You are on page 1of 14

REVIEWS

The bacterial epigenome


María A. Sánchez-​Romero and Josep Casadesús   *
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.

Methylome
In the second half of the 20th century, eukaryotic genomes cell lineages. At the turn of the century, DNA adenine
Overall DNA methylation were found to contain information superimposed over methylation was found to be essential for the patho-
pattern in a genome. the nucleotide sequence1. In multicellular eukaryotes, genesis of Salmonella enterica subsp. enterica serovar
this additional epigenetic information controls differen- Typhimurium11,12, and later of other pathogens13,14.
Adaptive value
tiation and development, and its perturbation can cause Together, these studies extended to bacteria the notion
In population genetics, the
contribution of a phenotypic pathological conditions2. The main epigenetic signals in that DNA methylation can exert epigenetic control over
trait to the fitness of an eukaryotic genomes are DNA methylation3 and post-​ the phenotype6,7,15–17. The possibility that phosphoro-
individual or a population. translational modification of histones4, and both mech- thioation of the DNA backbone may provide additional
anisms contribute to establish the transcription patterns epigenetic marks, perhaps in a manner complementary
Restriction–modification
(R-​M) systems
that govern the formation of specialized cell types. DNA to DNA methylation, remains open18,19.
Machine systems that methylation controls the binding of proteins to DNA and A hurdle in the investigation of the roles of DNA
eliminate foreign DNA provides a primary layer of information to shape the methylation in bacteria was that most studies focused
molecules by endonucleolytic eukaryotic epigenome3. DNA methylation is also found on 6mA, which is a base modification that was difficult
cleavage and protect the
in bacterial genomes, where it controls DNA–protein to detect. This technical limitation has been overcome
genome by modification of the
cognate endonuclease target. interactions as in eukaryotes5–8 (Box 1). through the development of DNA-​sequencing techno­l­
A frequent type of protection The methylated bases found in bacteria are N6-methyl-​ ogies that enable the high-​throughput analysis of DNA
is DNA methylation. adenine (6mA) and C5-methyl-​cytosine (5mC), which methylation across the genome (the methylome). Two
also exist in eukaryotes, and N4-methyl-​cytosine (4mC), such technologies, single-​molecule real-​time sequenc-
which is found only in bacteria and archaea (Table 1). ing (SMRT)20,21 and nanopore sequencing22, are currently
Base methylation is post-​replicative, and the bacte- ‘gold standards’ in the study of bacterial methylomes23.
rial genome is methylated at both DNA strands in the SMRT technology permits researchers to infer the pres-
absence of DNA replication. Upon DNA replication, ence of methylated bases on the template DNA strand
base methyl­ation is present in one strand only (hem- from kinetic signatures that are specific for each type
imethylation)5. Early studies in Escherichia coli and of methylated base20. Nanopore sequencing identifies
other gammaproteobacteria unveiled roles for DNA methylated nucleotides by detecting changes in electric
adenine methylation in chromosome replication, mis- current density upon electrophoretic passage of the sam-
match repair, control of transposon activity and regu- ple through an orifice22. Methylome analysis has shown
lation of transcription5,8. Proteins that can discriminate that base methylation is widespread in the genomes
the methyl­ation state (methylated or hemimethyl­ated) of bacteria and archaea24, including the small genomes of
of cognate binding sites were also identified5. In alpha­ certain (but not all) obligate parasites25. The wealth
Departamento de Genética, proteobacteria, DNA adenine methylation was found to of information provided by methylome analysis has
Facultad de Biología,
Universidad de Sevilla,
govern the cell cycle9. Seminal studies in uropathogenic also updated (and sometimes changed) classic notions
Sevilla, Spain. E. coli unveiled a role for DNA adenine methylation in about the physiological relevance and adaptive value of
*e-​mail: casadesus@us.es the formation of phenotypic variants of bacterial cells10, DNA methylation in bacteria. For instance, the DNA
https://doi.org/10.1038/ in a manner reminiscent of the involvement of DNA methyl­t ransferases of restriction–modification (R-​M )
s41579-019-0286-2 cytosine methylation in the formation of eukaryotic systems were thought to function in genome defence

Nature Reviews | Microbiology


Reviews

Box 1 | The epigenomes of eukaryotes and bacteria DNA methyltransferases are active as monomers37. For
the methylation reaction, the target base is rotated out
• In eukaryotes, epigenetic modification of the genome involves DNA methylation3 (‘flipped out’) of the DNA helix and inserted into the
and histone modification4. Bacteria lack histones, and epigenetic control relies on catalytic pocket of the enzyme 35,36. Depending on
DNA methylation only6. the position of the target base in the double helix, DNA
• In eukaryotes, de novo and maintenance forms of DNA methylation are performed methyltransferases are classified into two groups: exo-
by separate enzymes2. Bacterial DNA methyltransferases have both de novo and cyclic (for 6mA and 4mC) and endocyclic (for 5mC)37.
maintenance activities37.
Aside from this difference, all DNA methyltransferases
• In eukaryotes, two main mechanisms exist to erase DNA methylation marks: active show structural similarity, and the SAM-​binding domain
demethylation by dedicated proteins (Tet enzymes), and passive demethylation by
(or catalytic domain) is especially well conserved across
the hindrance of DNA methylase activity upon DNA replication35. In bacteria, DNA
demethylation is usually passive66, and the relevance of active demethylation by
kingdoms. By contrast, the target recognition domain
DNA repair remains to be evaluated82. (TRD) shows high variability in sequence and structure,
• In both bacteria and eukaryotes, transcriptional repression by DNA methylation is
which correlates with the manifold target specificities38.
common3,6. Transcriptional activation of bacterial genes under DNA methylation Structural similarity and conservation of motifs between
control often involves demethylation (partial or complete, single- or double-​stranded) different groups of DNA methyltransferases may be
of promoters or regulatory regions57,72,89,90,94,158. indicative of a monophyletic origin, and diversification
• The methylated base typically involved in the control of eukaryotic transcription is across kingdoms may have occurred upon the insertion
C5-methyl-​cytosine3, whereas in bacteria it is often N6-methyl-​adenine7,14. However, and permutation of domains39.
direct control of bacterial transcription by C5-methyl-​cytosine has been demonstrated DNA methyltransferases associated with R-​M sys-
recently126. Transcriptional control by N4-methyl-​cytosine may also exist130. tems are abundant, with an average of more than two
• In multicellular eukaryotes, the DNA methylation pattern of the genome is R-​M systems per genome40,41. Species with higher num-
reprogrammed during gametogenesis and during early embryonic development2. bers of R-​M systems exist, and extreme examples are
In bacteria, reprogramming does not occur, and the DNA methylation pattern can be found in Helicobacter pylori strains that encode more
transmitted unaltered across generations. However, the acquisition and loss of DNA than 50 R-​M systems42. In addition, many bacterial and
methyltransferase genes41 and recombinational shuffling of DNA methyltransferase archaeal genomes harbour at least one orphan DNA
domains27,33,143 can produce novel methylation patterns in bacterial genomes.
methyltransferase24. Orphan DNA methyltransferase
• In both bacteria and eukaryotes, DNA methylation controls the formation of phenotypic genes are also found in about one fifth of phage genomes43.
variants of genetically identical cells. However, DNA methylation-​dependent formation
Most orphan DNA methyltransferases are conserved at
of bacterial cell lineages can show programmed reversion (phase variation)15,27,93,111.
the genus level; the most highly represented are the Dam
family in gammaproteobacteria and the CcrM family in
only, and housekeeping functions were ascribed to alphaproteobacteria24. Bioinformatic detection of a gene
solitary (‘orphan’) DNA methyltransferases not asso- encoding a DNA methyltransferase does not imply that
ciated with R-​M systems. This distinction is not valid the enzyme is active, or even expressed: for example,
anymore, as a number of R-​M methyltransferases have transcription of the gene encoding the YhdJ methyltrans-
been shown to have roles in addition to genome defence, ferase of gammaproteobacteria is low or absent under
including transcriptional regulation and the formation laboratory conditions44.
of phenotypic cell variants26–28. The initial notion that Most DNA methyltransferases recognize specific
solitary DNA methyltransferases were rare also did not DNA targets: for instance, Dam and CcrM have a single
hold up, as methylome analysis has shown that they are target site (GATC and GANTC, respectively), whereas
abundant24. Finally, the semantics of the bacterial epige- Dcm has two target sites, CCAGG and CCTGG5. Orphan
nome, which traditionally had centred on 6mA, has been DNA methyltransferases that have several TRDs (and
extended to 5mC and 4mC. thus are multispecific) have been described in Bacillus
In this Review, we summarize historic and recent subtilis phages43. Nonspecific DNA methyltransferases
developments in epigenetic signalling by DNA methyl­ also exist, but they will not be discussed here45.
ation, with an emphasis on its contribution to pheno- The DNA methyltransferases of R-M systems methyl­
typic heterogeneity in bacterial populations. This bias ate DNA in a distributive manner; that is, they methylate
may be justified by the growing evidence that DNA only one target upon DNA binding37. Distributive
methylation-​mediated control of lineage formation is DNA methylation can be seen as a crucial adaptation
a widespread phenomenon in bacteria14,15,27,28, whereas of these enzymes to their biological function, as the need
the control of physiological processes such as chromo- to rebind DNA after each methylation event can slow
some replication and mismatch repair may be restricted down the methylation of phage DNA, thus facilitating
to certain bacterial clades8. An emphasis on phenotypic restriction by the cognate R-M endo­nuclease37. Orphan
heterogeneity may be further warranted by theoretical DNA methyltransferases are generally processive, and
analysis that has outlined its adaptive value29,30. We also so methylate multiple targets without dissociating
discuss, often in a speculative manner, evolutionary from DNA37,46. However, the CcrM methyl­transferase
aspects of bacterial DNA methylation. Complementary of alphaproteobacteria seems to be distributive47. Pro­
information can be found in other reviews27,31–34. cessivity can vary depending on the genome context;
for instance, certain DNA sequences that flank the
DNA methyltransferases GATC site can decrease the processivity of the Dam
The formation of 6mA, 5mC and 4mC is catalysed by methyltransferase48.
DNA methyltransferases, which transfer a methyl group Bacterial DNA methyltransferases can be essential or
from S-​adenosyl-methionine (SAM) to DNA35,36. Most dispensable7,13,49. In the genus Yersinia, Dam-​dependent

www.nature.com/nrmicro
Reviews

SOS regulon
methylation is essential in Yersinia enterocolitica, and Hemimethylation and demethylation
Regulatory network that dispensable in two related species, Yersinia pseudo­ Bacterial DNA methyltransferases are active on both
responds to DNA damage tuberculosis and Yersinia pestis50–52. Growth conditions nonmethylated and hemimethylated DNA46,63, thus
in gammaproteobacteria. can also determine whether a DNA methyltransferase is acting as both writers of the methylome and copyists
essential or dispensable: for instance, the CcrM methyl­ that transmit the methylation pattern of the genome to
ase of Caulobacter crescentus is only essential in rich daughter cells. Because every round of DNA replica-
medium 53. The phenotypes of strains that lack or tion produces hemimethylated DNA from methylated
overproduce a DNA methyltransferase can be indic- DNA, methylome inheritance requires methylation of
ative of the physiological roles that DNA methyla- the newly synthesized DNA strand (Fig. 1). The duration
tion might have in the species under study14,49,54,55. of the hemimethylated state after DNA replication varies
Moreover, altered patterns of gene expression can among bacterial clades, and a crucial factor is the avail-
provide evidence that the methylation state of promot- ability of the DNA methyltransferase. In gammaproteo-
ers or regulatory regions controls the activity of the bacteria, Dam methylase is synthesized at every stage of
transcriptional machinery56,57. However, altered gene growth64, and the hemimethylated state of GATC sites is
expression does not necessarily imply direct transcrip- short-​lived because the Dam methylase trails the DNA
tional control, because indirect effects are possible. For replication fork at a short distance, estimated to be less
instance, increased levels of SAM in DNA methylase than 10 kbp65. By contrast, the genome of alphaproteo-
mutants may induce changes in gene expression due bacteria remains hemimethylated during most of the cell
to alterations in one-​carbon metabolism58. Similarly, cycle, because the CcrM methylase is only synthesized
transcriptome differences can reflect indirect control. at the final stage of chromosome replication9,32,34.
For instance, upregulation of the SOS regulon in dam As a rule, hemimethylation after DNA replication is
mutants of E. coli and S. enterica is not caused by Dam-​ transient if the cognate DNA methyltransferase is avail-
dependent transcriptional control, but by DNA strand able. However, at specific sites methylation of the daugh-
breakage59,60. Enigmatic cases of post-​transcriptional ter strand can be hindered by the binding of proteins that
regulation by Dam-​dependent methylation have also prevent DNA methyltransferase activity, causing methyl­
been described61,62, and a tentative explanation is that ation loss in a passive manner (Fig. 1). This phenome-
Dam methylation may regulate transcription of genes non is well documented in E. coli and S. Typhimurium:
involved in control of mRNA stability, mRNA translation stable hemimethylated GATC sites are formed when a
or other post-​transcriptional events. DNA-​binding protein protects hemimethylated DNA

Table 1 | Examples of writers and readers of the bacterial epigenome


Methylated base Writer Target readers Physiological process or
sequence phenotype under control
(5′–3′)
N6-methyl-​adenine Dam GATC DnaA102, SeqA86, Chromosome replication and
MutHLS105, RNA segregation86,102,104, nucleoid
H 3C
NH polymerase91, organization87,164, mismatch repair105,
transposase91,134, transposition91,134, conjugal transfer88,
N N Lrp69,88,89,163, OxyR70–72,94, motility56, synthesis and secretion
Fur73 and HdfR74,75 of virulence determinants11,12,56,62,165,
N N
H envelope structure166, bacteriophage
resistance107 and antibiotic resistance155
CcrM GANTC GcrA90, MucR76,167 and Cell cycle control32,34
RNA polymerase53,57
ModA13 AGAAA Unknown Antibiotic resistance, epithelial cell
invasion and biofilm formation113
SpnD39III Allele-​specific Unknown Opacity (capsule synthesis) and
(six variants) virulence123
C5-methyl-​cytosine Dcm CCAGG and Unknown General stress response128 and
CCTGG drug transport129
NH2
H3C JHP1050 GCGC Unknown Cell morphology , competence,
N outer membrane composition and
copper resistance126
N O
H

N4-methyl-​cytosine M2.Hpy.AII TCTTC Unknown Virulence (adhesion to host cells)130


H3 C
NH

N O
H

Nature Reviews | Microbiology


Reviews

from Dam methylase activity6,14,66. If Dam methyla- OxyR70–72, Fur73 and HdfR74,75 are examples of DNA-​
tion hindrance persists during two consecutive rounds binding proteins that are involved in ‘passive’ erasure of
of genome replication, a nonmethylated GATC site is methyl groups (see below). Undermethylated GANTC
formed14,15. Undermethylation requires that the pro- sites have also been detected in alphaproteobacterial
cessivity of Dam methylase is reduced, which occurs at genomes, and an example of protection from CcrM
GATC sites that are flanked by specific, A–T-​rich DNA methylation has been shown to involve a transcription
sequences48,67. The effect of sequences outside the GATC factor, MucR76. Passive demethylation may be a common
site is not surprising, as DNA-​bound Dam methyltrans- phenomenon in the bacterial world: methylatable sites
ferase spans 10 bp68. The transcription factors Lrp69, that lack methylation in one or both DNA strands are

a b 5′ 3′

Methylated 5′ 3′
MBF Dam
G C
CH3 A T G G
A CH3 CH3 A C A CH3
T
C G T T C T
C A T C Methylation
G A hindrance
3′ 5′ G

3′ 5′
5′ 3′
DNA replication

5′ 3′ 5′ 3′ Hemimethylated
(stable)
G C G C
CH3 A T A T
5′ 3′ T A T A CH3
C G C G
Hemimethylated
Dam 3′ 5′ 3′ 5′

G G DNA replication
CH3 A C A CH3
T T C
C A T TC 5′ 3′
A
G G

3′ 5′
5′ 3′ G G Methylation
A C A hindrance
T T C T
C A T C
G A
G

3′ 5′
5′ 3′

Nonmethylated
Methylated
G C G C G C G C
CH3 A T CH3 A T A T A T
T A CH3 T A CH3 T A T A
C G C G C G C G

Most GATC sites Special GATC sites

Fig. 1 | Methylation, hemimethylation and passive demethylation of gATc sites in gammaproteobacteria.


a | The bacterial genome is methylated at both DNA strands in the absence of DNA replication. Upon DNA replication,
methylation is present in the template strand only (hemimethylation). Hemimethylation after DNA replication is transient
as Dam, an orphan DNA methyltransferase that trails the DNA replication fork , targets GATC sites for methylation to
restore the methylated state. This cycle occurs once per round of DNA replication and affects most GATC sites5. b | Binding
of a protein that functions as a methylation-​blocking factor (MBF) can prevent methylation of a hemimethylated GATC
site by the Dam methyltransferase. If hindrance of methylation persists during two consecutive rounds of DNA replication,
a nonmethylated GATC site is formed66. DNA methylation hindrance by blocking factors occurs only at GATC sites flanked
by DNA sequences that decrease the processivity of the Dam methylase48 (not shown). Undermethylated (hemimethylated
or nonmethylated) GATC site states can be transmitted to daughter cells in certain cases.

www.nature.com/nrmicro
Reviews

common in the genomes of gamma- and alphaproteo- nonmethylated sites can be heritable, thus transmitting
bacteria24,77–80, and some of those sites are located within epigenetic states to daughter cells6,15. In gammaproteo­
protein-​binding sites24,80. In E. coli, the methylation state bacteria, Dam-​dependent methylation patterns are
of certain GATC sites depends on growth conditions, found at the transcriptional control regions of bistable
which suggests that environmental effects can contribute loci, and the elements involved in the formation of OFF
to shape the methylome81. and ON states are more complex than those in Fig. 2a.
‘Active’ DNA demethylation has been described in For instance, the E. coli pap operon contains six bind-
E. coli, and its physiological relevance remains to be fully ing sites for Lrp and two GATC sites93; the S. enterica
understood. AlkB, a DNA repair protein that removes opvAB and gtr operons contain four OxyR binding sites
alkyl groups from nucleic acids, also targets 6mA and and four GATC sites72,94; the E. coli sci1 locus contains
removes its methyl group82. Because AlkB expression three Fur binding sites and three GATCs73; and the
is strongly induced upon DNA damage, widespread S. Typhimurium std operon contains one binding site
demethylation of 6mA may occur under such con- for HdfR and three GATC sites75 (Fig. 2b). In all cases, the
ditions. It is thus conceivable that the demethylation ON and OFF states are dictated by the binding pattern
of physiologically relevant, critical GATC sites might of the cognate protein. Binding causes passive demethyl­
contribute to survival, perhaps by delaying genome ation, which can increase the affinity of the regulator
replication or cell division82. towards its cognate site (or sites), thus generating a posi­
tive feedback loop that stabilizes the epigenetic state,
Readout of methylation marks which can sometimes be heritable.
The methyl group of 6mA, 5mC and 4mC protrudes Certain bistable loci that are controlled by Dam-​
from the major groove of the double helix, which pro- dependent methylation switch between alternative
vides a platform for DNA-​binding proteins to bind to states, and the system is thus reversible (‘phase-​variable’;
cognate nucleotide sequences. As a consequence, the see below)15,66. Switching between states requires DNA
methylation state of critical sites can either favour or replication with concomitant formation of hemimethyl-
hinder the interaction between a DNA-​binding domain ated DNA, and a crucial factor that determines the fre-
of the protein and a cognate DNA site7. In addition, base quency of switching is the affinity of the DNA-​binding
methylation can induce or enhance DNA curvature83–85, protein for its binding sites72,95 (Fig. 2a). Ancillary factors
which can further modulate DNA–protein interactions. may affect switching: for instance, in the pap operon a
Inhibition of the activity of DNA restriction endo­ protein named PapI is necessary for switching from OFF
nuclease by methylation of the target is a classic example to ON and for maintenance of the ON state95.
of a methylation-​sensitive DNA–protein interaction. Frequencies of ON–OFF switching are locus specific.
However, discrimination of DNA methylation states For instance, under laboratory conditions the GtrOFF and
can be far more complex. Certain methylation readers GtrON subpopulations of S. enterica have roughly iden-
(for example, the GATC sequestration factor SeqA) tical sizes96, whereas the OpvAB and Std switches are
can bind both methylated and hemimethylated DNA lopsided towards the OFF state72,97. However, switching
targets, albeit with different affinities86,87. Other pro- can be under environmental influence: for instance, the
teins (for example, the transcription factor Lrp) bind StdOFF and StdON subpopulations have different sizes in
both hemi­methylated and nonmethylated targets, but the laboratory and in the animal intestine97,98 (see below).
the binding patterns and the affinities are different88,89. Additional evidence for environmental control has come
Another remarkable bias in protein binding to hemi- from the observation that housekeeping proteins respon-
methylated sites is strand specificity, which can favour sive to environmental cues can alter switching frequen-
binding to only one of the hemimethylated DNA spe- cies. Examples include the global regulator CRP99, the
cies produced by DNA replication89–91. An unproven signal transduction proteins CpxAR100 and the nucleoid
but reasonable speculation is that the overall methyl­ proteins H-​NS and HU72,101.
ation state of the genome may also influence the bind-
ing of proteins to specific regions, by moulding nucleoid Cell cycle signalling by DNA methylation
topology34,92. In gammaproteobacteria, the initiation of chromo-
Especially complex DNA–protein interactions occur some replication and segregation of newly replicated
at regions that harbour several protein-​binding sites, nucleoids is controlled by readers that discriminate
which in turn contain methylatable motifs6,15,66. In such methylation from hemimethylation. One such reader is
cases the protein-​binding pattern determines the methyl­ DnaA, which initiates chromosome replication if GATC
ation state, because methylation hindrance causes passive sites within the origin of replication (oriC) are methyl-
demethylation of specific sites. In turn, the methyl­ ated102. Another reader of the methylation state is SeqA,
ation pattern determines the protein-​binding pattern. which binds hemimethylated GATC sites at the oriC
The same protein is thus the eraser of methyl groups and delays Dam methyltransferase activity86. Transient
and the reader of the resulting DNA methylation pattern. hemi­methylation of the oriC by SeqA also represses
In the imaginary example presented in Fig. 2a, the exist- transcription of the nearby dnaA gene, which contains
ence of two methylatable binding sites permits alternative, GATC sites in the promoter region103. In addition, the
mutually exclusive binding patterns, and each binding binding of SeqA to hemimethylated GATC sites gov-
pattern determines a distinct DNA methyl­ation pattern, erns the condensation and segregation of newly repli-
because methylation is hindered at the sites bound by cated nucleoids104. Transient hemimethylation upon
the regulator6,66. Such combinations of methylated and replication fork passage is also a signal for repair of

Nature Reviews | Microbiology


Reviews

mismatched nucleotide pairs, which relies on the ability transcription upon passage of the replication fork.
of the MutHLS complex to cleave the daughter strand at Classic examples in gammaproteobacteria are the trans-
hemimethylated GATCs105. posase (tnp) gene of insertion element IS10 (ref.91) and
In C. crescentus, the initiation of chromosome repli­ the plasmid-​borne traJ gene, which encodes an activa-
cation by DnaA requires GANTC methylation at the tor of conjugal transfer88,89. Because the hemimethyla-
replication origin (Cori)32,34. However, Cori remains tion state is transient, the expression of tnp and traJ is
spontaneously hemimethylated, without sequestration, short-​lived, and synthesis of the corresponding gene
because the CcrM methylase becomes available only at products is restrained. An additional sophistication of
the end of every round of chromosome replication32. tnp and traJ hemimethylation readout is asymmetry:
Hence, the DnaA proteins of gamma- and alphaproteo- the activation of tnp and traJ transcription takes place
bacteria read analogous signals at oriC and Cori, respec- in only one of the hemimethylated DNA species pro-
tively, but the mechanisms that control methylation of duced by DNA replication89,91. The adaptive value of
the replication origin are different7,32. strand-​specific transcription of tnp and traJ may be
Reading of DNA hemimethylation signals by RNA tentatively understood: asymmetric reading restrains
polymerase and transcription factors can activate synthesis of dangerous (transposase) or energetically

a b OFF state ON state


Lrp Lrp Papl
GA T C CH3 CH3
pap
State A State B
CH3 CH3
GATC 1 GATC 2 GATC 1 GATC 2
CH3 CH3
Fur
CH3 CH3 CH3
CH3 CH3 sci1
DNA replication CH3 CH3 CH3

OxyR OxyR
CH3 CH3 CH3 CH3 CH3 CH3
gtr
CH3 CH3 CH3 CH3

CH3 CH3
OxyR OxyR
CH3 CH3 CH3 CH3
opvAB
CH3 CH3 CH3 CH3

CH3 CH3
CH3 CH3 CH3 CH3 HdfR
std
CH3 CH3
CH3 CH3 CH3 CH3

Fig. 2 | control of bistable loci by the formation of alternative Dam six binding sites for Lrp in pap93, four for OxyR in gtr94 and opvAB106, three for
methylation patterns. a | Shown is an imaginary locus that harbours two Fur in sci1 (ref.73), and one binding site for HdfR in std75. The transcription
protein-​binding sites, each containing a GATC (GATC 1 and GATC 2). factor binding sites contain GATC sites: two in pap93, three in sci1 (ref.73) and
Two binding patterns are possible, and each binding pattern causes passive std75, and four in opvAB107 and gtr94. The methylation state of such sites
demethylation of a specific GATC site because methylation is hindered at is dictated by the binding pattern of the transcription factor: GATCs
the sites bound by the regulator. Nonmethylation can increase the affinity located within sites bound by the transcription factor undergo passive
of the protein for its cognate binding site, thus creating a positive feedback demethylation, while GATCs within unbound sites are methylated. This
loop that stabilizes the DNA methylation pattern and makes it heritable. mutual exclusion between Dam methylation and transcription factor
However, the system can be reset when hemimethylated DNA substrates binding produces distinct patterns of DNA methylation in the regulatory
are formed upon DNA replication. Switching occurs if the protein moves region of OFF and ON cells. In PapOFF cells, the upstream GATC site is
from one cognate site to the other, and a major factor that determines the methylated and the downstream GATC is nonmethylated, and the opposite
frequency of switching in each direction is the affinity of the protein GATC methylation pattern is found in the PapON lineage93. In the regulatory
towards each binding site72,95. Ancillary factors (not shown) can skew regions of gtr and opvAB, cells in OFF and ON states display opposite Dam
switching, either in a stochastic manner or in response to environmental methylation patterns involving two methylated and two nonmethylated
signals100,101. b | Epigenetic states of bistable loci of Escherichia coli (pap and GATC sites72,94. The formation of nonmethylated GATC sites is likewise
sci1) and Salmonella enterica (gtr, opvAB and std) under the control of Dam observed in Sci1OFF cells as the consequence of Fur binding73, and in StdON
methylation. The diagrams show regulatory regions upstream of the cells as the consequence of HdfR binding75. Under laboratory conditions,
promoters, with the promoter-​proximal side on the right. Transcription is the formation of ON and OFF subpopulations is reversible (phase variable)
repressed in the OFF state and activated in the ON state. In all cases, the in pap10, gtr94, opvAB106 and std97, but not in sci1 (ref.73). In pap, switching to
regulatory region contains binding sites for a transcription factor: ON also requires an ancillary factor, PapI95.

www.nature.com/nrmicro
Reviews

expensive (conjugation) cell products. Asymmetric state quickly reproduces a population in which >99%
transcription from a hemimethylated promoter has also of cells are OpvABOFF (Fig. 3a). The phase variation of
been observed in the creS gene of C. crescentus76, which the opvAB operon may thus be viewed as a trade-​off
suggests that the transcription factor involved (GrcA) between virulence and bacteriophage resistance107.
may be a hemimethylation reader with DNA strand Another phase-​variable locus, std, provides a unique
specificity. An attractive speculation is that asymmetric example of the power of Dam-​dependent phase varia-
reading of GANTC hemimethylation may contribute to tion to produce phenotypic diversity. The std operon is
differentiation of the two cell types34. found in the genome of S. Typhimurium and encodes
fimbriae that facilitate adhesion to the mucus layer
Bistability and phase variation of the large intestine98,108. In addition, the std operon
Heritable DNA adenine methylation patterns. The encodes two transcription factors, StdE and StdF, that
formation of alternative Dam-​dependent methylation are activators or repressors of more than one hundred
patterns, a phenomenon discussed above, controls the genes97. Pleiotropic control thus splits the bacterial pop-
expression of bistable loci and produces phenotypic ulation into two subpopulations that differ in multiple
variants of genetically identical cells in gammaproteo­ phenotypic traits, a phenomenon that may be viewed
bacteria66. Dam-​dependent bistability can be phase-​ as a rudimentary differentiation program97 (Fig. 3b). The
variable15. Paradigms of Dam-​dependent phase variation molecular mechanisms that control std phase variation
in E. coli are the pap operon of uropathogenic strains, are more complex than those in other Dam methylation-​
which encodes fimbriae for adhesion to the urinary dependent loci: std transcription is activated by HdfR,
epithelium 93, and the agn43 gene, which encodes a transcription factor of the LysR family, and the tran-
a non-​f imbrial adhesin involved in bacterial auto­ scription of hdfR is activated by an std product, StdE75.
aggregation70,71. Descriptions of Dam-​dependent control The binding of HdfR upstream of the std promoter is
of pap and agn43 phase variation can be found in other hindered by Dam-​dependent methylation, and HdfR
reviews6,7,66,93 and will not be discussed here. binding prevents methylation of two GATC sites in the
Natural isolates of non-​typhoidal S. enterica carry regulatory region75 (Fig. 2b). In addition, the formation of
one to four operons that encode glucosyltransferases, StdON cells requires autogenous activation of transcrip-
which add sugars to the lipopolysaccharide O-​antigen96. tion by StdF, which binds the std promoter in a Dam
Such operons, named gtr, are associated with prophages methylation-​independent fashion75.
or prophage remnants present in the genome, and each
operon may produce a distinct modification of the Phase-​variable DNA methyltransferases of R-​M systems.
O-​antigen. The resulting variants of the bacterial sur- Two decades ago, a Haemophilus influenzae gene (mod)
face may adapt Salmonella to interact with different host homologous to genes encoding the DNA adenine
receptors, and they may also alter phage susceptibility96. methyltransferases of type III R-​M systems was found
The expression of gtr is phase-​variable, and the GtrOFF to undergo phase variation due to contraction and/or
and GtrON subpopulations harbour alternative patterns expansion of nucleotide repeats109. Another phase-​
of methylated and nonmethylated GATC sites within a variable mod gene belonging to a type III R-​M system
regulatory region upstream of the gtr promoter94. The was thereafter described in H. pylori, and phase vari-
transcription factor OxyR is both an eraser of methyl ation was found to be caused by length variation of a
groups and a reader of the resulting gtr methylation homopolymeric nucleotide tract in the 5′-coding region
patterns: OxyR binding to upstream cognate sites pre- of a gene located upstream of mod110. The type III
vents methylation of GATC1 and GATC2 and activates R-​M Mod enzyme of H. influenzae turned out to be more
transcription, whereas binding to downstream sites pre- than just a modification enzyme involved in genome pro-
vents methylation of GATC3 and GATC4 and represses tection: transcriptome analysis identified genes that were
transcription94,96 (Fig. 2b). either up- or downregulated in a mod mutant, which
Another phase-​variable operon under the control indicates that adenine methylation of specific genome
of Dam-​dependent methylation and OxyR is opvAB, sequences controlled the expression of specific genes.
whose products are cytoplasmic membrane proteins Switching in the availability of active DNA methyltrans-
that reduce the length of the S. enterica lipopolysaccha- ferase thus produced a phase-​variable regulon (‘phase­
ride O-​antigen106. As in gtr, the control of phase varia- varion’)111. An additional, remarkable observation was
tion is transcriptional. A regulatory region upstream of that the restriction enzyme of certain type III R-​M sys-
the opvAB promoter contains four OxyR binding sites tems was inactive in a number of Haemophilus strains,
and four methylatable GATC motifs; OpvAB OFF and which suggests that such R-​M systems had evolved into
OpvABON cell lineages display opposite patterns of OxyR epigenetic regulators of gene expression112. Further stud-
binding, which in turn generate opposite patterns of ies described 6mA phasevarions in Neisseria meningitidis
GATC methylation72 (Fig. 2b). Transcriptional switching and Neisseria gonorrhoeae113. Phase variation involved
is skewed towards the OpvABOFF state, and the OpvABON variations in the length of simple sequence repeats
lineage comprises only 0.3% of cells106. Shortening of the (Fig. 4), and the loci under the control of Mod-​mediated
O-​antigen renders OpvAB ON cells resistant to phages methylation included genes with roles in envelope
that use the O-​antigen as a receptor. OpvAB ON cells structure, virulence and stress responses114. The power
Homopolymeric nucleotide
tract
are unable to infect animal hosts107; however, as soon of phasevarions as sources of epigenetic variation is
DNA region that contains only as phage challenge ceases, phase variation produces remarkable: in contrast with the phase variation systems
AT or GC nucleotide pairs. OpvABOFF cells, and lopsided switching towards the OFF that generate heterogeneity of a single phenotypic trait,

Nature Reviews | Microbiology


Reviews

a OpvABOFF b
Long O-antigen Fimbriae
Flagella
T3SS
Dam-dependent
StdON switching StdOFF

OpvABON ILEUM
Short O-antigen

LPS Dam-dependent
switching

OpvABOFF OpvABON Selection of M cell Epithelial cell


the OpvABON
subpopulation
CAECUM

Bacteriophage
Reconstruction of the Selection of Mucus layer
orignal population, the OpvABON
predominantly virulent subpopulation

Resuscitation of Goblet cell


the OpvABOFF
subpopulation

Fig. 3 | Formation of subpopulations controlled by Dam-​dependent methylation. a | Phase-​variable transcription


of the Salmonella enterica opvAB operon produces a lineage of OpvABOFF cells with long O-​antigen chains in the
lipopolysac­charide (LPS) (OpvABOFF) and a lineage with shorter O-​antigen chains (OpvABON)106. Shortening of the
O-​antigen renders the OpvABON lineage avirulent but resistant to bacteriophages (many Salmonella-​targeting phages
use the O-​antigen as a receptor). In the presence of a phage, the OpvABOFF subpopulation will be eliminated, while the
OpvABON subpopulation will survive. When the phage challenge ceases, OpvABOFF cells produced by phase variation
will survive, and virulence will be regained. However, a small subpopulation of avirulent OpvABON cells will sustain
preadaptation to future phage challenge107. b | Phase-​variable transcription of the std operon produces subpopulations
of StdOFF and StdON cells97. The StdOFF lineage is motile (producing flagella) and synthesizes the type 3 secretion system
(T3SS), promoting invasion of epithelial cells in the ileum of infected animals, which is the initial stage of systemic
Salmonella spp. infection97,162. In turn, the StdON lineage produces fimbriae that permit adhesion to the mucus layer
of the caecum in the large intestine98,108 but harbours neither flagella nor a T3SS97. Adhesion of the Std fimbriae to
the caecal mucosa increases inflammation98, which promotes the survival of Salmonella. StdOFF cells can thus cause
acute infection, whereas StdON cells cause chronic infection, and std phase variation can be viewed either as a division
of labour or as a bet-​hedging strategy.

bacterial lineages under phasevarion control can differ the presence of 6mA at promoters or transcription
in multiple phenotypic traits. control regions120.
Additional examples of phase-​variable type III R-​M Phase variation of DNA adenine methyltransferases
systems have been described in the past decade33,115–117, belonging to type I R-​M systems was first identified in
and bioinformatic analysis predicts that nearly one fifth Mycoplasma pulmonis121, and later in other human patho­
of type III mod genes may be phase variable, as judged gens27,122. A frequent mechanism of switching is recom-
from the presence of sequence repeats117. In certain bac- bination between inverted repeats of type I R-​M (hsd)
terial species (for example, Neisseria spp.), mod alleles genes, which can be present in multiple copies, complete
show high homology except in the target recognition or incomplete, in a genome27 (Fig. 4). DNA inversion by
domain, thus providing evidence for rapid evolution site-​specific recombination and the expansion and/or
by recombinational shuffling113. Target diversifica- contraction of single sequence repeats can also mediate
tion, together with frequent horizontal transfer of mod the switching of hsd genes27,33 (Fig. 4). An example of the
Transparent–opaque genes118, may further promote phenotypic diversity119. contribution of type I R-​M systems to phase variation
transition A DNA adenine methyltransferase of a phase-​ has been described in Streptococcus pneumoniae, in
Formation of phenotypic variable type II R-​M system has been shown to regulate which genetic rearrangements generate six 6mA vari­
variants (phase variation) of gene expression and virulence in Campylobacter jejunii, ants of a type I DNA methyltransferase that controls the
Streptococcus pneumoniae
where phase variation is controlled by the insertion or transparent–opaque transition123–125. Each DNA methyl­
that is involved in
pneumococcal carriage deletion of repeat units. However, it remains to be estab- transferase variant generates a distinct methylation
and invasive infection. lished whether or not gene expression control involves pattern in the genome, which results in different gene

www.nature.com/nrmicro
Reviews

a ON–OFF switching b Methylome diversification


IR IR IR IR
Genome methylated
ON (full length) Recombination between
inverted repeats
SSR

Contraction and/or
expansion of repeats

Stop Variation of genome methylation pattern


SSR

OFF (truncated)

Genome not methylated Shuffling of target


recognition domains

Fig. 4 | Mechanisms of switching and methylome diversification in phasevarions. a | Phase variation (ON–OFF
switching) by the expansion and/or contraction of single sequence repeats (SSRs). Frameshifting in the coding sequence
of a DNA methyltransferase gene causes premature termination, as a stop codon is introduced, generating a cell lineage
whose genome lacks DNA methylation27,33,114. However, further frameshifting events can either revert or suppress the
frameshift, and the restoration of a functional reading frame can permit DNA methyltransferase synthesis and genome
methylation. If base methylation affects the DNA–protein interactions that are crucial for gene expression, each bacterial
lineage will show a distinct expression pattern of methylation-​sensitive loci114. b | Formation of DNA methyltransferase
variants by recombination between inverted repeats (IRs) or between target recognition domains. DNA methyltransferase
variants with distinct specificities generate allele-​specific patterns of genome methylation, which can in turn have
phenotypic consequences27.

expression profiles and produces pneumococcal lineages DNA methylation and bacterial evolution
with different virulence capacities123–125. DNA methylation is both a conservative and an inno-
vative force in bacterial evolution. R-​M systems func-
Transcriptional control by 5mC and 4mC tion as barriers for the acquisition of genetic elements
JHP1050 is a 5mC DNA methyltransferase derived from by horizontal transfer26,40. Furthermore, R-​M systems
an ancestral R-​M system that is highly conserved among can limit recombination between bacterial clades that
H. pylori strains126. Mutants that lack JHP1050 show harbour distinct R-​M repertoires131,132. A likely mecha­
alterations in adherence to host cells, natural competence nism is cleavage of incoming DNA molecules, with a
for DNA uptake, bacterial cell shape and susceptibility to concomitant shortening of recombination tracts. These
copper126. These phenotypes correlate with altered pat- conservative roles are strengthened by the behaviour of
terns of gene expression, and site-​directed mutagenesis of R-​M systems as toxin–antitoxin addiction modules: if
GCGC targets at promoter regions has provided evidence an R-​M system is lost, the decrease in the number of
for direct transcriptional control by 5mC126. The forma- DNA methyltransferase molecules may permit restric-
tion of 5mC may further influence gene expression in an tion, and a single chromosome break can be sufficient to
indirect manner, perhaps by changes in DNA topology, cause cell death40,133. Among orphan methyltransferases,
as was previously proposed for 6mA92. Dam contributes to genome maintenance by the repres-
In Vibrio cholerae, the VhcM orphan 5mC methyl- sion of transposable elements91,134 and prophages135,136,
transferase is necessary for optimal growth, both in vitro as well as by reduction of the mutation rate via mis-
and during infection, and modulates stress responses127. match repair105. Interestingly, Dam methyltransferase
Transcriptome changes in vhcM deletion mutants may can also be viewed as an addictive component of the
be a consequence of direct transcriptional control, but genome, as its loss causes genome instability upon DNA
indirect effects are also conceivable127. A similar situation strand breakage59,137.
may apply to upregulation of the general stress response A major role of both Dam and phase-​variable DNA
in E. coli mutants lacking the Dcm 5mC methyltrans- methyltransferases is the formation of phenotypic line-
ferase128, as well as to RpoS-​dependent control of a drug ages, which enables division of labour in a community or
transporter129. prepares the community for future changes in the envi-
The literature also contains one example of epigenetic ronment (bet-​hedging)138. An organism can only have a
signalling by 4mC: M2.Hpy.AII, a DNA methyltrans- limited set of traits, and improvement of a given pheno­
ferase that introduces a methyl group at the N4 position type comes at the expense of another phenotype139.
in the first C of TCTTC motifs, controls gene expression Lineage formation solves this problem by allotting spe-
and virulence-​related traits in H. pylori130. As in other cific phenotypes to each cell type in a manner that can
studies discussed above, transcriptional control may be benefit the whole population. Formation of epigenetic
either direct or indirect130. cell variants may also provide a strategy for mutation

Nature Reviews | Microbiology


Reviews

avoidance: mutations are often deleterious, and their costs. An example is provided by the Dam methylation-​
accumulation in a population can decrease fitness140,141. dependent OpvAB bet-​hedging system of S. enterica,
In fact, bacterial populations with high mutation rates which confers bacteriophage resistance by modification
adapt quickly to a given environment but are outcom- of the O-​antigen107 (see above). In an opvAB deletion
peted by non-​mutators in the long run142. By contrast, mutant strain, bacteriophage-​resistant mutants can be
non-​mutational variation can adapt bacterial popu- isolated; however, such mutants often harbour mutations
lations to environmental challenge with lower fitness that impair fitness107.

Box 2 | Applications of DNA methylation in biotechnology and synthetic biology


Lack of DNA methylation attenuates virulence, sometimes severely, in a large number of bacterial pathogens13. In species
in which DNA methylation is essential and mutants are not viable, the attenuation of virulence has been observed
following the overproduction of DNA methyltransferases13. This relationship between DNA methylation and virulence has
fostered novel preventive and therapeutic approaches to combat bacterial infections. For instance, live vaccines against
Salmonella spp. have been developed using strains that carry dam mutations, alone or combined with other mutations
that attenuate virulence159,160. Another example is an oral Yersinia pseudotuberculosis vaccine, which has been shown to
be protective in mice52. A different approach involves the use of DNA methyltransferase inhibitors as antibacterial agents152.
In gammabacterial pathogens in which Dam-​dependent methylation is dispensable, the inhibition of Dam methylase
activity can produce phenocopies of dam mutants, thus reducing virulence without affecting viability. As a consequence,
the selection of inhibitor-​resistant variants may not be strong. An additional advantage is that inhibitors of DNA adenine
methylation may be efficient against various bacterial pathogens, as DNA adenine methyltransferases show structural
similarity in different bacterial clades38. Inhibitors of the DNA adenine methyltransferases of alphaproteobacteria and of
Gram-​positive bacteria have been also described153,154. The inhibition of DNA adenine methylation was initially considered
harmless for the mammalian host. However, the recent discovery of DNA adenine methylation in the human genome161 raises
the concern of possible side effects.
A recent study has suggested the possibility of using Dam methylase inhibitors to enhance the therapeutic activity of
antibiotics155. Lack of Dam methylation increases the bactericidal activity of β-​lactams, because exposure to the antibiotic
causes oxidative damage, which in turn induces SOS-​dependent error-​prone repair. In wild-​type Escherichia coli,
Dam-dependent mismatch repair corrects the errors. However, in the absence of Dam methylation, the MutHLS mismatch
repair system generates double-​strand breaks in the DNA. Such breaks further activate the SOS response, thus generating
a toxic feedback loop that potentiates the lethal action of the antibiotic155.
DNA methylation may also have application in phage therapy: the use of phages that encode DNA methyltransferases
can be expected to boost the elimination of infectious bacteria, by delaying the ability of the host R-​M systems to kill the
incoming phage43.
An appealing but largely unexplored application of bacterial DNA methylation is the development of DNA methylation-​
based biosensors (see the figure). An example is provided by a recent study that describes synthetic memory systems that
are able to store information in E. coli in the form of CcrM-​dependent methylation patterns156. In the negative control
system (see the figure, part a), signal detection dislodges a repressor from the control region of a DNA methylase gene
(mtase), and methylation of the control region prevents repressor binding. The resulting feedback loop will keep the
sensor active even if the signal disappears, and the DNA methylation pattern will be transmitted to daughter cells,
thus providing memory of a past event156. In the positive control system (see the figure, part b), signal detection enables
a transcription factor to activate the expression of a DNA methylase gene. In both systems, genome methylation will
occur in response to the signal. The addition of a fluorescent protein gene downstream of the DNA methylase gene
(not shown in the figure) can help detect sensor activation. Variants of these systems have been engineered to detect
various environmental stimuli including heat, DNA-​damaging agents and nutrients. A device for signal reset can be added
to memory sensors (see the figure, part c): induced synthesis of a protease (Lon) permits the degradation of an engineered
DNA methylase variant that contains a protease-​sensitive tag 156.

a Negative control b Positive control c Signal reset


OFF state OFF state

mtase mtase Signal


lon
Repressor
Inactive activator

ON state ON state Protease

mtase
Signal
mtase

DNA methyltransferase Activator Protease-sensitive tag


Signal
Inactive mtase
repressor

www.nature.com/nrmicro
Reviews

Although Dam methylation-​dependent switches and may represent a burden in others. However, asymmet-
phasevarions may have analogous roles in the forma- ric mutation towards AT seems to be a universal phe-
tion of bacterial lineages, they must be viewed as the nomenon in bacterial genomes146, and the bias may be
products of distinct evolutionary strategies, each with explained by a high rate of 5mC deamination147.
advantages and drawbacks. Dam-​dependent switches
are the products of long-​term co-​evolution with the core Conclusions and future perspectives
genome, and the formation of Dam-​dependent methyl- Conservation of the DNA methyltransferase structure
ation patterns may be an especially robust mechanism suggests its early evolutionary origin38, and the wide-
for transmission of epigenetic memory to daughter cells. spread methylation of bacterial and archaeal genomes
Furthermore, crosstalk with the housekeeping machin- may be indicative of positive selection during evolu-
ery may permit adjustment of the switching frequency tion24. A conceivable scenario is that DNA methylation
in response to environmental cues72,100,101. However, arose as a genome defence mechanism associated with
integration into host regulatory networks may heav- restriction40. An alternative possibility is that restric-
ily constrain further evolution. By contrast, stochastic tion enzymes did not initially evolve to destroy foreign
switching of phasevarions may be prone to perturbation DNA molecules, but to guarantee that DNA methyl­
but at the same time be more evolvable, and independ- transferases remained active 24. High rates of hori-
ence from host genome control may further increase zontal transfer of DNA methyltransferase genes have
evolvability. Recombination between DNA methyl- contributed to the spread of DNA methylation in the
transferase alleles may be a powerful mechanism for the bacterial world41, and exchange and shuffling of DNA
generation of DNA methyltransferase alleles with novel methyltransferase genes have promoted methylome
TRDs, especially in species that harbour multiple DNA diversification24,27,118,143.
methyltransferase genes113,118. During evolution, and perhaps very early, the addi-
High rates of horizontal transfer of DNA methylase tion of epigenetic information to the nucleotide sequence
genes, either R-​M or orphan, can further promote turned out to confer adaptive traits to bacterial cells. One
bacterial diversification40,41. Indeed, DNA-​recognition such trait is the ability to form phenotypic cell variants,
domains seem to be highly mobile143. In R-​M systems, often in a reversible manner15,27,111. An asset of reversibil-
a change of DNA methylation specificity can permit ity is the ability to restore phenotypic heterogeneity after
the acquisition of novel foreign elements, including a bottleneck, a circumstance that is especially frequent
prophages, transposons, plasmids and other mobile ele- inside animal hosts27. The abundance of phasevarions in
ments, perhaps accompanied by the loss of resident, old bacterial pathogens may support this view27,33,114.
components of the peripheral genome41. Furthermore, In certain bacterial clades, long-​term co-​evolution
the acquisition of a novel methylation pattern may mod- with the host genome has permitted DNA methyltrans-
ify the transcriptome if the transcriptional machinery ferases to participate in the control of housekeeping
of the recipient cell contains appropriate readers. The processes, including the cell cycle32,34, chromosome
fate of the new-​born pattern of epigenetic regulation replication and segregation6–8, DNA repair105 and the
will be determined by natural selection, and it might secretion of virulence determinants13. Functional analy­
instantly produce a novel bacterial clade, in the case sis of bacterial methylomes may reveal novel roles in
of high adaptive value. Shuffling and exchange of TRD years to come.
modules may thus be viewed as a trial-​and-error game Modification of the host epigenome by bacterial path-
for bacterial diversification. ogens is a well-​established phenomenon, the significance
The use of 5mC as an epigenetic mark has an enig- of which remains to be understood148–150. Epigenetic
matic side because 5mC is unstable in all cells, including memory of a bacterial infection might benefit either
bacteria, and 5mC deamination produces thymine144. the host (for example, by boosting immune responses),
In the resulting T:G mismatches, discrimination of the the pathogen (for example, by weakening immunity) or
incorrect base is not feasible, as both T and G are nor- both organisms (for example, by facilitating commensal
mal bases. If left unrepaired, a GC to AT transition will or symbiotic co-​existence). An intriguing possibility is
occur upon DNA replication, and one of the daughter that DNA methyltransferases of intracellular pathogens
DNA molecules will contain a base pair substitution. may directly methylate the eukaryotic genome151.
Gammaproteobacteria possess a repair system (very Future applications of bacterial DNA methylation in
short patch repair) that corrects T:G mismatches by biotechnology may include the use of inhibitors of DNA
replacing T with C145. However, a system of this kind adenine methylation as antibacterial drugs152–155 and
is absent in many bacterial clades: for instance, 5mC the design of synthetic switches, sensors and memory
deamination contributes to the high mutation frequency devices156,157 (Box 2).
of H. pylori126. One can speculate that a high mutation
rate may have adaptive value in certain species and Published online xx xx xxxx

1. Holliday, R. Epigenetics: a historical overview. 4. Lawrence, M., Daujat, S. & Schneider, R. Lateral 7. Wion, D. & Casadesus, J. N6-methyl-​adenine:
Epigenetics 1, 76–80 (2006). thinking: How histone modifications regulate gene an epigenetic signal for DNA–protein interactions.
2. Henikoff, S. & Greally, J. M. Epigenetics, cellular expression. Trends Genet. 32, 42–56 (2016). Nat. Rev. Microbiol. 4, 183–192 (2006).
memory and gene regulation. Curr. Biol. 26, 5. Marinus, M. G. Methylation of DNA in Escherichia coli 8. Løbner-​Olesen, A., Skovgaard, O. & Marinus, M. G.
R644–R648 (2016). and Salmonella. Cell. Mol. Biol. 782–791 (1996). Dam methylation: coordinating cellular processes.
3. Jones, P. A. Functions of DNA methylation: islands, 6. Casadesus, J. & Low, D. Epigenetic gene regulation Curr. Opin. Microbiol. 8, 154–160 (2005).
start sites, gene bodies and beyond. Nat. Rev. Genet. in the bacterial world. Microbiol. Mol. Biol. Rev. 70, 9. Stephens, C., Reisenauer, A., Wright, R. & Shapiro, L.
13, 484–492 (2012). 830–856 (2006). A cell cycle-​regulated bacterial DNA methyltransferase

Nature Reviews | Microbiology


Reviews

is essential for viability. Proc. Natl Acad. Sci. USA 93, pathogens-​phase-variable epigenetic regulators 55. Shell, S. S. et al. DNA methylation impacts gene
1210–1214 (1996). evolving from restriction-​modification systems. expression and ensures hypoxic survival of
10. Blyn, L. B., Braaten, B. A., White-​Ziegler, C. A., Microbiology 165, 917–928 (2019). Mycobacterium tuberculosis. PLOS Pathog. 9,
Rolfson, D. H. & Low, D. A. Phase-​variation of This recent review covers phasevarions. e1003419 (2013).
pyelonephritis-​associated pili in Escherichia coli: 34. Mohapatra, S. S., Fioravanti, A. & Biondi, E. G. 56. Balbontin, R. et al. DNA adenine methylation
evidence for transcriptional regulation. EMBO J. 8, DNA methylation in Caulobacter and other regulates virulence gene expression in Salmonella
613–620 (1989). Alphaproteobacteria during cell cycle progression. enterica serovar Typhimurium. J. Bacteriol. 188,
This study provides the first description of Trends Microbiol. 22, 528–535 (2014). 8160–8168 (2006).
bacterial lineage formation under the control 35. Jurkowska, R. Z. & Jeltsch, A. Mechanisms and 57. Gonzalez, D., Kozdon, J. B., McAdams, H. H., Shapiro, L.
of DNA methylation. biological roles of DNA methyltransferases and DNA & Collier, J. The functions of DNA methylation by CcrM
11. Heithoff, D. M., Sinsheimer, R. L., Low, D. A. methylation: from past achievements to future in Caulobacter crescentus: a global approach. Nucleic
& Mahan, M. J. An essential role for DNA adenine challenges. Adv. Exp. Med. Biol. 945, 1–17 (2016). Acids Res. 42, 3720–3735 (2014).
methylation in bacterial virulence. Science 284, 36. Cheng, X. Structure and function of DNA 58. Friso, S., Udali, S., De Santis, D. & Choi, S. W.
967–970 (1999). methyltransferases. Annu. Rev. Biophys. Biomol. One-​carbon metabolism and epigenetics. Mol. Asp.
12. Garcia-​Del Portillo, F., Pucciarelli, M. G. & Casadesus, J. Struct. 24, 293–318 (1995). Med. 54, 28–36 (2017).
DNA adenine methylase mutants of Salmonella 37. Bheemanaik, S., Reddy, Y. V. & Rao, D. N. 59. Torreblanca, J. & Casadesus, J. DNA adenine methylase
typhimurium show defects in protein secretion, cell Structure, function and mechanism of exocyclic DNA mutants of Salmonella typhimurium and a novel
invasion, and M cell cytotoxicity. Proc. Natl Acad. methyltransferases. Biochem. J. 399, 177–190 Dam-​regulated locus. Genetics 144, 15–26 (1996).
Sci. USA 96, 11578–11583 (1999). (2006). 60. Marinus, M. G. Recombination is essential for
13. Marinus, M. G. & Casadesus, J. Roles of DNA adenine 38. Malone, T., Blumenthal, R. M. & Cheng, X. Structure-​ viability of an Escherichia coli dam (DNA adenine
methylation in host-​pathogen interactions: mismatch guided analysis reveals nine sequence motifs methyltransferase) mutant. J. Bacteriol. 182,
repair, transcriptional regulation, and more. FEMS conserved among DNA amino-​methyltransferases, 463–468 (2000).
Microbiol. Rev. 33, 488–503 (2009). and suggests a catalytic mechanism for these 61. Campellone, K. G. et al. Increased adherence
14. Sanchez-​Romero, M. A., Cota, I. & Casadesus, J. enzymes. J. Mol. Biol. 253, 618–632 (1995). and actin pedestal formation by dam-​deficient
DNA methylation in bacteria: from the methyl group 39. Bujnicki, J. M. Sequence permutations in the enterohaemorrhagic Escherichia coli O157:H7.
to the methylome. Curr. Opin. Microbiol. 25, 9–16 molecular evolution of DNA methyltransferases. Mol. Microbiol. 63, 1468–1481 (2007).
(2015). BMC Evol. Biol. 2, 3 (2002). 62. Lopez-​Garrido, J. & Casadesus, J. Regulation of
15. Casadesus, J. & Low, D. A. Programmed heterogeneity: 40. Ershova, A. S., Rusinov, I. S., Spirin, S. A., Salmonella enterica pathogenicity island 1 by DNA
epigenetic mechanisms in bacteria. J. Biol. Chem. 288, Karyagina, A. S. & Alexeevski, A. V. Role of restriction-​ adenine methylation. Genetics 184, 637–649 (2010).
13929–13935 (2013). modification systems in prokaryotic evolution and 63. Herman, G. E. & Modrich, P. Escherichia coli dam
16. Casadesús, J. & Torreblanca, J. in Epigenetic ecology. Biochem. 80, 1373–1386 (2015). methylase. Physical and catalytic properties of the
Mechanisms of Gene Regulation (eds. Russo, V. E. A., 41. Oliveira, P. H., Touchon, M. & Rocha, E. P. The interplay homogeneous enzyme. J. Biol. Chem. 257, 2605–2612
Martienssen, R. A. & Riggs, A. D.) 141–153 of restriction-​modification systems with mobile genetic (1982).
(Cold Spring Harbor Laboratory, 1996). elements and their prokaryotic hosts. Nucleic Acids 64. Boye, E., Marinus, M. G. & Lobner-​Olesen, A.
17. Atack, J. M., Tan, A., Bakaletz, L. O., Jennings, M. P. Res. 42, 10618–10631 (2014). Quantitation of Dam methyltransferase in
& Seib, K. L. Phasevarions of bacterial pathogens: 42. Nobusato, A., Uchiyama, I. & Kobayashi, I. Diversity Escherichia coli. J. Bacteriol. 174, 1682–1685 (1992).
methylomics sheds new Light on old enemies. of restriction-​modification gene homologues in 65. Campbell, J. L. & Kleckner, N. The rate of Dam-​mediated
Trends Microbiol. 26, 715–726 (2018). Helicobacter pylori. Gene 259, 89–98 (2000). DNA adenine methylation in Escherichia coli. Gene 74,
18. Chen, C. et al. Convergence of DNA methylation and 43. Murphy, J., Mahony, J., Ainsworth, S., Nauta, A. 189–190 (1988).
phosphorothioation epigenetics in bacterial genomes. & van Sinderen, D. Bacteriophage orphan DNA 66. Low, D. A. & Casadesus, J. Clocks and switches:
Proc. Natl Acad. Sci. USA 114, 4501–4506 (2017). methyltransferases: insights from their bacterial bacterial gene regulation by DNA adenine methylation.
19. Wang, L., Jiang, S., Deng, Z., Dedon, P. C. & origin, function, and occurrence. Appl. Env. Microbiol. Curr. Opin. Microbiol. 11, 106–112 (2008).
Chen, S. DNA phosphorothioate modification— 79, 7547–7555 (2013). 67. Peterson, S. N. & Reich, N. O. Competitive Lrp
a new multi-​functional epigenetic system in bacteria. 44. Broadbent, S. E., Balbontin, R., Casadesus, J., and Dam assembly at the pap regulatory region:
FEMS Microbiol. Rev. 43, 109–122 (2019). Marinus, M. G. & van der Woude, M. YhdJ, a implications for mechanisms of epigenetic regulation.
20. Flusberg, B. A. et al. Direct detection of DNA nonessential CcrM-​like DNA methyltransferase of J. Mol. Biol. 383, 92–105 (2008).
methylation during single-​molecule, real-​time Escherichia coli and Salmonella enterica. J. Bacteriol. 68. Horton, J. R., Liebert, K., Bekes, M., Jeltsch, A. &
sequencing. Nat. Methods 7, 461–465 (2010). 189, 4325–4327 (2007). Cheng, X. Structure and substrate recognition of
21. Fang, G. et al. Genome-​wide mapping of methylated 45. Murray, I. A. et al. The non-​specific adenine DNA the Escherichia coli DNA adenine methyltransferase.
adenine residues in pathogenic Escherichia coli using methyltransferase M.EcoGII. Nucleic Acids Res. 46, J. Mol. Biol. 358, 559–570 (2006).
single-​molecule real-​time sequencing. Nat. Biotechnol. 840–848 (2018). 69. van der Woude, M. W., Braaten, B. A. & Low, D. A.
30, 1232–1239 (2012). 46. Urig, S. et al. The Escherichia coli dam DNA Evidence for global regulatory control of pilus expression
22. Clarke, J. et al. Continuous base identification methyltransferase modifies DNA in a highly in Escherichia coli by Lrp and DNA methylation: model
for single-​molecule nanopore DNA sequencing. processive reaction. J. Mol. Biol. 319, 1085–1096 building based on analysis of pap. Mol. Microbiol. 6,
Nat. Nanotechnol. 4, 265–270 (2009). (2002). 2429–2435 (1992).
23. Beaulaurier, J., Schadt, E. E. & Fang, G. Deciphering 47. Albu, R. F., Jurkowski, T. P. & Jeltsch, A. The 70. Wallecha, A., Munster, V., Correnti, J., Chan, T. &
bacterial epigenomes using modern sequencing Caulobacter crescentus DNA-(adenine-​N6)- van der Woude, M. Dam- and OxyR-​dependent phase
technologies. Nat. Rev. Genet. 20, 157–172 (2019). methyltransferase CcrM methylates DNA in a variation of agn43: essential elements and evidence
This review summarizes the technologies for distributive manner. Nucleic Acids Res. 40, for a new role of DNA methylation. J. Bacteriol. 184,
mapping bacterial methylomes. 1708–1716 (2012). 3338–3347 (2002).
24. Blow, M. J. et al. The epigenomic landscape of 48. Peterson, S. N. & Reich, N. O. GATC flanking 71. Waldron, D. E., Owen, P. & Dorman, C. J. Competitive
prokaryotes. PLOS Genet. 12, e1005854 (2016). sequences regulate Dam activity: evidence for interaction of the OxyR DNA-​binding protein and the
25. Lluch-​Senar, M. et al. Comprehensive methylome how Dam specificity may influence pap expression. Dam methylase at the antigen 43 gene regulatory
characterization of Mycoplasma genitalium and J. Mol. Biol. 355, 459–472 (2006). region in Escherichia coli. Mol. Microbiol. 44,
Mycoplasma pneumoniae at single-​base resolution. This study describes DNA sequence elements 509–520 (2002).
PLOS Genet. 9, e1003191 (2013). that influence the processivity of the Dam 72. Cota, I. et al. OxyR-​dependent formation of DNA
26. Vasu, K. & Nagaraja, V. Diverse functions of restriction-​ methyltransferase. methylation patterns in OpvABOFF and OpvABON cell
modification systems in addition to cellular defense. 49. Payelleville, A. et al. DNA adenine methyltransferase lineages of Salmonella enterica. Nucleic Acids Res.
Microbiol. Mol. Biol. Rev. 77, 53–72 (2013). (Dam) overexpression impairs Photorhabdus 44, 3595–3609 (2016).
27. De Ste Croix, M. et al. Phase-​variable methylation and luminescens motility and virulence. Front. Microbiol. 73. Brunet, Y. R., Bernard, C. S., Gavioli, M., Lloubes, R. &
epigenetic regulation by type I restriction-​modification 8, 1671 (2017). Cascales, E. An epigenetic switch involving overlapping
systems. FEMS Microbiol. Rev. 41, S3–S15 (2017). 50. Julio, S. M. et al. DNA adenine methylase is essential Fur and DNA methylation optimizes expression
28. Tan, A., Atack, J. M., Jennings, M. P. & Seib, K. L. for viability and plays a role in the pathogenesis of of a type VI secretion gene cluster. PLOS Genet. 7,
The capricious nature of bacterial pathogens: Yersinia pseudotuberculosis and Vibrio cholerae. e1002205 (2011).
phasevarions and vaccine development. Front. Infect. Immun. 69, 7610–7615 (2001). 74. Jakomin, M., Chessa, D., Baumler, A. J. & Casadesus, J.
Immunol. 7, 586 (2016). 51. Robinson, V. L., Oyston, P. C. & Titball, R. W. A dam Regulation of the Salmonella enterica std fimbrial
29. Thattai, M. & van Oudenaarden, A. Stochastic gene mutant of Yersinia pestis is attenuated and induces operon by DNA adenine methylation, SeqA, and HdfR.
expression in fluctuating environments. Genetics 167, protection against plague. FEMS Microbiol. Lett. 252, J. Bacteriol. 190, 7406–7413 (2008).
523–530 (2004). 251–256 (2005). 75. Garcia-​Pastor, L., Sanchez-​Romero, M. A., Jakomin, M.,
30. Kussell, E. & Leibler, S. Phenotypic diversity, population 52. Taylor, V. L., Titball, R. W. & Oyston, P. C. Oral Puerta-​Fernandez, E. & Casadesus, J. Regulation of
growth, and information in fluctuating environments. immunization with a dam mutant of Yersinia bistability in the std fimbrial operon of Salmonella
Science 309, 2075–2078 (2005). pseudotuberculosis protects against plague. enterica by DNA adenine methylation and transcription
31. Adhikari, S. & Curtis, P. D. DNA methyltransferases Microbiology 151, 1919–1926 (2005). factors HdfR, StdE and StdF. Nucleic Acids Res. 47,
and epigenetic regulation in bacteria. FEMS Microbiol. 53. Gonzalez, D. & Collier, J. DNA methylation by CcrM 7929–7941 (2019).
Rev. 40, 575–591 (2016). activates the transcription of two genes required for 76. Ardissone, S. et al. Cell cycle constraints and
32. Mouammine, A. & Collier, J. The impact of DNA the division of Caulobacter crescentus. Mol. Microbiol. environmental control of local DNA hypomethylation
methylation in Alphaproteobacteria. Mol. Microbiol. 88, 203–218 (2013). in alpha-​proteobacteria. PLOS Genet. 12, e1006499
110, 1–10 (2018). 54. Falker, S., Schilling, J., Schmidt, M. A. & Heusipp, G. (2016).
This recent article reviews DNA methylation in Overproduction of DNA adenine methyltransferase 77. Ringquist, S. & Smith, C. L. The Escherichia coli
alphaproteobacteria. alters motility, invasion, and the lipopolysaccharide chromosome contains specific, unmethylated dam
33. Phillips, Z. N., Husna, A. U., Jennings, M. P., O-​antigen composition of Yersinia enterocolitica. and dcm sites. Proc. Natl Acad. Sci. USA 89,
Seib, K. L. & Atack, J. M. Phasevarions of bacterial Infect. Immun. 75, 4990–4997 (2007). 4539–4543 (1992).

www.nature.com/nrmicro
Reviews

78. Wang, M. X. & Church, G. M. A whole genome epigenetic switch by CpxAR: phosphorylated CpxR 121. Sitaraman, R., Denison, A. M. & Dybvig, K. A unique,
approach to in vivo DNA–protein interactions in inhibits transition to the phase ON state by competition bifunctional site-​specific DNA recombinase from
E. coli. Nature 360, 606–610 (1992). with Lrp. Mol. Cell 16, 537–547 (2004). Mycoplasma pulmonis. Mol. Microbiol. 46,
79. Kozdon, J. B. et al. Global methylation state 101. White-​Ziegler, C. A., Angus Hill, M. L., Braaten, B. A., 1033–1040 (2002).
at base-​pair resolution of the Caulobacter genome van der Woude, M. W. & Low, D. A. Thermoregulation 122. Doberenz, S. et al. Identification of a Pseudomonas
throughout the cell cycle. Proc. Natl Acad. Sci. USA of Escherichia coli pap transcription: H-​NS is a aeruginosa PAO1 DNA methyltransferase, its targets,
110, E4658–E4667 (2013). temperature-​dependent DNA methylation blocking and physiological roles. MBio 8, e02312–e02316
80. Payelleville, A. et al. The complete methylome of an factor. Mol. Microbiol. 28, 1121–1137 (1998). (2017).
entomopathogenic bacterium reveals the existence of 102. Reyes-​Lamothe, R. & Sherratt, D. J. The bacterial 123. Manso, A. S. et al. A random six-​phase switch
loci with unmethylated adenines. Sci. Rep. 8, 12091 cell cycle, chromosome inheritance and cell growth. regulates pneumococcal virulence via global
(2018). Nat. Rev. Microbiol. 17, 467–478 (2019). epigenetic changes. Nat. Commun. 5, 5055 (2014).
81. Hale, W. B., van der Woude, M. W. & Low, D. A. 103. Campbell, J. L. & Kleckner, N. E. coli oric and the This study, together with Li et al. (2016) and
Analysis of nonmethylated GATC sites in the Escherichia dnaA gene promoter are sequestered from dam Oliver et al. (2017), characterizes a phase variation
coli chromosome and identification of sites that are methyltransferase following the passage of the mechanism that controls switching between
differentially methylated in response to environmental chromosomal replication fork. Cell 62, 967–979 pneumococcal phenotypic forms.
stimuli. J. Bacteriol. 176, 3438–3441 (1994). (1990). 124. Li, J. et al. Epigenetic switch driven by DNA inversions
82. Li, D. et al. Exocyclic carbons adjacent to the N6 of 104. Cagliero, C., Grand, R. S., Jones, M. B., Jin, D. J. dictates phase variation in Streptococcus pneumoniae.
adenine are targets for oxidation by the Escherichia & O’Sullivan, J. M. Genome conformation capture PLOS Pathog. 12, e1005762 (2016).
coli adaptive response protein AlkB. J. Am. Chem. Soc. reveals that the Escherichia coli chromosome is 125. Oliver, M. B., Basu Roy, A., Kumar, R., Lefkowitz, E. J.
134, 8896–8901 (2012). organized by replication and transcription. Nucleic & Swords, W. E. Streptococcus pneumoniae TIGR4
This report describes active demethylation of 6mA Acids Res. 41, 6058–6071 (2013). phase-​locked opacity variants differ in virulence
during DNA repair. 105. Modrich, P. Methyl-​directed DNA mismatch correction. phenotypes. mSphere 2, e00386-17 (2017).
83. Polaczek, P., Kwan, K. & Campbell, J. L. GATC motifs J. Biol. Chem. 264, 6597–6600 (1989). 126. Estibariz, I. et al. The core genome m5C
may alter the conformation of DNA depending on 106. Cota, I., Blanc-​Potard, A. B. & Casadesus, J. STM2209- methyltransferase JHP1050 (M.Hpy99III) plays
sequence context and N6-adenine methylation status: STM2208 (opvAB): a phase variation locus of an important role in orchestrating gene expression
possible implications for DNA–protein recognition. Salmonella enterica involved in control of O-​antigen in Helicobacter pylori. Nucleic Acids Res. 47,
Mol. Gen. Genet. 258, 488–493 (1998). chain length. PLOS ONE 7, e36863 (2012). 2336–2348 (2019).
84. Kimura, T., Asai, T., Imai, M. & Takanami, M. 107. Cota, I. et al. Epigenetic control of Salmonella enterica This article demonstrates direct transcriptional
Methylation strongly enhances DNA bending in O-​antigen chain length: a tradeoff between virulence control of bacterial genes by 5mC.
the replication origin region of the Escherichia coli and bacteriophage resistance. PLOS Genet. 11, 127. Chao, M. C. et al. A cytosine methyltransferase
chromosome. Mol. Gen. Genet. 219, 69–74 (1989). e1005667 (2015). modulates the cell envelope stress response in the
85. Diekmann, S. DNA methylation can enhance or induce 108. Chessa, D., Winter, M. G., Jakomin, M. & Baumler, A. J. cholera pathogen. PLOS Genet. 11, e1005666 (2015).
DNA curvature. EMBO J. 6, 4213–4217 (1987). Salmonella enterica serotype Typhimurium Std 128. Kahramanoglou, C. et al. Genomics of DNA cytosine
86. Waldminghaus, T. & Skarstad, K. The Escherichia coli fimbriae bind terminal alpha(1,2)fucose residues in methylation in Escherichia coli reveals its role in
SeqA protein. Plasmid 61, 141–150 (2009). the cecal mucosa. Mol. Microbiol. 71, 864–875 stationary phase transcription. Nat. Commun. 3, 886
87. Sanchez-​Romero, M. A. et al. Dynamic distribution of (2009). (2012).
SeqA protein across the chromosome of Escherichia 109. De Bolle, X. et al. The length of a tetranucleotide 129. Militello, K. T., Mandarano, A. H., Varechtchouk, O.
coli K-12. Mbio 1, e00012-10 (2010). repeat tract in Haemophilus influenzae determines & Simon, R. D. Cytosine DNA methylation influences
88. Camacho, E. M. & Casadesus, J. Conjugal transfer the phase variation rate of a gene with homology to drug resistance in Escherichia coli through increased
of the virulence plasmid of Salmonella enterica is type III DNA methyltransferases. Mol. Microbiol. 35, sugE expression. FEMS Microbiol. Lett. 350,
regulated by the leucine-​responsive regulatory protein 211–222 (2000). 100–106 (2014).
and DNA adenine methylation. Mol. Microbiol. 44, 110. de Vries, N. et al. Transcriptional phase variation 130. Kumar, S. et al. N4-cytosine DNA methylation regulates
1589–1598 (2002). of a type III restriction-​modification system in transcription and pathogenesis in Helicobacter pylori.
89. Camacho, E. M. & Casadesus, J. Regulation of traJ Helicobacter pylori. J. Bacteriol. 184, 6615–6623 Nucleic Acids Res. 46, 3429–3445 (2018).
transcription in the Salmonella virulence plasmid (2002). 131. Budroni, S. et al. Neisseria meningitidis is structured
by strand-​specific DNA adenine hemimethylation. 111. Srikhanta, Y. N., Maguire, T. L., Stacey, K. J., in clades associated with restriction modification
Mol Microbiol 57, 1700–1718 (2005). Grimmond, S. M. & Jennings, M. P. The phasevarion: systems that modulate homologous recombination.
90. Fioravanti, A. et al. DNA binding of the cell cycle a genetic system controlling coordinated, random Proc. Natl Acad. Sci. USA 108, 4494–4499 (2011).
transcriptional regulator GcrA depends on N6-adenosine switching of expression of multiple genes. Proc. Natl 132. Nandi, T. et al. Burkholderia pseudomallei sequencing
methylation in Caulobacter crescentus and other Acad. Sci. USA 102, 5547–5551 (2005). identifies genomic clades with distinct recombination,
Alphaproteobacteria. PLOS Genet. 9, e1003541 (2013). 112. Fox, K. L. et al. Haemophilus influenzae phasevarions accessory, and epigenetic profiles. Genome Res. 25,
91. Roberts, D., Hoopes, B. C., McClure, W. R. & have evolved from type III DNA restriction systems 608 (2015).
Kleckner, N. IS10 transposition is regulated by DNA into epigenetic regulators of gene expression. Nucleic 133. Kobayashi, I. Behavior of restriction-​modification
adenine methylation. Cell 43, 117–130 (1985). Acids Res. 35, 5242–5252 (2007). systems as selfish mobile elements and their impact
92. Camacho, E. M. et al. Regulation of finP transcription 113. Srikhanta, Y. N. et al. Phasevarions mediate random on genome evolution. Nucleic Acids Res. 29,
by DNA adenine methylation in the virulence plasmid switching of gene expression in pathogenic Neisseria. 3742–3756 (2001).
of Salmonella enterica. J. Bacteriol. 187, 5691–5699 PLOS Pathog. 5, e1000400 (2009). 134. Tomcsanyi, T. & Berg, D. E. Transposition effect of
(2005). 114. Srikhanta, Y. N., Fox, K. L. & Jennings, M. P. adenine (Dam) methylation on activity of O end
93. van der Woude, M., Braaten, B. & Low, D. Epigenetic The phasevarion: phase variation of type III DNA mutants of IS50. J. Mol. Biol. 209, 191–193 (1989).
phase variation of the pap operon in Escherichia coli. methyltransferases controls coordinated switching 135. Alonso, A., Pucciarelli, M. G., Figueroa-​Bossi, N.
Trends Microbiol 4, 5–9 (1996). in multiple genes. Nat. Rev. Microbiol. 8, 196–206 & Garcia-​del Portillo, F. Increased excision of the
94. Broadbent, S. E., Davies, M. R. & van der Woude, M. (2010). Salmonella prophage ST64B caused by a deficiency
W. Phase variation controls expression of Salmonella 115. Kwiatek, A., Mrozek, A., Bacal, P., Piekarowicz, A. & in Dam methylase. J. Bacteriol. 187, 7901–7911
lipopolysaccharide modification genes by a DNA Adamczyk-​Poplawska, M. Type III methyltransferase (2005).
methylation-​dependent mechanism. Mol. Microbiol. M.NgoAX from Neisseria gonorrhoeae FA1090 136. Murphy, K. C., Ritchie, J. M., Waldor, M. K.,
77, 337–353 (2010). regulates biofilm formation and interactions with Lobner-​Olesen, A. & Marinus, M. G. Dam
95. Hernday, A. D., Braaten, B. A. & Low, D. A. The human cells. Front. Microbiol. 6, 1426 (2015). methyltransferase is required for stable lysogeny of
mechanism by which DNA adenine methylase and 116. Blakeway, L. V. et al. Moraxella catarrhalis restriction-​ the Shiga toxin (Stx2)-encoding bacteriophage 933W
PapI activate the pap epigenetic switch. Mol. Cell 12, modification systems are associated with phylogenetic of enterohemorrhagic Escherichia coli O157:H7.
947–957 (2003). lineage and disease. Genome Biol. Evol. 10, J. Bacteriol. 190, 438–441 (2008).
96. Davies, M. R., Broadbent, S. E., Harris, S. R., 2932–2946 (2018). 137. Wang, T. C. & Smith, K. C. Inviability of dam recA and
Thomson, N. R. & van der Woude, M. W. Horizontally 117. Atack, J. M., Yang, Y., Seib, K. L., Zhou, Y. & dam recB cells of Escherichia coli is correlated with
acquired glycosyltransferase operons drive Jennings, M. P. A survey of Type III restriction-​ their inability to repair DNA double-​strand breaks
salmonellae lipopolysaccharide diversity. PLOS Genet. modification systems reveals numerous, novel produced by mismatch repair. J. Bacteriol. 165,
9, e1003568 (2013). epigenetic regulators controlling phase-​variable 1023–1025 (1986).
97. Garcia-​Pastor, L., Sanchez-​Romero, M. A., regulons: phasevarions. Nucleic Acids Res. 46, 138. Veening, J. W., Smits, W. K. & Kuipers, O. P. Bistability,
Gutierrez, G., Puerta-​Fernandez, E. & Casadesus, J. 3532–3542 (2018). epigenetics, and bet-​hedging in bacteria. Annu. Rev.
Formation of phenotypic lineages in Salmonella A comprehensive bioinformatic search for phase-​ Microbiol. 62, 193–210 (2008).
enterica by a pleiotropic fimbrial switch. PLOS Genet. variable type III R-​M DNA methyltransferases. 139. Maynard-​Smith, J. Evolution and the Theory of Games
14, e1007677 (2018). 118. Gawthorne, J. A., Beatson, S. A., Srikhanta, Y. N., (Cambridge University Press, 1982).
98. Suwandi, A. et al. Std fimbriae–fucose interaction Fox, K. L. & Jennings, M. P. Origin of the diversity in 140. Roth, J. R., Kugelberg, E., Reams, A. B., Kofoid, E. &
increases Salmonella-​induced intestinal inflammation DNA recognition domains in phasevarion associated Andersson, D. I. Origin of mutations under selection:
and prolongs colonization. PLOS Pathog. 15, modA genes of pathogenic Neisseria and Haemophilus the adaptive mutation controversy. Annu. Rev.
e1007915 (2019). influenzae. PLOS ONE 7, e32337 (2012). Microbiol. 60, 477–501 (2006).
99. Weyand, N. J., Braaten, B. A., van der Woude, M., 119. Srikhanta, Y. N. et al. Methylomic and phenotypic 141. Tanner, J. R. & Kingsley, R. A. Evolution of Salmonella
Tucker, J. & Low, D. A. The essential role of the analysis of the ModH5 phasevarion of Helicobacter within hosts. Trends Microbiol. 26, 986–998 (2018).
promoter-​proximal subunit of CAP in pap phase pylori. Sci. Rep. 7, 16140 (2017). 142. Turrientes, M. C. et al. Normal mutation rate variants
variation: Lrp- and helical phase-​dependent activation 120. Anjum, A. et al. Phase variation of a Type IIG arise in a mutator (MutS) Escherichia coli population.
of papBA transcription by CAP from -215. Mol. restriction-​modification enzyme alters site-​specific PLOS ONE 8, e72963 (2013).
Microbiol. 39, 1504–1522 (2001). methylation patterns and gene expression in 143. Furuta, Y. & Kobayashi, I. Movement of DNA sequence
100. Hernday, A. D., Braaten, B. A., Broitman-​Maduro, G., Campylobacter jejuni strain NCTC11168. Nucleic recognition domains between non-​orthologous
Engelberts, P. & Low, D. A. Regulation of the pap Acids Res. 44, 4581–4594 (2016). proteins. Nucleic Acids Res. 40, 9218–9232 (2012).

Nature Reviews | Microbiology


Reviews

144. Coulondre, C., Miller, J. H., Farabaugh, P. J. & Gilbert, W. 155. Cohen, N. R. et al. A role for the bacterial GATC 165. Badie, G., Heithoff, D. M. & Mahan, M. J. LcrV
Molecular basis of base substitution hotspots in methylome in antibiotic stress survival. Nat. Genet. synthesis is altered by DNA adenine methylase
Escherichia coli. Nature 274, 775–780 (1978). 48, 581–586 (2016). overproduction in Yersinia pseudotuberculosis and
145. Lieb, M. & Bhagwat, A. S. Very short patch repair: 156. Maier, J. A. H., Mohrle, R. & Jeltsch, A. Design of is required to confer immunity in vaccinated hosts.
reducing the cost of cytosine methylation. Mol. synthetic epigenetic circuits featuring memory effects Infect. Immun. 72, 6707–6710 (2004).
Microbiol. 20, 467–473 (1996). and reversible switching based on DNA methylation. 166. Pucciarelli, M. G., Prieto, A. I., Casadesus, J.
146. Hershberg, R. & Petrov, D. A. Evidence that mutation Nat. Commun. 8, 15336 (2017). & Garcia-​del Portillo, F. Envelope instability in DNA
is universally biased towards AT in bacteria. PLOS 157. Olivenza, D. R. et al. A portable epigenetic switch adenine methylase mutants of Salmonella enterica.
Genet. 6, e1001115 (2011). for bistable gene expression in bacteria. Sci. Rep. 9, Microbiology 148, 1171–1182 (2002).
147. Lobry, J. R. & Sueoka, N. Asymmetric directional 11261 (2019). 167. Fumeaux, C. et al. Cell cycle transition from S-​phase to
mutation pressures in bacteria. Genome Biol. 3, 158. Nou, X. et al. Regulation of pyelonephritis-​associated G1 in Caulobacter is mediated by ancestral virulence
RESEARCH0058 (2002). pili phase-​variation in Escherichia coli: binding of the regulators. Nat. Commun. 5, 4081 (2014).
148. Pereira, J. M., Hamon, M. A. & Cossart, P. A lasting PapI and the Lrp regulatory proteins is controlled
impression: epigenetic memory of nacterial infections? by DNA methylation. Mol. Microbiol. 7, 545–553 Acknowledgements
Cell Host Microbe. 19, 579–582 (2016). (1993). Work in the authors’ laboratory is supported by grant
149. Niller, H. H. & Minarovits, J. Patho-​epigenetics of 159. Heithoff, D. M. et al. Salmonella DNA adenine BIO2016-75235-P from the Ministerio de Ciencia, Innovación
infectious diseases caused by intracellular bacteria. methylase mutants confer cross-​protective immunity. y Universidades of Spain and the European Regional Fund.
Adv. Exp. Med. Biol. 879, 107–130 (2016). Infect. Immun. 69, 6725–6730 (2001). They are grateful to M. van der Woude and L. García-​Pastor
150. Bierne, H. in Epigenetics of Infectious Diseases 160. Heithoff, D. M., House, J. K., Thomson, P. C. & for discussions.
(eds Doerfler, W. & Casadesus, J.) 113–158 Mahan, M. J. Development of a Salmonella cross-​
(Springer, 2017). protective vaccine for food animal production systems.
Author contributions
151. Chernov, A. V. et al. Mycoplasma CG- and GATC-​ Vaccine 33, 100–107 (2015).
J.C. and M.A.S.-R. contributed to discussion of the content,
specific DNA methyltransferases selectively and 161. Xiao, C. L. et al. N6-methyladenine DNA modification
wrote the article, reviewed and edited the manuscript before
efficiently methylate the host genome and alter the in the human genome. Mol. Cell 71, 306–318.e7
submission, and researched data for the article.
epigenetic landscape in human cells. Epigenetics 10, (2018).
303–318 (2015). 162. Lopez-​Garrido, J. & Casadesus, J. Crosstalk between
152. Mashhoon, N., Pruss, C., Carroll, M., Johnson, P. H. virulence loci: regulation of Salmonella enterica Competing interests
& Reich, N. O. Selective inhibitors of bacterial DNA pathogenicity island 1 (SPI-1) by products of the std The authors declare no competing interests.
adenine methyltransferases. J. Biomol. Screen 11, fimbrial operon. PLOS ONE 7, e30499 (2012).
497–510 (2006). 163. Braaten, B. A. et al. Leucine-​responsive regulatory Peer review information
153. Benkovic, S. J. et al. Identification of borinic esters protein controls the expression of both the pap and Nature Reviews Microbiology thanks M. R. Oggioni and the
as inhibitors of bacterial cell growth and bacterial fan pili operons in Escherichia coli. Proc. Natl Acad. other, anonymous, reviewer(s) for their contribution to the peer
methyltransferases, CcrM and MenH. J. Med. Chem. Sci. USA 89, 4250–4254 (1992). review of this work.
48, 7468–7476 (2005). 164. Waldminghaus, T., Weigel, C. & Skarstad, K.
154. Ceccaldi, A. et al. Identification of novel inhibitors of Replication fork movement and methylation govern Publisher’s note
DNA methylation by screening of a chemical library. SeqA binding to the Escherichia coli chromosome. Springer Nature remains neutral with regard to jurisdictional
ACS Chem. Biol. 8, 543–548 (2013). Nucleic Acids Res. 40, 5465–5476 (2012). claims in published maps and institutional affiliations.

www.nature.com/nrmicro

You might also like