Sie sind auf Seite 1von 33

Enterococcal Genetics

KEITH E. WEAVER
Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069

ABSTRACT The study of the genetics of enterococci has enously acquired DNA (6). Further genome sequencing
focused heavily on mobile genetic elements present in these has revealed extensive variability in the proportion of
organisms, the complex regulatory circuits used to control
horizontally acquired DNA in E. faecalis strains, with the
their mobility, and the antibiotic resistance genes they frequently
carry. Recently, more focus has been placed on the regulation of
presence of clustered regularly interspaced short palin-
genes involved in the virulence of the opportunistic pathogenic dromic repeats (CRISPR) with CRISPR-associated (Cas)
species Enterococcus faecalis and Enterococcus faecium. Little protein (CRISPR-Cas) loci playing an important role in
information is available concerning fundamental aspects of DNA regulating MGE acquisition (7, 8). More recent genomic
replication, partition, and division; this article begins with a brief analysis has revealed that MGEs are important drivers of
overview of what little is known about these issues, primarily by genome diversity and evolution in Enterococcus faecium
comparison with better-studied model organisms. A variety of as well (9).
transcriptional and posttranscriptional mechanisms of regulation
While work on enterococcal MGEs continues apace,
of gene expression are then discussed, including a section on the
genetics and regulation of vancomycin resistance in enterococci. other aspects of enterococcal genetics are beginning to
The article then provides extensive coverage of the pheromone- come into sharper focus. While the basic mechanisms of
responsive conjugation plasmids, including sections on regulation chromosome replication, partition, and cell division re-
of the pheromone response, the conjugative apparatus, and main poorly defined, progress on other chain-forming
replication and stable inheritance. The article then focuses on ovococci provides a framework for further detailed anal-
conjugative transposons, now referred to as integrated, ysis. Meanwhile, substantial progress has been made in
conjugative elements, or ICEs, and concludes with several
understanding transcription and posttranscriptional reg-
smaller sections covering emerging areas of interest concerning
the enterococcal mobilome, including nonpheromone plasmids
ulation of gene expression, intercellular communication,
of particular interest, toxin-antitoxin systems, pathogenicity and global response via second messengers, particularly
islands, bacteriophages, and genome defense. of genes involved in virulence of the two major pathogens

Interest in enterococcal genetics began with three land- Received: 4 December 2018, Accepted: 8 January 2019,
Published: 8 March 2019
mark discoveries: (i) identification of the first conjugative Editors: Vincent A. Fischetti, The Rockefeller University, New York,
plasmids whose transfer systems are induced by an iden- NY; Richard P. Novick, Skirball Institute for Molecular Medicine, NYU
tifiable signal (1), (ii) identification of the first “transpo- Medical Center, New York, NY; Joseph J. Ferretti, Department of
Microbiology & Immunology, University of Oklahoma Health
sons” capable of intercellular (conjugative) transposition Science Center, Oklahoma City, OK; Daniel A. Portnoy, Department
(2, 3), and (iii) the acquisition of vancomycin resistance of Molecular and Cellular Microbiology, University of California,
Berkeley, Berkeley, CA; Miriam Braunstein, Department of
(4, 5). Since the most prevalent antibiotic resistance genes Microbiology and Immunology, University of North Carolina-Chapel
are located on plasmids and transposons, early work on Hill, Chapel Hill, NC, and Julian I. Rood, Infection and Immunity
enterococcal genetics focused heavily on mobile genetic Program, Monash Biomedicine Discovery Institute, Monash
University, Melbourne, Australia
elements (MGEs). Examination of the complete sequence Citation: Weaver K. 2019. Enterococcal Genetics. Microbiol
of a vancomycin-resistant clinical isolate of Enterococ- Spectrum 7(2):GPP3-0055-2018. doi:10.1128/microbiolspec.GPP3-
cus faecalis, V583, reaffirmed the importance of MGEs 0055-2018.
in the evolution of this species, revealing that over a Correspondence: Keith E. Weaver, kweaver@usd.edu
© 2019 American Society for Microbiology. All rights reserved.
quarter of the genome consists of mobile and/or exog-

ASMscience.org/MicrobiolSpectrum 1
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

of the genus, E. faecalis and E. faecium (for a compre- (mutS and mutL homologs annotated as hexA and
hensive review see 10). This article will focus primarily on hexB, respectively), and numerous other nucleotide re-
the most recent advances in this field, with references to pair proteins, including seven potential mutT homo-
detailed reviews where available. Unless otherwise stated, logs. It should be noted that the previously designated
identification of E. faecalis genes orthologous to genes uvrABC genes located on plasmid pAD1 (17) are not
in other bacteria utilized the V583 genome for searches homologs of the excision repair enzymes that usually
(6), and gene annotations where given will use the V583 bear this name but are instead associated with a lesion
designation. bypass polymerase. Multiple homologs of genes involved
in recombination repair were also identified, including
genes for a LexA-like regulator, RecA, a RexAB DNA
CHROMOSOMAL REPLICATION, PARTITION, processing enzyme, a RuvAB Holliday junction heli-
AND CELL DIVISION case, a PriA replication restart primase, a RecFOR com-
The fundamentals of chromosomal DNA replication in plex, and a variety of other recombination proteins. A
enterococci are probably similar to those in the model recombination-deficient mutant of E. faecalis, UV202,
organism Bacillus subtilis (11). The E. faecalis genome has a phenotype similar to recA mutants in other bacteria
includes homologs of all known components of the typ- (18). Sequencing and complementation analyses con-
ical Gram-positive DNA polymerase III (Pol III) holo- firmed that this phenotype is due to a single point mu-
enzyme, including a replicative DNA helicase (which is tation that results in a G265D change in the RecA protein
named DnaB according to the Escherichia coli conven- (19). Other putative recombinational repair proteins have
tion rather than DnaC as in B. subtilis), helicase loaders, not been characterized.
primase, clamp and clamp loaders, and DnaE- and PolC- The E. faecalis genome includes topoisomerases I to
type catalytic alpha subunits. Most have also been iden- IV, a Streptococcus pneumoniae condensin homolog, and
tified in the E. faecium genome. It has been proposed that a homolog for the nucleoid associated protein HU, sug-
PolC may be the primary leading strand DNA poly- gesting that the chromosomal DNA is organized and
merase, while DnaE plays dual roles as the lagging strand compacted as in other bacteria. No replication termina-
polymerase and as an error-prone polymerase during tor protein is annotated in the genome, and no identifi-
DNA repair in B. subtilis and Streptococcus pyogenes able homolog of the B. subtilis Rtp terminator could be
(12, 13). The E. faecalis PolC and DnaE proteins have identified. In fact, Rtp appears to be limited to the Bacillus
been purified and found to have characteristics similar to genus, suggesting that proteins involved in termination of
the B. subtilis homologs, even cross-reacting with anti- replication are poorly conserved. A single XerD homo-
bodies generated against B. subtilis PolC and DnaE (14), log, involved in chromosome dimer resolution in other
suggesting that E. faecalis and B. subtilis enzymes are bacteria, is annotated in the E. faecalis genome, but
structurally and functionally similar. The putative en- several other integrase/recombinases have been identified
terococcal oriC has a bipartite organization with DnaA that could play the role of the XerCD resolution recom-
binding sites flanking the dnaA gene as in B. subtilis and binase. The C-terminus of the cell division protein FtsK has
distinct from E. coli. been implicated in organizing both the XerCD dimer res-
In addition to DNA Pol III, the E. faecalis V583 chro- olution complex and the topoisomerase IV decatenation
mosome also encodes a DNA Pol I homolog, two putative complex near the chromosomal replication terminus for
phage-encoded DNA polymerases, and one member of efficient separation of daughter chromosomes in E. coli
the mutagenic Y-family polymerases (15) that have been (20, 21). The C-terminal domain of E. faecalis FtsK is 42%
shown to be involved in lesion bypass in other organisms. identical to the C-terminus of E. coli FtsK, but the N-
Y-family polymerases are commonly found on bacterial terminal domains of the two proteins are not similar,
plasmids (16), and all three of the plasmids in E. faecalis suggesting that they may play similar roles at the end of
V583 encode at least one member of the family. The pre- chromosomal replication but different roles in cell division.
viously described uvrA gene of E. faecalis plasmid pAD1 The division/cell wall cluster of E. faecalis was iden-
(17) also belongs to this group and was shown to impart tified in strain A24836 (22) and appears to be identical
increased resistance to UV exposure. in the V583 genome (note that divIB and ftsQ are or-
Other genes associated with DNA repair present in thologs). This cluster includes ftsZ, the gene for the bac-
the E. faecalis genome include homologs required for terial tubulin homolog essential for contraction of the cell
transcription-coupled DNA repair (mfd), nucleotide ex- membrane during cell division, two other cell division
cision repair (uvrA, uvrB, and uvrC), mismatch repair genes, ftsA and ftsQ, and several genes involved in pep-

2 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

tidoglycan synthesis. There are also multiple ftsW- and is required to determine how similar coordination of
ftsK-like genes in the V583 genome that may be involved chromosome partition and cell division are in the en-
in cell division. Details of the mechanism of FtsZ ring terococci and streptococci.
closure have been examined in S. pneumoniae (23–25)
and may be applicable to enterococci.
In the model rod-shaped organisms E. coli and B. sub- REGULATION OF GENE EXPRESSION
tilis the primary systems responsible for division site se- Like many other aspects of enterococcal genetics,
lection and coordination with chromosome segregation regulation of gene expression was initially investigated
are the Min and nucleoid occlusion systems (26, 27). in the MGEs of E. faecalis. These include the two-
In the Min system, the MinCD division inhibitor func- component signal transduction system (TCSTS) regula-
tions with a topological specificity determinant (MinE in tion of transposon-borne vancomycin resistance genes,
E. coli and DivIVA in B. subtilis) to prevent division site signal peptide-dependent and antisense RNA-mediated
placement at the poles, while nucleoid occlusion prevents regulation of conjugation, and antisense RNA regula-
septum closure over the bulk chromosomal DNA. Gram- tion of plasmid-encoded toxin-antitoxin (TA) genes. The
positive cocci lack recognizable genes for MinCD and mechanisms of regulation of these systems will be exam-
known nucleoid occlusion systems, and evidence suggests ined in detail in later sections of the article. This section
that nucleoid occlusion does not function in these organ- will focus on more recent advances in the understanding
isms (28, 29). Recent work in S. pneumoniae suggests of the regulation of enterococcal chromosomal genes.
that proper origin placement and chromosome compac- The E. faecalis genome encodes genes for RNA poly-
tion drive division site selection, and a protein unique merase α, β, β′, ω, and δ subnits and four recognizable
to the Enterococcaceae and Streptococcaceae families, σ factors. The σ factor genes include a putative house-
MapZ (30), is required for proper perpendicular orien- keeping σ factor (rpoD or sigA), two members of the
tation of the division plane (31). The mechanism of origin extracytoplasmic family (ECF), and a σ54-like sigma fac-
placement remains unclear but may involve as yet un- tor (rpoN). σ54 is unique among bacterial sigma fac-
identified proteins, coupling of transcription to segrega- tors in that σ54 RNA polymerase is unable to form open
tion (32), or entropic effects (31). complexes and initiate transcription without the help of
The S. pneumoniae system provides a useful model for ATP-dependent activator proteins (35). Such “enhancer
understanding enterococcal partition and cell division. binding proteins” are highly conserved, and genes en-
Enterococcal division is at least superficially similar to coding four such proteins have been identified in the
streptococcal cell division, and the enterococcal chro- E. faecalis genome (36). All belong to the LevR subfam-
mosome includes orthologs for many of the important ily of σ54 activators, which includes a conserved DEAH
players, including genes for partition, chromosome com- helicase motif (37). Immediately downstream of each of
paction, and MapZ. But there are also several potentially these genes is a putative σ54 promoter and operons en-
significant differences. First, the S. pneumoniae parB coding four distinct sugar phosphotransferase systems
gene, whose product is required for proper partition and (PTSs) of the mannose family (36). Mutation of σ54, en-
recruitment of the nucleoid organizing protein Smc (33), hancer binding protein MptR, or PTS component MptD
is located immediately adjacent to the origin region and is resulted in decreased sensitivity to class IIa bacteriocins,
not genetically linked to a gene for the ParA protein, implicating MptD as the bacteriocin receptor and σ54
which is normally essential for ParB-mediated partition and MptR as its regulators (36, 38). V583 σ54 mutants
function. The closest S. pneumoniae parB homolog on showed increased biofilm formation while decreasing au-
the E. faecalis genome (EF3298) is not closely linked to tolysis, cell death, and extracellular DNA release (39).
the putative origin region and has a clear parA partner This phenotype was independent of the four known en-
(EF3299). Gene order at the E. faecalis and E. faecium hancer binding proteins. Because biofilm development is
origins conform more closely to that found in B. subtilis believed to be dependent on extracellular DNA, it was
than in S. pneumoniae (34). Further, the homology be- proposed that σ54 mutants adapt an alternative matrix
tween S. pneumoniae and enterococcal MapZ is pri- for biofilm establishment. The E. faecium genome also
marily in the C-terminal peptidoglycan binding domain encodes an rpoN and five enhancer binding proteins of
and does not extend to the transmembrane and cyto- the LevR family, but the functions of the genes are so far
plasmic domains potentially involved in midcell divi- undefined.
some placement. While these observations may simply ECF σ factors generally facilitate transcription of genes
represent variations on a general theme, further research involved in bacterial stress responses. They are seques-

ASMscience.org/MicrobiolSpectrum 3
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

tered by membrane-localized anti-σ factors and released also modulate intrinsic cephalosporin resistance by an
by stress-induced anti-σ factor degradation by regulated unknown mechanism (58–61).
intramembrane proteolysis (40, 41). The SigV ECF of The GrvRS TCSTS system, also known as EtaRS, is
E. faecalis was shown to contribute to survival following orthologous to the important S. pyogenes virulence reg-
heat, acid, and ethanol stress (42) and to play a key role in ulatory TCSTS CovRS (62). In E. faecalis it has been
lysozyme resistance and virulence (43). The gene adjacent demonstrated to be involved in virulence as well as re-
to sigV, rsiV, encodes the anti-σ factor of the system (42) sistance to low pH and high temperature (63). GrvRS has
and is targeted by the Eep membrane-embedded zinc also been implicated in the regulation of the important
metalloprotease and at least one other unidentified pro- extracellular matrix adhesin Ace (64), catalase (65), and
tease for degradation in response to lysozyme and per- the heat shock proteins DnaK and GroEL (66). It seems
haps other stresses (44). The function of the second E. likely that the GrvRS regulon will continue to grow with
faecalis ECF σ factor is unknown. further investigation.
TCSTSs have emerged as ubiquitous bacterial signal- Disruption of RR 04, 08, and 18 (as defined in 46)
ing systems for regulating gene expression in response to each resulted in sensitivity to heat, and transcription of
environmental conditions (45). The canonical TCSTSs HK-RR 07, 13, and 14 was induced by heat, suggesting
are composed of a sensor histidine kinase (HK) that that these TCSTSs may also play a role in stress resis-
modulates a response regulator (RR) via phosphotrans- tance (66).
fer. The RR is frequently a transcriptional regulator. In E. faecium the EntKR TCSTS regulates expression
Analysis of the E. faecalis genome revealed the pres- of the bacteriocin enterocin A (67). EntKR is a quorum
ence of 17 HK/RR pairs and 1 orphan RR (46). The best- sensing system that responds to a small peptide produced
characterized enterococcal TCSTSs are the Van system, from the same locus, designated EntF. There appear to
required for regulation of vancomycin resistance genes be no close homologs to the EntKR system in E. fae-
(see below), and the Fsr system. The Fsr system regulates calis. The ChtRS TCSTS contributes to resistance to both
production of two proteases, gelatinase and serine pro- bacitracin and the disinfectant chlorhexidine (68), and
tease, that are associated with virulence and biofilm for- CroRS is associated with intrinsic cephalosporin resis-
mation (47–51). Fsr is a quorum sensing system that tance (57). Clear homologs of E. faecalis HK-RR 01, 03,
responds to the accumulation of a peptide lactone, des- 04, 05, 07, 09, and 10 (as defined in 46) are present in the
ignated GBAP (52). GBAP is the product of a small open E. faecium genome sequence. In addition, a system with
reading frame, fsrD, that is processed and secreted by linked homologs of the S. aureus agrA, agrB, and agrC
FsrB (53). GBAP is sensed at high cell density by the FsrC genes is present in E. faecium, but this system is not
HK, which then activates the FsrB RR by phosphoryla- closely related to the E. faecalis fsr system. The E. fae-
tion. Phosphorylated FsrB then activates transcription cium genome contains 10 more annotated RRs and 9
from the gelE-sprE target promoter and the fsrBDC pro- more HKs that either have no strong homology to coun-
moter, completing the autoinduction loop. The fsr system terparts in the E. faecalis genome or are no more closely
is analogous to the well-studied agr system of Staphylo- related to E. faecalis genes than to genes from any other
coccus aureus (54) except for the absence of the global Firmicutes. Therefore, E. faecalis and E. faecium have
small RNA (sRNA) regulator RNA III; FsrA appears to be about the same number of TCSTSs but not necessarily the
the terminal regulator of the Fsr system. same complement of homologs.
The CroRS TCSTS is essential for PBP5-mediated The E. faecalis cyl cytolysin-producing operon is in-
intrinsic cephalosporin resistance in both E. faecalis and duced by a quorum sensing mechanism operating through
E. faecium (55–57). The CroS HK is a sensor of cell a two-component system, but in this case the sensor
wall homeostasis and is stimulated to phosphorylate the and regulator show no homology to HK/RR of typical
CroR RR in response to a number of cell wall-active TCSTSs (69). Active cytolysin is composed of two post-
antibiotics. The CroR-regulated effector genes respon- translationally modified peptides, CylLL and CylLS, the
sible for cephalosporin resistance have not been identi- latter acting as the autoinducer of the system. CylR2 is
fied, but neither PBP5 itself nor its substrates appear to a DNA binding protein that represses expression of the
be the primary target. In some E. faecalis strains, cross- cyl operon encoding the structural genes for the toxin,
regulation is observed with a second TCSTS CisRS (56). an enzyme for modification of the toxin peptides, and
The situation is further complicated by the involvement of an ABC transporter for secretion. The structure of the
a transmembrane serine/threonine kinase, IreK, its cog- CylR2-promoter interaction has been determined (70).
nate phosphatase, IreP, and their substrate, IreB, which The CylLS inducing signal is transmitted to CylR2 via

4 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

the membrane receptor CylR1. The system is sensitive these regulators, acting as a transcriptional activator of
to both bacterial cell density and the presence of target at least six genes encoding catalase, glutathione reduc-
cells. Target cells are detected by preferential binding of tase, two subunits of alkyl hydroperoxide reductase,
CylLL from the dual toxin complex, releasing CylLS to thiol peroxidase, and HypR itself through binding to a
induce the operon (71). “HypR box” (84, 85). HypR mutants are deficient in
The E. faecalis genome includes numerous other pu- macrophage survival and virulence.
tative regulatory proteins in a variety of other classes, Transcriptomic analyses have identified a number of
but few of these regulators have been characterized. The regulatory networks and putative regulatory proteins
E. faecalis version of the catabolite control protein gene, involved in the virulence of E. faecalis. These have been
ccpA, has been mutationally inactivated and shown to recently reviewed (10). Information relevant to stress re-
regulate a variety of glucose starvation proteins and was sponses in E. faecalis may also be found in a recent re-
capable of complementing a B. subtilis ccpA mutant (72). view on the stress physiology of lactic acid bacteria (86).
CcpA/P-Ser-Hpr binding to catabolite responsive ele- Posttranscriptional regulation by various RNA-
ments (cre) in E. faecalis has been shown to participate in mediated mechanisms has recently increased in promi-
(i) the repression of citrate utilization operons through nence in the regulation of gene expression in bacterial
transcriptional inhibition of both the structural genes cells. As in most other bacteria, the first examples of
and the gene for the GntR-type citrate-responsive posi- RNA-mediated regulation in enterococci were identified
tive regulator CitO (73), (ii) the partial repression of in MGEs (see below). More recently, a number of ex-
genes for malate utilization, including maeKR encoding amples of RNA-mediated regulation were identified on
a malate response TCSTS (74), and (iii) repression of the enterococcal chromosome. The best studied of these
the agmatine deiminase pathway in cooperation with the is regulation of the eut regulon, which encodes genes
mannose PTS and in opposition to the AguR-positive essential for ethanolamine (EA) utilization (87, 88) (Fig. 1).
regulator (75). CcpA has also been shown to be impor- Regulation of the eut regulon requires a response to
tant for the growth and virulence of E. faecium (76). two regulatory inputs: the presence of EA substrate and
Metal homeostasis is frequently important in bacterial the availability of adenosyl cobalamine (AdoCbl), a co-
pathogens, and regulatory responses to copper and man- factor in the first enzymatic step of EA catabolism. The
ganese have been investigated in the enterococci. Copper presence of EA is sensed by the EutVW TCSTS. Bind-
uptake in Enterococcus hirae is encoded in the four-gene ing of EA by the EutW HK leads to phosphotransfer
cop operon consisting of the copA and copB genes, to and dimerization of the EutV RR. Unlike most other
which encode proteins for copper uptake and efflux, re- RRs, EutV is an RNA binding protein in the ANTAR
spectively, copY, which encodes a copper-responsive re- family of antiterminator proteins (89). EutV∼P dimers
pressor, and copZ, which encodes a copper chaperone bind to and stabilize a dual hairpin structure in nas-
that delivers copper from CopA to CopY (77). At low cent RNA that precludes formation of multiple intrinsic
copper levels, CopY in complex with Zn(II) binds to terminators in four eut polycistronic RNAs, allowing
DNA sequences overlapping the promoter as a dimer and transcriptional read-through into downstream eut genes.
inhibits transcription. At high copper levels, CopZ de- AdoCbl is sensed via a unique riboswitch located up-
livers Cu(I) to CopY, displacing Zn(II) and causing dis- stream of the eutG structural gene, designated EutX. In
sociation from the promoter. CopZ complexed with Cu the absence of AdoCbl, EutX adopts a structure that
(I) is subject to proteolysis, preventing excessive accu- allows read-through to a EutV∼P dimer binding site.
mulation of intracellular copper, which can damage the The relative abundance of EutX allows sequestration of
cell. The Efa adhesin, encoded by the E. faecalis efaCBA EutV∼P dimers, preventing mRNA binding and anti-
operon, is regulated in response to manganese by the termination (+EA) (Fig. 1). AdoCbl binding to nascent
EfaR protein, a member of the DtsR/MntR repressor EutX causes a conformational change that allows for-
family (78). The EfaR binding motif was later identified mation of an intrinsic terminator that stops transcrip-
in the promoter regions of 30 genes. efaR mutation im- tion before the EutV∼P binding site (+AdoCbl) (Fig. 1),
paired biofilm formation, survival in macrophages, and allowing EutV∼P dimers to perform their antitermina-
oxidative stress tolerance, suggesting that EfaR is an tion function (+AdoCbl,+EA) (Fig. 1).
important modulator of virulence (79). Several other examples of posttranscriptional regula-
Four regulators, OxyR (80), PerR (81), Spx (82), and tion have been identified in the enterococcal core genome.
HypR (83), have been implicated in the response of E. The pyr operon of E. faecalis appears to be regulated
faecalis to oxidative stress. HypR is the best studied of by PyrR via an attenuation mechanism similar to that

ASMscience.org/MicrobiolSpectrum 5
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

FIGURE 1 Model for EutV- and EutX-mediated regulation of the eut regulon. In the absence
of either EA or AdoCbl (–EA), EutV∼P dimers are not available to disrupt terminators up-
stream of eut genes. In the presence of EA but the absence of AdoCbl (+EA), the EutX
riboswitch adopts a conformation that binds EutV∼P dimers, titrating them away from the
eut gene terminators. In the presence of AdoCbl but the absence of EA (+AdoCbl), EutX
adopts an alternative conformation that is not competent for binding EutV∼P dimers, but
such dimers are absent due to the lack of EA. In the presence of both EA and AdoCbl
(+AdoCbl, +EA), EutX is unable to bind EutV∼P dimers, which then interfere with termination
in eut transcripts, allowing the genes to be transcribed. (Adapted from reference 87.)

described for B. subtilis (90). Bioinformatic analysis sug- RNAs as transfer-messenger and 4.5S RNA. The specific
gests that genes involved in amino acid biosynthesis and functions and targets of these putative sRNAs and anti-
transport are subjected to T-box regulation in E. faecalis sense RNAs remains a mystery, although four have been
and E. faecium (91). A transcription attenuation mecha- implicated in modulating virulence and stress responses
nism for regulation of the tetM gene of Tn916 was pro- (98). E. faecium has also been subjected to experiments
posed to be based on the observation of (i) a short leader designed to detect putative regulatory RNAs with the
peptide encoded between the promoter and the TetM- identification of 61 sRNA candidates (99). Several of
encoding open reading frame, (ii) the existence of mul- these RNAs were demonstrated to be induced or re-
tiple alternative RNA secondary structures surrounding pressed during exposure to daptomycin or development
the leader peptide coding sequence, and (iii) the apparent of resistance to it. One putative sRNA, sRNA_0160, was
extension of a short transcript upon induction with tet- demonstrated to be downregulated in the presence of
racycline (92). RNase J2 has been implicated in the reg- daptomycin and repressed in a daptomycin-resistant
ulation of the ebp pilus operon in E. faecalis independent mutant. Further work will be required to determine the
of an effect on the level of the mRNA for EbpR, the target of this sRNA and its functional mechanism.
transcriptional activator of the operon (93). Mutation of
the E. faecalis rnjB gene encoding RNase J2 was further
shown to affect the expression of 62 genes and biofilm VANCOMYCIN RESISTANCE
formation, bile salt resistance, and virulence in a model Enterococci are intrinsically resistant to a variety of
system (94). commonly used antibiotics and have acquired resistance
Recently, a comprehensive map of all 5′ RNA ends to many others either by chromosomal mutation or ac-
in E. faecalis V583, discriminating processed from pri- quisition of MGEs (100). As such, the enterococci provide
mary ends, was obtained by a combination of differen- a collection and distribution center for the dissemination
tial tagging and RNA-seq (95). This study identified 85 of antibiotic resistance genes to other, potentially more
novel putative regulatory RNAs, which when added to pathogenic, Gram-positive bacteria. Dramatic evidence
those identified in previous studies (96, 97), brings the of this was provided by the characterization of the first
total number to >120. This includes putative trans-acting high-level vancomycin-resistant S. aureus (VRSA) iso-
sRNAs and antisense RNAs as well as such functional late (101, 102). Examination of the plasmid content of

6 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

the VRSA isolate and coisolated vancomycin-resistant of enzymes required to reprogram peptidoglycan syn-
E. faecalis (VRE) and vancomycin-sensitive S. aureus thesis. Nine genotypically and phenotypically distinct
(VSSA) strains suggested that a vanA-carrying transpo- systems designated VanA to VanE and VanG (reviewed
son, Tn1546, was delivered from the VRE to the VSSA in 107), VanL (108), VanM (109), and VanN (110) have
on a conjugative plasmid. The Tn1546 then transposed to been identified in VRE. Van gene clusters generally in-
a plasmid resident in the VSSA, and the donating plasmid clude three components: (i) a TCSTS, VanRS, required
was subsequently lost. Further studies have implicated for sensing the presence of the antibiotic and inducing
Inc18 plasmids, in particular, as the driving force for dis- the resistance operon; (ii) an essential three-gene core
semination of vancomycin resistance to S. aureus (103). encoding enzymes responsible for producing the D-Lac
However, even though the Tn1546-bearing plasmids are (VanH) or D-Ser (VanT) precursors, ligating those pre-
conjugative in S. aureus, vancomycin resistance has not cursors to D-Ala (VanA to VanN), and eliminating D-Ala-
become widespread in S. aureus infections. A total of only D-Ala dipeptides (VanX, VanY, and/or a VanXY fusion);
14 VRSA were isolated between 2002 and 2015 (104). and (iii) accessory genes performing various and some-
Whether this is due to some fundamental incompatibil- times unknown roles. While the core and accessory genes
ity between vancomycin resistance and pathogenesis in are generally transcribed as an operon, the TCSTS is
this species or some bottleneck in vanA transmission is transcribed from a separate promoter (Fig. 2).
unknown. The VanC, VanE, VanG, VanL, and VanN systems all
Vancomycin is a cell wall-active antibiotic that rep- encode low-level vancomycin resistance by producing pep-
resents the last line of defense against many multiply tidoglycan peptides terminating in D-Ala-D-Ser. All encode
resistant Gram-positive pathogens. Vancomycin and re- a VanXY bifunctional dipeptidase/carboxypeptidase for
lated glycopeptide antibiotics inhibit cell wall synthe- degrading D-Ala-D-Ala, a VanT serine racemase for pro-
sis by binding to the terminal D-Ala-D-Ala dipeptide of ducing D-Ser, and their respective ligases. The VanC sys-
peptidoglycan precursors, preventing their transfer from tems are responsible for the intrinsic resistance of motile
the lipid carrier to the cell wall by transglycosidases (105). enterococci, are chromosomally encoded, and do not ap-
Transpeptidases and D,D-carboxypeptidases involved in pear to be on mobile elements. Although VanC systems
cell wall synthesis are also inhibited. VRE circumvent appear to encode a TCSTS, resistance is either expressed
vancomycin action by substituting D-Ala-D-Lac or D-Ala- constitutively or only sluggishly induced (111, 112). The
D-Ser for the D-Ala-D-Ala dipeptide, decreasing the affinity VanE system is very similar to VanC in sequence and gene
of peptidoglycan precursors for glycopeptides. Hydrolysis organization but is present in an E. faecalis strain (113).
of pre-existing D-Ala-D-Ala dipeptides is also required for Although likely acquired from one of the intrinsically re-
full resistance (106). sistant enterococci, it is chromosomally located and could
Unlike resistance to most other antibiotics, resis- not be transferred to a vancomycin-susceptible strain.
tance to vancomycin involves the acquisition of a suite VanE is inducible in spite of a nonsense mutation in the

FIGURE 2 Organization of the VanA and VanB van-


comycin resistance systems. Gene functions are color
coded: green, sensory and regulatory; red, essential
resistance genes; blue, auxiliary resistance genes. De-
tailed functions of each gene are described in the text.
Green arrows show the position of the VanR inducible
promoters. The percentage identity of the VanB genes
to their VanA homologs is given below the individual
VanB genes. Homologs have identical names in both
systems except for the vanA and vanB ligases.

ASMscience.org/MicrobiolSpectrum 7
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

gene for the VanS HK, suggesting some level of cross-talk condition in vivo, with phosphorylation occurring either
with other cellular TCSTSs. The VanG system is also by acetyl phosphate or by cross-talk with other cellular
chromosomally located in an E. faecalis strain but resides HK. The primary role of VanS, then, is to dephosphor-
on a large (∼240 kb) mobile element that also encodes ylate VanR in the absence of the inducing signal (119,
an ermB erythromycin resistance gene (114). The VanG 120). The nature of the inducing signal is beginning to
system is inducible, probably due to the presence of a emerge. Given the differences in induction patterns and
functional VanRS TCSTS. VanL and VanN were re- the poor conservation of VanS, the signal may be dif-
cently described, with VanN being the first D-Ala-D-Ser- ferent for the VanA and VanB systems. Interestingly,
producing system in E. faecium (108, 110). single amino acid substitutions in the signaling domain
The VanA, VanB, VanD, and VanM systems produce of VanSB allow induction by teicoplanin (121). A re-
peptidoglycan precursors terminating in D-Ala-D-Lac. The cent report examining structural and activity effects on
VanA and VanB systems are widespread in both E. fae- VanSA of a range of potential ligands including vanco-
calis and E. faecium. In general, vancomycin resistance is mycin, teicoplanin, and a variety of peptidoglycan pre-
more prevalent in E. faecium than in E. faecalis (115). cursers identified the likely inducers as the glycopeptides
Phenotypically, VanA-type resistance is characterized by themselves (122).
high-level resistance to both vancomycin and teicoplanin, Both the VanA and VanB gene clusters are frequently,
while VanB-type resistance provides variable levels of re- if not always, associated with MGE. VanA is located on a
sistance to vancomycin only. Susceptibility to teicoplanin Tn3-type transposon, Tn1546, and is frequently found
in the VanB system is due to a failure to induce the re- on conjugative plasmids. In one E. faecium outbreak, a
sistance genes in response to this antibiotic; VanB cells unique mechanism of facilitating horizontal transfer of
induced with vancomycin are resistant to both antibi- VanA was observed (123). In this case, a nonconjugative
otics. Genotypically, the VanA and VanB systems are Tn1546-bearing plasmid was observed to integrate into
quite similar (Fig. 2). Each encodes a VanRS TCSTS, a a pheromone-responsive plasmid via a mechanism in-
VanH pyruvate dehydrogenase for production of D-Lac, volving a series of insertion sequences located outside of
their respective ligase VanA or VanB, and separate VanX Tn1546. Conjugative transfer was enhanced by the fact
dipeptidase and VanY carboxypeptidase enzymes. Only that plasmid fusion occurred within a homolog of prgX,
the vanH, vanA/B, and vanX genes are essential for re- the central negative regulator of the pheromone response
sistance. In addition, the VanA system encodes an addi- in pCF10-like plasmids (see below). The resulting in-
tional gene, vanZ, which confers low-level resistance to sertional inactivation of prgX derepressed conjugation
teicoplanin by an unknown mechanism, and the VanB functions, and transfer occurred in the absence of pher-
system encodes a gene, vanW (also present in VanG), of omone. Tn1546-like elements have also been identified
unknown function. on plasmids related to the conjugative non-pheromone-
The VanRS TCSTS of the VanA and VanB systems responsive plasmid pMG1 (124) (see below). VanB has
have been extensively investigated, and reference 116 been localized on a variety of MGEs. In some strains,
provides an excellent review. In vitro the VanRS sys- conjugative transfer of VanB has been associated with
tem functions much like other TCSTSs. The HK, VanS, the movement of large (90 to 250 kb), chromosomally
autophosphorylates its conserved histidine residue and located elements that may be complex conjugative trans-
then transfers the phosphate to the VanR RR (117). posons (125). VanB was further localized on one of these
Phosphorylated VanR (VanR∼P) produces a larger foot- elements to a 64-kb composite transposon, Tn1547, ca-
print and binds with higher affinity to its target pro- pable of transposition to plasmids under laboratory con-
moters than unphosphorylated VanR, consistent with ditions. In other cases, VanB resistance was associated
its in vivo role as a transcriptional activator (118). with conjugative plasmids (126), but the identification of
However, in the absence of VanS, dephosphorylation of similar genes on a variety of plasmids also suggested a
VanR∼P is unusually slow compared to related RR, and transposon location. A VanB gene cluster was localized
VanS stimulates dephosphorylation (117). In addition, on a Tn916-like element designated Tn5382 (127). Al-
VanR is capable of stimulating its own phosphoryla- though independent mobility of this element was not
tion in the absence of VanS, using acetyl phosphate. demonstrated, it appears to be associated with a larger,
These in vitro results are consistent with the observation 130- to 160-kb chromosomally located conjugative ele-
in vivo that inactivation of VanS results in constitutive ment. A similar element, designated Tn1549, was iden-
activation of vancomycin resistance rather than failure tified on a pAD1-like pheromone-responsive plasmid
to induce expression. Apparently, VanR∼P is the default (128). Mobility of chromosomally located vancomycin

8 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

resistance genes may also be accomplished by Hfr-like studied in bacteria other than the enterococci and have
transfer mechanisms as described below and in references been the subject of several excellent reviews (137, 139,
129 and 130. 140). Rep_3 replicons are ubiquitous in bacteria and
Unlike VanA and VanB, VanD systems provide con- include some of the best-studied bacterial plasmids, in-
stitutive vancomycin resistance due to various lesions in cluding F, P1, and pSC101 (141, 142). The RepA_N
the vanS gene (131–133). In spite of the presence of an plasmids are a class of replicons broadly distributed
apparently intact vanX gene, VanD systems produce very among the Firmicutes but whose individual members
little VanX D-Ala-D-Ala dipeptidase activity. Neverthe- have a narrow host range (143). They include both
less, peptidoglycan precursors in VanD strains contain conjugative and nonconjugative plasmids ranging in size
very little or no D-Ala-D-Ala terminal peptides due to from 3.3 to 281 kb. RepA_N proteins have also been
mutations in the chromosomally encoded ddl gene re- detected on streptococcal bacteriophages and integrated
sponsible for producing the D-Ala-D-Ala precursors (131, conjugative elements (ICEs). Of particular relevance to
133, 134). Thus, a combination of constitutive expres- enterococcal genetics are the RepA_N replicons that
sion of VanD and mutation of the chromosomal ddl encode conjugation systems that are induced by small
ensure vancomycin resistance. VanD systems have been peptide pheromones secreted by potential recipients, the
found only in E. faecium and do not appear to be mobile. pheromone-responsive plasmids. These plasmids repli-
The complex nature of the vancomycin resistance cate primarily in E. faecalis and remain one of only two
systems argues against their acquisition by accumulation plasmid systems in which a specific signal for induction
of mutations or piecemeal acquisition of individual genes. of conjugation has been identified (the quorum sensing
More than likely, they were acquired en masse from system of Agrobacterium Ti plasmids is the other [144]).
a single source. In this context it is interesting to note These plasmids will be discussed below, but the reader is
that the gene organization and sequence of the VanA referred to detailed reviews that discuss their replication,
and VanB systems are highly similar to self-protection stable inheritance, and conjugation (137, 145–150).
systems present in glycopeptide-producing species (135, Pheromone-responsive plasmids range in size from 37
136). to 92 kb and are maintained at a low copy number in
their native host. In general, the pheromone-responsive
Resistance to Other Antibiotics conjugation system works as follows. Conjugation in cells
In addition to vancomycin, enterococci express intrinsic containing pheromone-responsive plasmids is normally
and/or acquired resistance to a wide variety of other anti- repressed in the absence of an appropriate recipient.
biotics, including daptomycin, aminoglycosides, rifampi- When cocultivated with or exposed to culture filtrates
cin, quinolones, macrolide/lincosamide/streptogramin B, of plasmid-free cells, plasmid-containing cells respond by
β-lactams, and linezolid. For information on the genetics producing a plasmid-encoded surface adhesin called ag-
of these resistance mechanisms and the MGEs with which gregation substance (AS). AS binds to a ligand called
they are associated the reader is referred to a recent ex- enterococcal binding substance that is present on the
tensive review (100). surface of both donor and recipient cells, leading to the
formation of macroscopic cellular aggregates that pro-
vide the contact required for plasmid transfer. Once
transfer is complete, production of the pheromone spe-
PHEROMONE-RESPONSIVE PLASMIDS cific for the transferred plasmid is shut down in recipient
The enterococci host a multitude of plasmids, including cells by the combined action of two plasmid-encoded
both theta and rolling circle replicating replicons (RCRs) components: a peptide that competitively inhibits pher-
(137). The majority of these plasmids fall into six clas- omone activity and a membrane protein that reduces
ses based on conserved domains in their putative ini- pheromone secretion. These inhibitor mechanisms are
tiator proteins: three RCR classes (Rep_trans, Rep_2, specific for the cognate pheromone of the particular
and Rep_1) and three theta classes (Rep_3, inc18, and plasmid acquired; production of pheromone(s) specific
RepA_N). These plasmids have been further subdivided for other plasmids continues, allowing the recipient to
into 10 families (of 19 present in the Firmicutes) based induce a mating response in other potential donors and
on initiator sequence homology (138). RCR and inc18 continue to collect plasmids. A cartoon showing the
plasmids generally have a broad host range and are relevant players involved in the pheromone response,
widely disseminated in Gram-positive and even some how they interact, and the organization of the plasmid-
Gram-negative bacteria. They have been extensively encoded regulatory genes is shown in Fig. 3.

ASMscience.org/MicrobiolSpectrum 9
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

FIGURE 3 Regulatory circuitry of the pheromone response. (A) Events occurring at the cell
surface. Important chromosomally encoded determinants are colored yellow with the
exception of the pheromone itself, which is shown as blue circles. Plasmid-encoded de-
terminants are color coordinated with their genes shown in panel B. The inhibitor peptide
(ip) is shown as red circles and competitively inhibits pheromone binding to TraC. TraB
inhibition of pheromone secretion is depicted here as sequestration, but may also involve
peptide degradation. (B) Events occurring at the DNA level. Binding of pheromone by TraA
links events at the cell surface with events at the DNA level. A conformational change in
TraA due to pheromone binding is indicated by the change in shape of the molecule and
the change in dimerization state, although more subtle conformational changes are likely.
Pheromone-free TraA binds Po and inhibits transcription. Direction of transcription from
Po and Pa is indicated by the arrows. The antisense RNA, generalized to aR in this figure,
stimulates termination at t1, indicated by the green arrow. Positive regulatory elements vary
between different pheromone-responsive plasmids, and their mechanisms of inducing
downstream transcription of the conjugation structural genes may also vary. For simplicity,
the more common gene names and order are used. It should be noted that the gene order
of the traC and traB genes are reversed in pAD1. The RepA gene is shown to orient the
reader relative to Fig. 4. (C) Events at the RNA level. Relative levels of RNA produced from
the Po and Pa promoters under uninduced (red) and induced (green) conditions are
depicted by the sizes of the arrows.

Regulation of the Pheromone Signal production of pheromones specific for pAD1 (cAD1),
Transduction Circuit pCF10 (cCF10), and pAM373 (cAM373) was confirmed
All sex pheromones and inhibitors characterized to date by cloning and mutagenesis experiments (152–154). Mu-
are linear, hydrophobic peptides of seven or eight amino tations of the mature lipoprotein-encoding portion of the
acids. The genetic determinants responsible for the pro- genes for cAD1 and cAM373, cad and camE, respec-
duction of five pheromones were identified in the E. tively, had no effect on pheromone production or mat-
faecalis V583 genome and found to be encoded within ing potential, suggesting that the lipoproteins themselves
signal peptide coding sequences of different lipopro- are not involved in conjugation. Pheromone sequences
tein genes (151). The involvement of these genes in the are generally found at the C-terminus of the lipoprotein

10 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

signal peptide, indicating that signal peptidase II is re- Results with three TraA/PrgX proteins, those from pAD1,
quired for the initial processing step. Maturation of the pPD1, and pCF10, show that the repressors bind both the
pheromone is then completed by the site 2 protease Eep in stimulatory pheromone peptides and their cognate in-
most cases (155). As described above, Eep belongs to a hibitor peptides (166, 168, 169). In pPD1 and pCF10,
family of intramembrane processing proteins involved both inhibitor- and pheromone-bound repressors bind
in regulated intramembrane proteolysis in organisms the promoter required for expression of the conjugative
as diverse as bacteria and humans (156). The plasmid- apparatus (Po in Fig. 3) but with subtly different con-
encoded genes for the inhibitor peptides (iAD1, iCF10, figurations that make the difference between prevent-
iAM373, etc.) encode prepeptides resembling unattached ing or allowing RNA polymerase binding (168, 170).
lipoprotein signal peptides (157) and therefore require In pPD1 this involves the binding of TraA to alternate
only Eep for processing. Some variability is observed in sites within the promoter region (168). In pCF10 it in-
the processing of specific pheromones, suggesting further volves subtle conformational changes in the peptide-
complications and potential alternative pathways. For bound PrgX tetramers that allow greater competition
example, Eep-processed cCF10 is further processed by with RNA polymerase promoter binding in the inhibitor-
removal of three C-terminal amino acids by an unknown bound than in the pheromone-bound state (170). Thus,
mechanism (153, 158), and Eep is not required for cOB1, competition between pheromone and inhibitor peptides
cAM373, or iAM373 processing (155, 158). Interest- occurs at two levels, import via TraC/PrgZ and inter-
ingly, both cAD1 and cAM373 activities can be detected action with repressor protein TraA/PrgX. The complex
in some S. aureus strains. In both cases the responsible dual peptide regulation of TraA/PrgX systems is unique
peptides are produced from the signal sequences of lipo- among RRNPP family members, giving rise to several
proteins unrelated to those that produce the same pep- interesting evolutionary questions that have been ad-
tides in E. faecalis (154, 159). Linear synthetic peptides of dressed elsewhere (148).
the appropriate sequence function as effective inducing As shown in Fig. 3, even in the absence of pheromone,
signals for the cognate plasmids, indicating that no fur- transcription from Po is not completely repressed. In fact,
ther processing is necessary. Hydrophobic septa- and oc- basal-level expression is required to maintain the unin-
tapeptide pheromones are pumped out of the membrane duced state since the gene for the inhibitor peptide, ip in
by a membrane ABC pump (160). Release of lipoprotein Fig. 3, is under Po control. In fact, results in pCF10 sug-
signal sequence fragments appears common among the gest that pheromone induction results in only a 5-fold or
Firmicutes, and perhaps pheromone-responsive plasmids less increase in transcription from Po (147, 171). Thus,
evolved to take advantage of this process. It is interesting the pheromone response must rely on regulatory mech-
and perhaps surprising that so far, this efficient cell-cell anisms beyond simple derepression of transcription from
communication system is mainly found in the species E. Po. Multiple lines of evidence suggest that this added
faecalis and is not more widespread. layer of regulation occurs at the t1 terminator (Fig. 3),
Pheromones are imported into plasmid-containing cells which separates the Po promoter from the rest of the
by the chromosomally encoded oligopeptide permease conjugation genes. First, pPD1 TraA has been shown to
(Opp) system in combination with a plasmid-encoded bind to both t1 and t2 either alone or when iPD1-bound
OppA homolog, TraC/PrgZ, that provides increased sen- but not when cPD1-bound (168). It is proposed that this
sitivity and specificity for the cognate pheromone (161– facilitates termination in the uninduced state, but pre-
164). The chromosomal Opp system transports sufficient cisely how this occurs is not known. Second, transcrip-
pheromone to induce conjugation at high peptide con- tion from the Pa promoter results in the production of
centrations, but TraC/PrgZ allows responses to phero- an antisense regulatory RNA (aR in Fig. 3) that binds
mone levels of 10–10 to 10–11 M (161). Evidence indicates to the nascent Po transcript, stabilizing a conformation
that inhibitor peptides compete with their cognate pher- including an intrinsic terminator. mD and Qa antisense
omones for TraC/PrgZ binding and that this at least par- RNAs have been identified and described in pAD1 and
tially accounts for their function (but see below) (102, pCF10, respectively (172, 173), and their sequences and
165). location are highly conserved in other pheromone plas-
Once inside the cell, pheromone is bound by a key mids. Details of this countertranscript-driven attenuation
negative regulatory protein, TraA/PrgX (161, 166). These mechanism have been described for the pCF10 system
proteins belong to the RRNPP (Rap, Rgg, NprG, PlcR, (174, 175). In pCF10, PrgX and Qa are transcribed from
and PrgX) family of linear peptide pheromone-responsive Pa, and PrgX is required both for positive autoregulation
regulators that are widespread in Firmicutes (167). of Pa and proper processing of Qa (171). In both the pAD1

ASMscience.org/MicrobiolSpectrum 11
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

and pCF10 systems, pheromone induction results in a PrgY proteins appear to be part of an orthologous family
significant decrease in antisense RNA production (176– of proteins found in all domains of life but only spottily
178). Based on these observations, it was proposed that, distributed (188). For instance, there are representatives
under noninducing conditions, the level of antisense RNA in humans, plants, and Gram-negative bacteria but not in
is precisely poised to ensure that all Po transcripts adopt a any other Firmicutes species outside of the enterococci.
termination-competent structure. Even a moderate in- This phylogenetic distribution suggests that horizontal
crease in transcription from Po tips the balance in favor of gene transfer has been critical for their distribution. No
antitermination by three amplification mechanisms: (i) function has been assigned for any of the TraB/PrgY
titration of the Pa transcript by the excess Po transcript, orthologs, but homology suggests that they are involved
(ii) transcriptional interference favoring the Po transcript in some form of peptide signaling or processing. Inter-
and further reducing Pa transcript levels (179), and (iii) estingly, pAM373 appears to control extracellular pher-
RNase III-mediated degradation of the complex between omone levels without a clear TraB/PrgY homolog (189).
the Po and Pa transcripts, which reduces translation of Altogether then, pheromone plasmids encode three
TraA/PrgX (180). These overlapping regulatory mecha- proteins, TraC/PrgZ, TraA/PrgX, and TraB/PrgY, that
nisms allow the cell to precisely set the pheromone: bind their cognate pheromone yet are divergent enough
inhibitor ratio high enough to prevent fruitless induction to be able to selectively respond only to the pheromone
of the costly conjugation apparatus but low enough to specific for their encoding plasmid. Structural analyses
allow efficient transfer of the plasmid DNA when suffi- of these proteins suggest that the pheromone binding
cient numbers of recipients are present. Tight regulation pockets did not diverge from a single ancestral domain
at t1 is most likely required to prevent transcription of but rather evolved independently and convergently (148).
strong downstream positive regulators. In the case of Assuming that all pheromone plasmids descended from
pAD1, this regulator is TraE1, which positively auto- a single ancestor, divergence to respond to new phero-
regulates its own transcription and is essential for ex- mone and inhibitor peptides requires coordinated evolu-
pression of the downstream structural genes required for tion of the binding pockets of all three proteins. How
conjugation (177, 181). The pCF10 plasmid lacks a these specificity switches occur is an intriguing evolu-
TraE1 homolog but produces several RNAs in the re- tionary question.
gion immediately downstream of t1 and t2 that posi-
tively regulate downstream structural genes (182–184). The Conjugation Apparatus
The mechanisms of action of these regulators are un- Downstream of the positive regulatory elements of pAD1,
known but may involve both stimulation of transcrip- transcription of a contiguous region of ∼20 kb is induc-
tional read-through and translation. ible by pheromone (190). In most pheromone plasmids,
The inhibitor peptide is essential for modulating the the first two genes of this region encode a surface exclu-
plasmid-containing cell’s response to exogenous phero- sion protein and AS (191). Both proteins encode signal
mone, but it is not sufficient to prevent self-induction by sequences and LPXTG cell wall-anchoring motifs and
endogenously produced pheromone. A second plasmid- have been demonstrated to be present on the surface
encoded protein, TraB/PrgY, is required to specifically of only pheromone-induced plasmid-bearing cells (192).
reduce secretion of the cognate pheromone (185–187). The surface exclusion proteins limit conjugation between
The pCF10-encoded PrgY protein has been studied in strains harboring the same pheromone plasmid (193,
detail (158, 188). It consists of four transmembrane do- 194). AS is essential for the cellular aggregation response
mains with the N-terminal two-thirds of the protein lo- to pheromone and efficient mating in broth but is not
cated on the outer surface of the cytoplasmic membrane. required for plasmid transfer on solid surfaces, suggesting
The cCF10 pheromone binding region has been mapped that it is not an integral part of the mating pore (195). AS
to an approximately 120-amino acid region in the extra- is a cell wall-anchored protein and is highly conserved in
cytoplasmic domain adjacent to the first transmembrane all pheromone plasmids except pAM373. pAM373 does
segment. PrgY has been proposed to function by speci- not encode an entry exclusion function (196) and encodes
fically binding to cognate pheromone as it emerges from an AS that is half the size of other AS proteins and shows
the membrane, either following or concomitant with only localized homology (189). AS facilitates intercellu-
cleavage by Eep. The protein then either degrades the lar aggregation by binding to an enterococcal binding
peptide directly or chaperones it to a cell wall bound substance which is present on the surface of both donor
protease for degradation (147). Recent data favor direct and recipient cells. Lipoteichoic acid (LTA) appears to
degradation (G. Dunny, personal communication). TraB/ be at least one component of enterococcal binding sub-

12 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

stance, since free LTA inhibits aggregation (197) and cells inverted repeat (216). The relaxase of these plasmids is
containing mutations affecting LTA are poor recipients unique in that it lacks the “three-histidine motif” com-
(198). Mutagenesis and in vitro binding analysis of the mon to most other plasmid relaxases (217) and instead
AS from pCF10 has identified two distinct aggregation relies on a catalytic domain related to the PD-(D/E)XK
domains, one of which binds tightly to LTA in vitro (199, family of restriction endonucleases for nicking activ-
200). However, pCF10 AS also binds LTA from E. hirae ity. Thus, the pAD1 TraX relaxase is the prototype for
which does not aggregate with AS-expressing E. faecalis a distinct family of relaxases designated MOBC (218).
cells, suggesting that LTA binding may not be sufficient pAD1 also contains a second oriT located within the repA
for aggregation. AS has also been implicated as a viru- initiator protein gene that is similar to IncP-type oriTs
lence factor in several pathogenicity models (201–205) (209), but this oriT is utilized with 1,000-fold lower ef-
and is induced by plasma by means of the pheromone- ficiency than oriT2 and its biological relevance is un-
sensing machinery (206). A recent study demonstrated clear. The pCF10 relaxase, PcfG, is unrelated to pAD1
that the pCF10 surface exclusion protein, AS, and the TraX and instead is closely related to the pheromone-
product of the prgC gene immediately downstream all independent Lactococcus lactis plasmid pRS01 relaxase
play roles in conjugation, biofilm formation, and viru- (210, 219, 220). A comparative study of the pCF10 and
lence (207). Another gene located between the AS gene pRS01 systems identified related accessory proteins im-
and prgC, designated prgU, encodes a small protein with portant for assembly of the relaxosome. In spite of clear
an RNA binding fold that mitigates the toxicity associated sequence homology between the pCF10 and pRS01 re-
with overproduction of AS and other related adhesins in laxases and their target oriTs, each system effectively
multiple Gram-positive bacteria (208). Its mechanism of mobilized only its cognate target (221).
action remains to be determined.
Comparison of the sequences downstream of the AS Replication and Stable Inheritance
gene of five completely sequenced pheromone-responsive On the opposite side of the pheromone-response regu-
plasmids, pAD1 (209), pAM373 (189), pCF10 (210), latory module from the conjugation structural genes
pTEF1, and pTEF2 (6), show a high degree of conser- are the genes required for replication and stable inher-
vation in the presumed structural genes required for itance (189, 222–224). The genes of the basic replicon
conjugation. Comparison of these sequences to those of include a replication initiator protein RepA/PrgW, a
conjugative systems of both Gram-negative and Gram- partition system RepBC/PrgPO, and an antisense RNA-
positive bacteria suggests that they are orthologous to the regulated type I toxin-antitoxin (TA-1) module desig-
family of protein and DNA secretion machineries col- nated par (Fig. 4). The repA/prgW genes belong to the
lectively referred to as type 4 secretion systems, or T4SSs RepA_N family of replication initiator proteins, which
(211). Functional studies of the products of these genes along with their associated partition modules, have been
from pheromone-responsive plasmids are limited, but recently reviewed (143). RepA_N initiator proteins are
work on pCF10 has identified PcfC as the coupling pro- not restricted to pheromone plasmids and are wide-
tein responsible for bringing the nicked DNA substrate spread among conjugative and nonconjugative plasmids
to the secretion apparatus (212, 213), PrgJ as a VirB4- in the Firmicutes. The genes encoding RepA_N initiators
like ATPase in the T4SS apparatus (214), and PrgK as a encode a centrally located series of repeats whose or-
peptidoglycan hydrolase required to generate localized ganization and sequence vary between plasmids and, in
lesions in the cell wall for assembly of the T4SS (215). some cases, have been demonstrated to function as an
Within the conserved region encoding the T4SS is a origin of replication (oriV) (225–228). Work on pAD1
short segment of localized reduced homology. In pAD1, has shown that (i) oriV is sufficient to support plasmid
this region has been shown to encode a functional origin replication if its RepA_N initiator, RepA, is supplied
of transfer (oriT2) located between a relaxase gene, traX, in trans, (ii) the repA gene itself is sufficient to support
and traW, the pAD1 coupling protein homolog (209). A plasmid replication if RepA is overexpressed from an
similarly located oriT was identified in pAM373, and the artificial promoter, and (iii) RepA binds specifically to
two oriTs were found to be mobilized only by their oriV in double-stranded DNA and nonspecifically to
cognate plasmids. Plasmid-specific single-strand nicking single-stranded DNA (225). The pCF10 initiator pro-
by the putative relaxase was demonstrated within a large, tein, PrgW, binds cCF10 pheromone, and provision of
conserved inverted repeat in oriT. Specificity was im- cCF10 in a host not normally permissive for plasmid
parted by nonconserved direct repeats, presumed to be replication, L. lactis, allows plasmid establishment (229).
the relaxase binding site, adjacent to the conserved The role of cCF10 in plasmid replication and whether

ASMscience.org/MicrobiolSpectrum 13
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

FIGURE 4 Genetic organization of a general pheromone-responsive plasmid replicon.


Gene names are given as pAD1/pCF10. The stipled box within repA/prgW represents the
origin of replication (oriV) to which the repA/prgW product binds. The lined boxes at each
end of the repB/prgP-repC/prgO operon represent the likely centromere-like sites to
which the repC/prgO products bind and, with the repB/prgP product, direct plasmid
partition. The organization of the par region is blown up below the replicon. The arrow-
heads at each end of the locus marked P represent the promoters for the RNAI and RNAII
transcripts. The extent and direction of transcription of the transcripts are shown above and
below the genes for RNAII and RNAI, respectively. The fst gene is designated by the di-
agonally lined box, and the sequence of the peptide is shown at the bottom. DRa and DRb
are direct repeats in the DNA sequence that provide complementarity between RNAI and
RNAII when transcribed in opposite directions. Overlapping transcription at the bidirec-
tional transcriptional terminator also provides complementarity.

this observation can be extended to other pheromone- initiator protein is superimposed on handcuffing in the
responsive plasmids has not been determined. The mech- staphylococcal plasmids (232). No analogous antisense
anisms of strand opening, initiation of replication, and RNA has yet been detected, either physically or bioin-
regulation of initiation frequency have yet to be deter- formatically, on the pheromone plasmids.
mined for the pheromone-responsive plasmids. How- The pheromone-responsive plasmid partition systems
ever, work with the RepA_N proteins of staphylococcal are variants of the previously described type Ib class
plasmids pSK41 and pTZ2162 may be instructive (230). (233). Type I systems encode a well-conserved Walker-
Both domains flanking the central repeat region of these type ATPase designated ParA, a poorly conserved DNA
proteins adopt a structural fold similar to the DnaD pri- binding protein designated ParB, and a centromere-
mosome. However, the N-terminal domain has evolved like site to which ParB binds. In type Ib systems, the
to bind the oriV repeats bending the DNA and presum- ParA component lacks a DNA-binding domain, and the
ably facilitating separation of the DNA strands. The C- centromere-like site is located upstream of the parA-
terminal domain retains primosome function and recruits parB operon where binding of the ParB component can
DnaG primase to initiate replication. Furthermore, the autoregulate transcription as well as direct partition.
N-terminal domain was also demonstrated to be capable The RepB/PrgP and RepC/PrgO proteins correspond to
of bridging two plasmid origins, suggesting that it could type Ib ParA and ParB proteins, respectively. In all four
function in replication control by handcuffing (231). It pheromone-responsive plasmids examined, including
had been previously proposed that the pattern of con- pAD1, pCF10, pPD1, and pAM373, a series of repeats
servation of RepA_N proteins, with a highly conserved is present upstream of the repB/prgP gene (Fig. 4), with
N-terminal domain and genus-specific conservation of at least one repeat overlapping a putative promoter se-
the C-terminal domain, suggested a universally conserved quence, suggesting that protein binding to the repeats
function (e.g., origin binding) for the former and a genus- could suppress transcription. A series of similar repeats
specific function (e.g., interaction with host replication is also located downstream of the repC/prgO gene (143,
proteins) for the latter (143). These results support that 150). Each plasmid has repeats with a distinct DNA
prediction. A second method of copy number control in- sequence and organization. For example, pAD1 contains
volving an antisense RNA that regulates expression of the 26 TAGTARRR (R = purine) repeats upstream of repB

14 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

in two clusters of 13 and 12 tandem repeats with a single antitoxin. Although the RNAI and RNAII genes overlap
isolated repeat overlapping the promoter. Three more only at the terminator, each reads in opposite directions
tandem repeats are located downstream of repC. pCF10 across a pair of direct repeats (DRa and DRb in Fig. 4).
contains six tandem ATATATNNN (N = any nucleo- Therefore, RNAII contains regions of complementarity
tide) repeats upstream of prgP with a single isolated re- with both the 3′ and 5′ ends of RNAI. Furthermore, the
peat overlapping the promoter and two sets of four RNAI repeats are positioned such that interaction with
tandem repeats downstream of prgO. Work with the RNAII sequesters the translation initiation region of fst,
pAD1 RepBC system indicates that it functions similarly preventing ribosome binding and translation (244, 245).
to other partition systems (234). Thus, RepBC, with the Formation of the RNAI-RNAII complex proceeds in
flanking repeats, is capable of stabilizing heterologous at least two steps, with the interaction initiating at a
plasmids, the upstream repeats act as a strong incom- U-turn motif in the RNAI terminator loop which stim-
patibility determinant, RepC binds specifically and with ulates interaction with the complementary sequences in
high affinity to the repeats, RepB binds the repeats only the RNAII terminator loop. Interaction then extends to
in the presence of RepC and ATP, and RepC down- the repeats at the 5′ end, suppressing translation (246)
regulates its own expression. Plasmids bearing either the (Fig. 5). An intramolecular stem-loop within RNAI that
full complement of 25 upstream repeats or the half site partially sequesters the fst ribosome binding site is re-
containing 12 repeats but not the downstream 3 repeats quired to inhibit translation until RNAII binding can be
can be stabilized by repBC supplied in trans. Sponta- completed (244, 247). A long distance interaction be-
neous expansion of the pAD1 repB upstream repeats tween sequences near the 3′ end of RNAI and its 5′
has also been associated with loss of regulation of the nucleotides is responsible for the stability of RNAI rela-
conjugative response and constitutive aggregation (235). tive to RNAII (248). Unlike most antisense RNA sys-
The molecular basis of this phenomenon is unknown but tems, binding of RNAI by RNAII results in formation of
may not be directly related to the normal function of a highly stable complex rather than specifically targeting
the repeats. RepB proteins are broadly distributed in the the complex for degradation (248, 249). Formation of
Firmicutes but are not phylogenetically congruent with such a stable complex allows the toxin-encoding RNAI
the associated replication initiator proteins, suggesting to persist in plasmid-containing cells. Over time, how-
mosaicism of partition and replication modules (143). ever, RNAII is slowly removed from the complex to al-
TA modules are ubiquitous on bacterial plasmids low translation of fst. The mechanism of this preferential
and function as stability determinants by programming degradation of RNAII from the complex is unknown.
plasmid-free segregants for death (236). To accomplish The Fst toxin sequence is shown in Fig. 4. It consists of
this, TA modules encode a stable toxin and an unstable a central hydrophobic region flanked by charged N- and
antitoxin; as long as the plasmid is retained, antitoxin is C-terminal tails. Overexpression of Fst has been shown
continuously replenished and the cell is protected, but if to be toxic in E. faecalis, S. aureus, B. subtilis, and E. coli
the plasmid is lost, the antitoxin is degraded and the (247, 250–252). In all four species overexpression re-
toxin kills the cell. There are now five classes of TA sults in condensation and mislocalization of the nucleoid
modules (237), but only two are currently relevant to and interference with proper partition and cell division.
enterococci. In TA-1 the antitoxin is an unstable small Membrane permeabilization is only a secondary effect.
regulatory RNA that binds to the toxin message and Internal expression of Fst and external addition of the
suppresses its translation. In TA-2 the antitoxin is an lantibiotic nisin synergistically effect cell killing, suggest-
unstable protein that binds to the toxin protein and in- ing that the two peptides have different but comple-
hibits its activity. A general review of these systems is mentary effects on the cell envelope (252). Saturation
available (238), and enterococcal TA systems have been mutagenesis experiments revealed that the central hy-
reviewed (137). Only one TA system has been charac- drophobic region and charged residues in the N-terminus
terized on pheromone-responsive plasmids, the TA-1 par were important for overexpression-mediated toxicity but
locus (see 137, 239–241 for extensive reviews). par en- that the C-terminal charged tail was nonessential (251).
codes two convergently transcribed RNAs, RNAI and An atomic resolution structure determined in a mem-
RNAII, initiating from each end of par and terminat- brane mimetic by nuclear magnetic resonance spectros-
ing at a common bidirectional intrinsic terminator (242, copy revealed that the essential hydrophobic region
243) (Fig. 4). RNAI encodes the toxic component of the forms an α-helix that spans the membrane with the
system, a 33-amino-acid peptide designated Fst. RNAII charged N- and C-termini protruding (253). The charged
is a 65-nucleotide small regulatory RNA that is the par C-terminal seven amino acids were disordered and pre-

ASMscience.org/MicrobiolSpectrum 15
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

FIGURE 5 Model of RNAI-RNAII interaction. RNAI is the larger, black structure and RNAII is
the smaller, blue structure. Stems, loops, and bulges are depicted in their approximate
locations and sizes as determined by experimentation. The red stem-loop structure within
RNAI sequesters the ribosome binding site and prevents translation until a complex is
formed. The initial interaction occurs at a U-turn motif in the terminator loop of RNAI (A),
followed by interaction between the complementary repeats in the 5′ end of each RNA
(B and C). Since RNAII-mediated protection from the RNAI-encoded toxin occurs in vivo even
when one of the repeats is mutated, the structure shown in panel C is apparently sufficient
to prevent translation in vivo. Once complex formation is complete (D), the structure is
extremely stable in vivo and in vitro perhaps due to the gap between the interacting repeats.

dicted to extend from the cytoplasmic side of the mem- The Ldr members are present in the γ-proteobacteria and
brane. The authors suggested that the primary function are regulated in a manner more similar to hok-sok fam-
of membrane insertion was to facilitate interactions with ily members. In spite of their phylogenetic distance, Ldr
a specific target, rather than being directed against the overexpression has effects on nucleoid condensation and
membrane itself. They also predicted that the disordered cell division similar to those Fst has in E. coli (247, 257).
C-terminus might become structured upon recognition of Like most other TA systems, the par-like Fst-encoding
the target. This would appear to contradict results indi- systems have both MGE and chromosomal members,
cating that the C-terminal amino acids are nonessential and like other TA systems, the function of the chromo-
for toxicity, but it should be noted that the expression somal members is obscure. One of these chromosomal
system used in reference 251 could not distinguish levels members, the toxin of which is annotated as EF0409, is
of toxicity. Therefore, it is possible that the toxicity is conserved in E. faecalis genomes (251). For purposes of
substantially decreased in the truncated mutants and nomenclature, we have established a convention of using
could even be an artifact of overexpression. This possi- the locus or MGE name as a subscript for the relevant
bility is under further investigation. element. While the parEF0409 and parpAD1 systems show
TA-1 type toxins in general are small proteins, less clear genetic similarity, the predicted RNAI-RNAII in-
than 60 amino acids, that have hydrophobic regions ty- teraction sites are divergent, and it has been demon-
pical of transmembrane domains (254). These toxins strated that the antitoxins do not cross-protect (258).
have been divided into eight superfamilies, and Fst is ParEF0409 is located between two paralogous mannitol-
the founding member of the Fst/Ldr family (255). The type phosphotransferase sugar transport systems, sug-
Fst members of this family are widespread among the gesting that it may be involved in regulating carbon flux.
Firmicutes and appear to be regulated and organized Transcriptomic analysis of cells overexpressing either
similarly to the pAD1 par determinant (251, 255, 256). FstpAD1 or FstEF0409 revealed that effects on the expres-

16 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

sion of a variety of transporters were predominant (258, The E. faecalis transposon Tn916 was the first well-
259), including effects on several genes for PTS compo- characterized self-conjugative transposon (2, 3) although
nents other than those to which parEF0409 is genetically large, conjugative chromosomal elements had been pre-
linked. It was also observed that interference with the viously identified in other streptococci (265). Conjuga-
function of ABC transporters rescued cells from FstpAD1 tive transposons were later found to be widespread in
overexpression (259). Utilizing a novel expression vector, the lactic acid bacteria, and mechanistically similar but
pCIE, that allows tight regulation and a large dynamic unrelated elements were identified in Bacteroides spe-
range of induction, the effects of low-level expression of cies (266). Characterization of new elements from both
FstpAD1 and FstEF0409 were compared. It was found that, Gram-positive and Gram-negative organisms and the
while there was some overlap, overexpression of the two reinterpretation of data on older elements formerly clas-
toxins had distinct transcriptomic effects (258). This sug- sified as integrative plasmids have revealed that MGEs
gests that the toxins have different targets or have dis- with characteristics similar to conjugative transposons
tinct effects on the same target. Finally, overexpression of (i.e., that excise from their chromosomal locus, circular-
the Fst proteins is approximately 10-fold more toxic to ize, transfer to a new host cell by conjugation, and in-
mannitol-grown than to glucose-grown cells (K. Weaver, sert in the recipient cell’s chromosome without extensive
unpublished observation), strengthening the circumstan- replication) are probably as ubiquitous in bacteria as
tial link between par function and carbon flux. Somewhat plasmids. Since the mobility of most of these elements
surprisingly a mutant deleted for the parEF0409 antitoxin is dependent on a tyrosine or serine recombinase rather
was isolated in E. faecalis strain EryS (98). The mutant than a DDE-type transposase and since many of them
strain showed increased virulence and improved survival have relatively site-specific integration preferences, the
in macrophages under several stress conditions. Prote- appropriateness of the term “conjugative transposons”
omic analysis performed on this strain showed signifi- has been questioned and alternative names have been
cant differences from the wild type, but these differences suggested, the most inclusive of which is ICE, for inte-
did not correlate with the transcriptome differences ob- grative and conjugative element (267). The full spectrum
served in strain OG1RF (258). These differences could of ICEs has been described in detail in numerous reviews
be strain-related, due to differences between transcription (268–271). This section will focus primarily on Tn916 and
and translation effects, or due to the accumulation of related elements and other ICE-related issues specific to
compensatory mutations in the deletion strain. Further enterococci. A map of Tn916 is shown in Fig. 6. For more
investigation of the enterococcal par systems should elu- detail on the Tn916-family of ICEs see references 272 and
cidate the function of the chromosomal parEF0409 locus, 273. For a comprehensive list of Tn916-like elements and
determine the mechanism of action of the Fst toxins, and other ICEs in enterococci see reference 137.
provide insight into the evolution of the par-like systems. Unlike transposons using a DDE-type transposase,
Tn916-like transposons are not flanked by direct repeats
of target sequence. Preferred insertion sites tend to be AT-
CONJUGATIVE TRANSPOSONS (ICES) rich and possess intrinsic curvature, but the DNA se-
The enterococci carry the full range of transposon vari- quence is not conserved and flanking sequences on each
eties found in other bacteria. Two Tn3-type transposons, end are heterologous (274, 275). Unlike many other ICEs,
Tn917 encoding erythromycin resistance and Tn1546 Tn916 has relatively little insertion site preference. Two
encoding vancomycin resistance, have been extensively transposon-encoded proteins, Int and Xis, are required
studied (260, 261), and Tn917 and its derivatives have for excision (276, 277). Int is a tyrosine recombinase
been used for transposon mutagenesis in a number of (278) with N-terminal and C-terminal DNA binding do-
Gram-positive organisms. The genome sequence of E. mains (279). The N-terminal domain (Int-N) binds via a
faecalis strain V583 revealed the presence of 38 insertion three-stranded β sheet to direct repeat sequences near
sequence elements, with ISEf1, IS256, and IS1216 being each end of Tn916 (280, 281). The C-terminal domain
the most frequent (6). Both IS256 and IS1216 have been (Int-C) binds to the transposon termini as well as to target
associated with simple composite transposons (123, 262) sequences. Int-C contains the conserved active site resi-
and large, complex, mosaic elements (263, 264). Many dues involved in nicking-closing. Like other tyrosine re-
of these elements contain antibiotic resistance genes, and combinases, Int produces staggered cuts at each end of the
some are conjugative by means that have yet to be fully integrated transposon. One cut is made flush with the end
explained (see below). For a more detailed description of of the transposon, producing a recessed 3′ end covalently
these elements see references 137 and 150. jointed to Int via a phosphotyrosine linkage. The second

ASMscience.org/MicrobiolSpectrum 17
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

FIGURE 6 Genetic organization of Tn916 and the relative positions of Int and Xis binding
sites. (A) Functions of Tn916 genes are color coded: blue, recombination; green and red,
positive and negative regulation, respectively; yellow, tetracycline resistance; magenta, con-
jugation. Gene names and open reading frame numbers are shown above the individual
genes. The origin of transfer is designated by the line between orfs 20 and 21, labelled oriT.
Promoters and the direction of transcription are designated by labeled arrows below the gene
line. (B) Binding sites for the Int-C and Int-N DNA binding domains and Xis are designated by
marked circles and triangles. Int and Xis binding sites are color coordinated with the genes
shown in panel A.

cut is made 6 nucleotides into the host sequence, leaving has dual roles in excision, both stimulating and inhib-
a free 5′ OH that attacks the phosphotyrosine linkage at iting the process (292).
the other end of the transposon, resulting in circulariza- Once the circular intermediate is formed, the trans-
tion (282). Since the joint is derived from chromosomal poson is transferred to the recipient cell by conjugation.
DNA at each end that is not complementary, a 6-bp DNA strand transfer is initiated by nicking at oriT di-
heteroduplex is formed in the circular intermediate, called rected by the Orf20 relaxase and the Int protein (293,
the coupling sequence (283). The fact that Int must bind 294). While the Orf20 protein is responsible for strand
and cleave at the coupling sequences, which vary from nicking, Int functions as a specificity factor, conferring
insert to insert, may explain the wide range of conju- both strand and sequence specificity on its endonucleo-
gation frequencies (from <10–9 to >10–4 per donor cell) lytic activity (294). Although Tn916 oriT bears simi-
observed using different donor strains (284). How the larities to IncP-type origins of transfer (293), Orf20 does
coupling sequences affect conjugation frequency is not not show similarity to any of the identified families of
known, but it does not appear to be due to effects on plasmid-encoded relaxase proteins (217). Instead, it ap-
integrase affinity (285) or on excision frequency (286). pears to have similarity to the Rep_trans family of RCR
Xis binds adjacent to the Int-N binding sites on both initiator proteins and may represent a novel solution to
ends of the transposon (287) and stimulates Int-mediated the problem of conjugative nicking (217, 294). On the
excision of Tn916 in E. coli (276, 288), L. lactis (289), opposite side of the oriT from orf20 is orf21, which en-
B. subtilis (277), and in vitro (290). In E. coli, Xis bind- codes a product in the FtsK/SpoIIIE/coupling protein
ing to the left end is implicated in playing an architec- superfamily of DNA translocases. It is likely that Orf21
tural role similar to λ-Xis in excision of bacteriophage connects the oriT/Orf20 complex to the T4SS proteins
λ (291). HU protein plays a significant role in excision in encoded in orfs14 to 16 (137). A combination of genetic
E. coli, but no host factors affecting excision have been analyses and DNA sequence determination have identi-
identified in Gram-positive hosts. Interestingly, Xis bind- fied a number of genes involved in Tn916 conjugation
ing to the right end of Tn916 inhibits excision, perhaps (295, 296), but the precise roles of none of these genes
by competing with Int-N for binding, suggesting that Xis have been experimentally determined.

18 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

Tn916 does not mobilize plasmids into which it has is a tyrosine recombinase, while the recombinase protein
integrated, nor does it mobilize chromosomal genes ad- of Tn5397 is a serine recombinase. Conversely, the re-
jacent to its integration site during intercellular trans- combination module of the VanB transposon Tn1549 is
position. This suggests that some mechanism suppresses closely related to the Tn916 int/xis genes, but most of
utilization of the oriT site until after the transposon has the conjugation genes are unrelated (128). Putative ICEs
excised from its host genome and circularized. Support- identified in the E. faecalis V583 genome sequence, EfaC2
ing this suggestion, induced expression of the transfer and EfaD2, include conjugation-related genes apparently
genes requires a functional int gene, and transcription is acquired from pAD1 and pAM373 (267). A more global
initiated from the orf7 promoter, which since it is lo- analysis including plasmids, bacteriophages, and genomic
cated at the opposite end of the transposon, is linked to islands as well as ICEs indicates that the degree of ex-
the transfer genes only in the circular intermediate (297). change between these elements is very high, suggesting
Activation of the orf7 promoter requires orf7 and orf8 that, rather than representing distinct groups, they rep-
and is repressed by orf9, but the mechanisms of regu- resent a continuum of structures that maximize mobility
lation are unknown. Conjugation is also induced by the under various circumstances (304).
presence of tetracycline, which induces transcription of
the tetM gene via ribosomal override of a transcriptional
attenuator at its 5′ end (92). Transcription then proceeds OTHER FEATURES OF THE ENTEROCOCCAL
through tetM, increasing transcription of the positive MOBILOME
regulators orf7 and orf8 and blocking the orf9 regu- The above discussion focused on MGEs that are unique
lator presumably by antisense RNA interactions and/or to the enterococci and/or were originally described in
transcriptional interference. This in turn increases tran- the enterococci, but the enterococcal “mobilome” is ex-
scription through int and xis, stimulating excision and traordinarily diverse and includes plasmids, insertion
circularization, leading to read through into the structural sequences, and transposons in a variety of families, group
conjugation genes (272). Recent evidence suggests that II introns, integrons, pathogenicity islands (PAIs), and
subinhibitory concentrations of other ribosome-targeting bacteriophages. A recent review provides tabular lists
antibiotics also stimulate Tn916 conjugation and may of many of these elements (137). For a comprehensive
contribute to its spread (298). overview of MGEs associated with the mobilome of clin-
Once a copy of the transposon is transferred, integra- ically important E. faecium and E. faecalis lineages, the
tion into the recipient chromosome requires Int but not reader is referred to a recent report (305). Enterococcal
Xis (277). Since the known tyrosine recombinases func- phages have also been recently reviewed (306). Addressed
tion only on double-stranded DNA, replacement syn- below are some issues of particular interest related to the
thesis of the transferred strand presumably takes place enterococcal mobilome.
prior to integration. Experiments with int- versions of
Tn916 where Int was provided in trans from a plasmid in Important Non-Pheromone-Responding
the donor revealed that int gene expression was not re- Plasmids
quired in the recipient for integration, suggesting that the pAMα1 is an ∼10-kb multicopy tetracycline resistance
Int protein might be transported with the transposon plasmid that is a composite of two replicons (307, 308),
DNA (299). Recipients frequently acquire multiple cop- an RCR replicon similar to Bacillus cereus plasmid pBC16
ies of the transposon, and there is no evidence that Tn916 that carries the tetracycline resistance gene but does not
expresses either an entry exclusion function or transpo- function in E. faecalis and a putative theta-replicon simi-
son immunity (145, 300). Interestingly, in cells harboring lar to pS86 that supports replication in E. faecalis. Each
more than one copy of the transposon, excision of one component plasmid encodes a relaxase gene, and the
element transactivates excision and mobility of other junction sites contain oriT sequences recognized by them.
elements located elsewhere in the genome (301). The oriT/relaxase complex allows mobilization of the
Sequence analysis of known and putative ICEs in se- plasmid in the presence of another conjugative plasmid.
quenced genomes suggests that these mobile elements are The presence of the oriT sites at the plasmid junctions
modular and that the modules may have different evo- suggests that the fusion giving rise to pAMα1 occurred via
lutionary histories (267, 302, 303). For example, Tn5397 a site-specific recombination event catalyzed by the re-
from Clostridium difficile is greater than 84% identical at laxases as was previously observed in other plasmids
the DNA sequence level to Tn916 across all of the con- (309, 310). Under tetracycline selection, cells accumulate
jugation-related genes and the tetM gene, but Tn916 Int pAMα1 derivatives containing tandem repeats of the

ASMscience.org/MicrobiolSpectrum 19
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

pBC16 replicon, amplifying the tetracycline resistance native host. The Txe toxin belongs to the RelE super-
gene. Amplification is initiated by a RecA-independent family of RNA interferases and was shown to cleave
recombination event requiring at least one of the relaxase mRNA one base downstream of the AUG initiation co-
genes and both oriT sequences (311) followed by more don (319, 320). Interestingly, Axe is more closely related
extensive RecA-dependent amplification between redun- to Doc, the antitoxin for the Phd toxin, than it is to RelB,
dant sequences in the duplicated region (312). Thus, the the antitoxin most commonly associated with RelE tox-
relaxases of pBC16 and pS86 perform three functions: ins, supporting the hypothesis that TA-II systems have
(i) stimulate cointegration to form pAMα1, (ii) facilitate undergone extensive recombinatorial mixing and match-
mobilization of the cointegrate, and (iii) trigger amplifi- ing during their evolution (321). The ωεζ TA-II system is
cation of the tetracycline resistance gene in the presence of commonly present on the broad-host-range Inc18 plas-
the antibiotic. mids (139), many representatives of which are present in
pMG1 is an ∼65-kb conjugative plasmid originally enterococci (322), including the VanA-encoding E. fae-
isolated from E. faecium and encoding high-level gen- cium plasmid pVEF3 (323), pAMβ1 (308), and the pher-
tamicin resistance (313). pMG1-like plasmids bearing omone plasmid pSL1 (324). Unlike most TA-II systems,
Tn1546 encoding the vanA determinant have been as- the ε antitoxin does not autoregulate the locus. Instead,
sociated with the spread of vancomycin resistance in it is regulated by an independent regulatory component,
enterococci (124, 314). Like the pheromone-responsive ω, which also regulates the plasmid active partition lo-
plasmids, pMG1 is capable of high-frequency transfer cus and the copy control factor CopS (325). ζ-family
in broth. Unlike pheromone plasmids, pMG1 transfer is toxins have been proposed to cause cell growth arrest
not induced by exposure to culture filtrates from po- by depleting ATP and GTP pools in the cell and may
tential recipient cells, conjugating broth cultures do not be associated with persistence to antibiotics (326). The
form macroscopic cellular aggregates, and transfer occurs ubiquitous higBA and mazEF chromosomally encoded
freely between E. faecium and E. faecalis cells. Further- TA-II systems are prevalent in E. faecium and E. faecalis
more, no homology between pMG1 and the conjuga- clinical isolates, and mazEF has been suggested as a
tive systems of pheromone-responsive plasmids or Inc18 target for novel antibacterials (327).
plasmids was observed (313). Rather, sequencing results In E. faecalis V583 a mazEF TA-II homolog is closely
indicated significant homology of suspected conjugation linked to a TA-I locus distantly related to B. subtilis txpA-
genes to those from pXO2, the capsule-encoding plasmid ratA (328). Upstream of the mazEF locus is a direct repeat
of Bacillus anthracis (315, 316). Microscopic aggregates motif that binds MazE and MazEF, suggesting that the
were observed during mating, and this phenomenon is locus is negatively autoregulated as is observed in other
related to a region of the plasmid encoding five open TA-II modules (329). It was also observed that ectopic
reading frames. One open reading frame encodes a large expression of MazE or MazEF stimulated transcription
protein with similarity to a pXO2-encoded surface protein of the ratA antitoxin RNA for the txp-ratA locus, and a
that could represent an “aggregation substance” unrelated sequence with similarity to the direct repeat motif was
to the pheromone plasmid AS (316). Genes potentially found upstream of the ratA gene. Thus, the MazE toxin
encoding the coupling protein, a VirB11-family protein, a suppresses the toxicity of both mazEF, by reducing over-
relaxase, and the oriT were also identified. A gene desig- all transcription, and the txpA-ratA locus, by increasing
nated traA appears to encode a key positive regulatory synthesis of the ratA antitoxin, which then targets the
protein (317). txpA mRNA for degradation. Conversely, one would
expect triggered degradation of MazE, by an as yet un-
TA Systems known mechanism, to activate toxicity of both systems.
In addition to parpAD1 and parEF0409, several other TA Thus far, this is the only example of coordinated regula-
systems have been identified on enterococcal plasmids tion of TA-I and TA-II loci, and the complete system has
and chromosomes. Par homologs are present on sev- been dubbed a TAI-II locus.
eral other enterococcal plasmids, although no chromo-
somal loci analogous to parEF0409 have been described PAIs and Intercellular Transfer
outside of E. faecalis (215, 256). The E. faecium multi- by Hfr Formation
resistance plasmid pRUM (318) and the related van- PAIs are chromosomal regions containing large clusters of
comycin resistance plasmid pS177 (319) encode a TA-II genes associated with bacterial virulence (330). Frequently,
locus designated Axe-Txe. Axe-Txe stabilizes heterolo- PAIs include genes associated with known MGEs, such as
gous plasmids in Bacillus thuringiensis, E. coli, and its phage- and ICE-related integration/excision modules and

20 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

plasmid-related conjugation genes, and some have been onstrated in E. faecium strain C68. Transfer in this case
shown to be either independently mobile (331) or mo- was mediated by pLRM23 and a second plasmid and
bilizable by other MGEs (332). The first enterococcal PAI appeared to proceed via cointegration of the plasmid
was identified in E. faecalis strain MMH594, and related with the donor chromosome, excision of the plasmid
islands were described in strains V586 and V583 (333). with neighboring chromosomal DNA, circularization in
This PAI is ∼150 kb long, encodes several pathogenicity the donor prior to transfer, integration of the chromo-
factors, including the enterococcal surface protein (esp), somal segment into the recipient chromosome by homo-
cytolysin, AS, a bile acid hydrolase, various surface pro- logous recombination, and subsequent loss of the unstable
teins, and general stress proteins, and contains numer- plasmid remnant (129). Whether this mechanism can also
ous transposons. The PAI also contains phage-related facilitate PAI transfer has not been determined.
integration/excision proteins, homologs of plasmid con-
jugation functions, and is flanked by 10-bp terminal di- Enterococcal Bacteriophage and Genome
rect repeats, suggesting that it could be independently Defense Systems
mobile. Genomic analysis of clinical E. faecalis isolates Bacteriophages provide a sizable fraction of the mobilome
revealed that PAI genetic composition is highly variable, of both E. faecalis and E. faecium, amounting to close to
suggesting the involvement of modular accretion during 10% of the genome of E. faecalis strain V583 (6) and 3 to
its evolution and dissemination (334, 335). Intercellular 5% of seven sequenced E. faecium strains (340). V583
transfer of either portions of the PAI (336, 337) or the contains seven putative prophages, five of which have
complete PAI (130, 338) has been demonstrated be- sufficient gene content to generate functional virions. The
tween E. faecalis strains and between E. faecalis and E. phage02 region was subsequently determined to be part of
faecium. Complete transfer from V583 was shown in the core genome of E. faecalis and is likely no longer
vitro to be independent of PAI-encoded mobility genes capable of mobilization (335). The phage07 region is ca-
and to frequently include chromosomal genes outside of pable of producing infectious virions but requires phage01
the element. This in vitro transfer was dependent on the as a helper phage to provide packaging and structural
conjugation functions of pTEF1 and pTEF2, two pher- elements (341, 242). Sequencing of multiple E. faecalis
omone-responsive conjugative elements present in the genomes has confirmed the variable presence of these
same strain, suggesting that the plasmids mobilized the prophage elements except for phage02 (335, 343, 344),
PAI via chromosomal integration, perhaps at IS256 ele- and the OG1RF genome contains only phage02 (345).
ments, followed by mobilization either by an Hfr- or Comparative genomic analysis of E. faecalis phages
F′-like mechanism (130). In contrast, precise excision, suggests that they have a modular structure much like
circularization, and recipient insertion of the PAI was bacteriophages infecting other bacteria (306). The mod-
observed in E. faecalis strain UW3114, suggesting that ules consist of three transcriptional units, one encoding
the PAI is capable of independent mobility (338), per- the lysogeny control functions, one encoding genes re-
haps at a rate substantially lower than that mediated quired for the lytic pathway, and a variable “cargo” re-
by mobilization. Interestingly, the PAI circular interme- gion encoding potential lysogenic conversion genes, some
diate was not detected in V583, suggesting that an im- of which could encode important virulence factors. Such
portant component for self-mobilization may be absent cargo genes include a set of putative membrane pro-
in that strain or not expressed under the conditions tested. teins in the φEF11 phage (346) and a ferrochelatase gene
However, even in UW3114, a coresident pheromone- and cold shock protein gene in phage04 (6). However,
responsive plasmid, pLG2, was transferred in parallel no systematic analyses have been performed to unequiv-
with the PAI and may have facilitated its transfer, and the ocally link phage determinants to enterococcal patho-
authors did not determine whether or which PAI genes genicity (306). Laboratory studies have demonstrated a
might be required for mobility. So whether the E. faecalis capacity for transduction by enterococcal phages, in-
PAI is fully self-mobile is still an open question, although cluding interspecies transfer of antibiotic resistance, so the
genes associated with excision, integration, and transfer potential for phage effects on virulence does exist (347,
appear to be intact. 348).
Similar esp-associated PAIs have also been identified CRISPR-Cas function as adaptive immune systems for
in E. faecium that may be subject to conjugative transfer bacteria, generally targeting the DNA of invading MGEs
(337, 339, 340). Plasmid-mediated transfer of chromo- for degradation (see 349 for a recent review). Three dis-
somal determinants, including determinants involved tinct type II-A CRISPR loci have been identified in
in β-lactam and vancomycin resistance, has been dem- E. faecalis: CRISPR1 and 3 are complete functional loci

ASMscience.org/MicrobiolSpectrum 21
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

with associated Cas genes, while the CRISPR2 locus is ters could be shared among enterococci and with other
an “orphan” locus harboring repeats and spacers of the genera. Further studies identified the enterococci as a
same family as CRISPR1 but lacking Cas genes (8, 345). promiscuous hub for the dissemination of a wide variety
CRISPR1-Cas and CRISPR3-Cas are variably present in of antibiotic resistance genes. The accumulation of ge-
E. faecalis genomes and never cooccur in the same strain, nomic information on the enterococci has further revealed
while CRISPR2 is ubiquitous and can thus be considered just how important MGEs are to enterococcal patho-
part of the core genome of this species. CRISPR1-Cas- genesis and evolution, particularly in the way that the
related systems have also been detected in several envi- CRISPR MGE defense system impacts the structure of
ronmental and nonclinical enterococcal species (350). E. faecalis and E. faecium populations.
Spacer sequences from all three CRISPR loci show iden- Complicated regulatory schemes and important anti-
tities to phage and plasmid sequences, suggesting that all biotic resistance phenotypes are not limited to enterococ-
have acquired MGE segments for genome defense (8). cal MGEs, and progress is now being made on important
However, CRISPR2 only functions when the Cas genes systems in the core genomes, particularly those of of
of CRISPR1 are provided in trans (351), suggesting that E. faecalis and E. faecium. Work is proceeding on the
strains containing only CRISPR2 should be more sen- intrinsic mechanisms of antibiotic resistance in these or-
sitive to MGE invasion. Indeed, the absence of com- ganisms, and the complex transcriptional and posttran-
plete CRISPR-Cas loci is associated with larger genomes, scriptional mechanisms of regulation of the EA regulon
higher percentages of horizontally acquired genes, greater presage more interesting regulatory phenomena to be
antibiotic resistance, the presence of certain virulence fac- discovered. While roles for enterococcal TCSTSs are be-
tors, and high-risk multilocus sequence type (7, 8). There- ing defined, genomic analyses have revealed a plethora
fore, E. faecalis CRISPR-Cas loci, in cooperation with of potential sRNA regulators about which we know very
restriction-modification systems (351), are effective in lim- little. To this point, most detailed genetic analyses have
iting invasion by MGEs, but loss of CRISPR-Cas facilitates been performed in vitro or have focused on the effects
the evolution of pathogenicity by allowing greater hori- of the various systems during pathogenic processes, but
zontal gene transfer. Recently, a method for activating the enterococci are common intestinal bacteria, and it is
CRISPR2 and using it to modify the E. faecalis population likely that events occurring within the gut in the presence
structure was proposed (352). of the microbiota are more relevant to normal entero-
A type II-A CRISPR-Cas locus has also been identified coccal physiology. Here again, studies with MGEs are
in E. faecium, related to the E. faecalis CRISPR1-Cas likely to lead the way. Clearly, much remains to be dis-
locus (8). Like in E. faecalis, the locus is not present in covered about the genetics of these interesting and clin-
all E. faecium strains and is absent in most multiple ically important Gram-positive bacteria.
antibiotic-resistant strains and in the major hospital-
adapted clade of this species (353). REFERENCES
1. Dunny GM, Brown BL, Clewell DB. 1978. Induced cell aggregation
and mating in Streptococcus faecalis: evidence for a bacterial sex phero-
mone. Proc Natl Acad Sci U S A 75:3479–3483 http://dx.doi.org/10.1073
CONCLUDING REMARKS /pnas.75.7.3479.
2. Gawron-Burke C, Clewell DB. 1982. A transposon in Streptococcus
From the beginning, enterococcal genetics has been fo- faecalis with fertility properties. Nature 300:281–284 http://dx.doi.org/10
cused on MGEs. Pioneering studies of the enterococci .1038/300281a0.
gave us one of the first examples of intercellular com- 3. Franke AE, Clewell DB. 1981. Evidence for a chromosome-borne re-
munication in the bacterial world in the pheromone- sistance transposon (Tn916) in Streptococcus faecalis that is capable of
“conjugal” transfer in the absence of a conjugative plasmid. J Bacteriol
inducible conjugative plasmids and the first chromo- 145:494–502.
somally located conjugative elements in the conjugative 4. Uttley AH, Collins CH, Naidoo J, George RC. 1988. Vancomycin-
transposons, later to be named ICEs after their distribu- resistant enterococci. Lancet 1:57–58 http://dx.doi.org/10.1016/S0140
tion was found to be ubiquitous in bacterial cells. Stud- -6736(88)91037-9.
ies of the complex regulatory circuits controlling the 5. Leclercq R, Derlot E, Duval J, Courvalin P. 1988. Plasmid-mediated
resistance to vancomycin and teicoplanin in Enterococcus faecium. N Engl
pheromone response and the conjugative mechanisms of J Med 319:157–161 http://dx.doi.org/10.1056/NEJM198807213190307.
ICEs continue to inform those interested in the molec- 6. Paulsen IT, Banerjei L, Myers GSA, Nelson KE, Seshadri R, Read TD,
ular mechanisms of gene regulation in prokaryotes in Fouts DE, Eisen JA, Gill SR, Heidelberg JF, Tettelin H, Dodson RJ,
Umayam L, Brinkac L, Beanan M, Daugherty S, DeBoy RT, Durkin S,
general. The importance of MGEs in enterococci became
Kolonay J, Madupu R, Nelson W, Vamathevan J, Tran B, Upton J, Hansen
even more apparent with the emergence of vancomycin T, Shetty J, Khouri H, Utterback T, Radune D, Ketchum KA, Dougherty
resistance and the multitude of ways the van gene clus- BA, Fraser CM. 2003. Role of mobile DNA in the evolution of vancomycin-

22 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

resistant Enterococcus faecalis. Science 299:2071–2074 http://dx.doi.org 23. Fleurie A, Lesterlin C, Manuse S, Zhao C, Cluzel C, Lavergne J-P,
/10.1126/science.1080613. Franz-Wachtel M, Macek B, Combet C, Kuru E, VanNieuwenhze MS,
7. Lindenstrauss AG, Pavlovic M, Bringmann A, Behr J, Ehrmann MA, Brun YV, Sherratt D, Grangeasse C. 2014. MapZ marks the division sites
Vogel RF. 2011. Comparison of genotypic and phenotypic cluster anal- and positions FtsZ rings in Streptococcus pneumoniae. Nature 516:259–
yses of virulence determinants and possible role of CRISPR elements to- 262 http://dx.doi.org/10.1038/nature13966.
wards their incidence in Enterococcus faecalis and Enterococcus faecium. 24. Jacq M, Adam V, Bourgeois D, Moriscot C, Di Guilmi A-M, Vernet
Syst Appl Microbiol 34:553–560 http://dx.doi.org/10.1016/j.syapm.2011 T, Morlot C. 2015. Remodeling of the Z-ring nanostructure during the
.05.002. Streptococcus pneumoniae cell cycle revealed by photoactivated locali-
8. Palmer KL, Gilmore MS. 2010. Multidrug-resistant enterococci lack zation microscopy. MBio 6:e01108-15 http://dx.doi.org/10.1128/mBio
CRISPR-cas. MBio 1:e00227-10 http://dx.doi.org/10.1128/mBio.00227-10. .01108-15.
9. Palmer KL, van Schaik W, Willems RJL, Gilmore MS. 2014. Entero- 25. Land AD, Tsui H-CT, Kocaoglu O, Vella SA, Shaw SL, Keen SK,
coccal genomics. In Gilmore MS, Clewell DB, Ike Y, Shankar N (ed), Sham L-T, Carlson EE, Winkler ME. 2013. Requirement of essential
Enterococci: from Commensals to Leading Causes of Drug Resistant In- Pbp2x and GpsB for septal ring closure in Streptococcus pneumoniae D39.
fection. Massachusetts Eye and Ear Infirmary, Boston, MA. https://www Mol Microbiol 90:939–955 http://dx.doi.org/10.1111/mmi.12408.
.ncbi.nlm.nih.gov/books/NBK190425/. 26. Adams DW, Wu LJ, Errington J. 2014. Cell cycle regulation by the
10. DebRoy S, Gao P, Garsin DA, Harvey BR, Kos V, Nes IF, Solheim M. bacterial nucleoid. Curr Opin Microbiol 22:94–101 http://dx.doi.org/10
2014. Transcriptional and post transcriptional control of enterococcal .1016/j.mib.2014.09.020.
gene regulation. In Gilmore MS, Clewell DB, Ike Y, Shankar N (ed), 27. Rowlett VW, Margolin W. 2015. The Min system and other nucleoid-
Enterococci: from Commensals to Leading Causes of Drug Resistant In- independent regulators of Z ring positioning. Front Microbiol 6:478
fection. Massachusetts Eye and Ear Infirmary, Boston, MA. https://www http://dx.doi.org/10.3389/fmicb.2015.00478.
.ncbi.nlm.nih.gov/books/NBK190422/. 28. Fadda D, Santona A, D’Ulisse V, Ghelardini P, Ennas MG, Whalen
11. Jameson KH, Wilkinson AJ. 2017. Control of initiation of DNA MB, Massidda O. 2007. Streptococcus pneumoniae DivIVA: localiza-
replication in Bacillus subtilis and Escherichia coli. Genes (Basel) 8:E22 tion and interactions in a MinCD-free context. J Bacteriol 189:1288–1298
http://dx.doi.org/10.3390/genes8010022. http://dx.doi.org/10.1128/JB.01168-06.
12. Le Chatelier E, Bécherel OJ, d’Alençon E, Canceill D, Ehrlich SD, 29. Pinho MG, Kjos M, Veening J-W. 2013. How to get (a)round: mech-
Fuchs RPP, Jannière L. 2004. Involvement of DnaE, the second replicative anisms controlling growth and division of coccoid bacteria. Nat Rev Mi-
DNA polymerase from Bacillus subtilis, in DNA mutagenesis. J Biol Chem crobiol 11:601–614 http://dx.doi.org/10.1038/nrmicro3088.
279:1757–1767 http://dx.doi.org/10.1074/jbc.M310719200. 30. Garcia PS, Simorre J-P, Brochier-Armanet C, Grangeasse C. 2016. Cell
13. Bruck I, Goodman MF, O’Donnell M. 2003. The essential C family division of Streptococcus pneumoniae: think positive! Curr Opin Micro-
DnaE polymerase is error-prone and efficient at lesion bypass. J Biol Chem biol 34:18–23 http://dx.doi.org/10.1016/j.mib.2016.07.014.
278:44361–44368 http://dx.doi.org/10.1074/jbc.M308307200. 31. van Raaphorst R, Kjos M, Veening J-W. 2017. Chromosome segre-
14. Foster KA, Barnes MH, Stephenson RO, Butler MM, Skow DJ, gation drives division site selection in Streptococcus pneumoniae. Proc
LaMarr WA, Brown NC. 2003. DNA polymerase III of Enterococcus Natl Acad Sci U S A 114:E5959–E5968 http://dx.doi.org/10.1073/pnas
faecalis: expression and characterization of recombinant enzymes encoded .1620608114.
by the polC and dnaE genes. Protein Expr Purif 27:90–97 http://dx.doi
32. Kjos M, Veening J-W. 2014. Tracking of chromosome dynamics in
.org/10.1016/S1046-5928(02)00577-6.
live Streptococcus pneumoniae reveals that transcription promotes chro-
15. Ohmori H, Friedberg EC, Fuchs RP, Goodman MF, Hanaoka F, mosome segregation. Mol Microbiol 91:1088–1105 http://dx.doi.org/10
Hinkle D, Kunkel TA, Lawrence CW, Livneh Z, Nohmi T, Prakash .1111/mmi.12517.
L, Prakash S, Todo T, Walker GC, Wang Z, Woodgate R. 2001. The Y-
33. Minnen A, Attaiech L, Thon M, Gruber S, Veening J-W. 2011. SMC
family of DNA polymerases. Mol Cell 8:7–8 http://dx.doi.org/10.1016
is recruited to oriC by ParB and promotes chromosome segregation in
/S1097-2765(01)00278-7.
Streptococcus pneumoniae. Mol Microbiol 81:676–688 http://dx.doi.org
16. Permina EA, Mironov AA, Gelfand MS. 2002. Damage-repair error- /10.1111/j.1365-2958.2011.07722.x.
prone polymerases of eubacteria: association with mobile genome elements.
34. Kunst F, Ogasawara N, Moszer I, Albertini AM, Alloni G, Azevedo V,
Gene 293:133–140 http://dx.doi.org/10.1016/S0378-1119(02)00701-1.
Bertero MG, Bessières P, Bolotin A, Borchert S, Borriss R, Boursier L, Brans
17. Ozawa Y, Tanimoto K, Fujimoto S, Tomita H, Ike Y. 1997. Cloning A, Braun M, Brignell SC, Bron S, Brouillet S, Bruschi CV, Caldwell B,
and genetic analysis of the UV resistance determinant (uvr) encoded on the Capuano V, Carter NM, Choi SK, Cordani JJ, Connerton IF, Cummings NJ,
Enterococcus faecalis pheromone-responsive conjugative plasmid pAD1. Daniel RA, Denziot F, Devine KM, Düsterhöft A, Ehrlich SD, Emmerson
J Bacteriol 179:7468–7475 http://dx.doi.org/10.1128/jb.179.23.7468-7475 PT, Entian KD, Errington J, Fabret C, Ferrari E, Foulger D, Fritz C, Fujita
.1997. M, Fujita Y, Fuma S, Galizzi A, Galleron N, Ghim SY, Glaser P, Goffeau
18. Yagi Y, Clewell DB. 1980. Recombination-deficient mutant of Strep- A, Golightly EJ, Grandi G, Guiseppi G, Guy BJ, Haga K, et al. 1997. The
tococcus faecalis. J Bacteriol 143:966–970. complete genome sequence of the Gram-positive bacterium Bacillus subtilis.
19. Weaver KE, Reddy SG. 2006. The recombination deficient Entero- Nature 390:249–256 http://dx.doi.org/10.1038/36786.
coccus faecalis UV202 strain is a recA mutant. Plasmid 55:164–168 http:// 35. Buck M, Gallegos M-T, Studholme DJ, Guo Y, Gralla JD. 2000. The
dx.doi.org/10.1016/j.plasmid.2005.10.001. bacterial enhancer-dependent sigma(54) (sigma(N)) transcription factor.
20. Ip SCY, Bregu M, Barre F-X, Sherratt DJ. 2003. Decatenation of J Bacteriol 182:4129–4136 http://dx.doi.org/10.1128/JB.182.15.4129-4136
DNA circles by FtsK-dependent Xer site-specific recombination. EMBO J .2000.
22:6399–6407 http://dx.doi.org/10.1093/emboj/cdg589. 36. Héchard Y, Pelletier C, Cenatiempo Y, Frère J. 2001. Analysis of
21. Espeli O, Lee C, Marians KJ. 2003. A physical and functional inter- σ(54)-dependent genes in Enterococcus faecalis: a mannose PTS permease
action between Escherichia coli FtsK and topoisomerase IV. J Biol Chem (EII(Man)) is involved in sensitivity to a bacteriocin, mesentericin Y105.
278:44639–44644 http://dx.doi.org/10.1074/jbc.M308926200. Microbiology 147:1575–1580 http://dx.doi.org/10.1099/00221287-147-6
22. Pucci MJ, Thanassi JA, Discotto LF, Kessler RE, Dougherty TJ. 1997. -1575.
Identification and characterization of cell wall-cell division gene clusters in 37. Débarbouillé M, Martin-Verstraete I, Klier A, Rapoport G. 1991. The
pathogenic gram-positive cocci. J Bacteriol 179:5632–5635 http://dx.doi transcriptional regulator LevR of Bacillus subtilis has domains homolo-
.org/10.1128/jb.179.17.5632-5635.1997. gous to both sigma 54- and phosphotransferase system-dependent regu-

ASMscience.org/MicrobiolSpectrum 23
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

lators. Proc Natl Acad Sci U S A 88:2212–2216 http://dx.doi.org/10.1073 54. Podbielski A, Kreikemeyer B. 2004. Cell density-dependent regulation:
/pnas.88.6.2212. basic principles and effects on the virulence of Gram-positive cocci. Int J
38. Dalet K, Briand C, Cenatiempo Y, Héchard Y. 2000. The rpoN gene Infect Dis 8:81–95 http://dx.doi.org/10.1016/j.ijid.2003.04.003.
of Enterococcus faecalis directs sensitivity to subclass IIa bacteriocins. 55. Comenge Y, Quintiliani R Jr, Li L, Dubost L, Brouard J-P, Hugonnet
Curr Microbiol 41:441–443 http://dx.doi.org/10.1007/s002840010164. J-E, Arthur M. 2003. The CroRS two-component regulatory system is
39. Iyer VS, Hancock LE. 2012. Deletion of σ(54) (rpoN) alters the rate of required for intrinsic β-lactam resistance in Enterococcus faecalis. J Bac-
autolysis and biofilm formation in Enterococcus faecalis. J Bacteriol 194: teriol 185:7184–7192 http://dx.doi.org/10.1128/JB.185.24.7184-7192
368–375 http://dx.doi.org/10.1128/JB.06046-11. .2003.
40. Ho TD, Ellermeier CD. 2012. Extra cytoplasmic function σ factor 56. Kellogg SL, Kristich CJ. 2016. Functional dissection of the CroRS two-
activation. Curr Opin Microbiol 15:182–188 http://dx.doi.org/10.1016 component system required for resistance to cell wall stressors in En-
/j.mib.2012.01.001. terococcus faecalis. J Bacteriol 198:1326–1336 http://dx.doi.org/10.1128
/JB.00995-15.
41. Missiakas D, Raina S. 1998. The extracytoplasmic function sigma
factors: role and regulation. Mol Microbiol 28:1059–1066 http://dx.doi 57. Kellogg SL, Little JL, Hoff JS, Kristich CJ. 2017. Requirement of the
.org/10.1046/j.1365-2958.1998.00865.x. CroRS two-component system for resistance to cell wall-targeting anti-
microbials in Enterococcus faecium. Antimicrob Agents Chemother 61:
42. Benachour A, Muller C, Dabrowski-Coton M, Le Breton Y, Giard
e02461-16 http://dx.doi.org/10.1128/AAC.02461-16.
J-C, Rincé A, Auffray Y, Hartke A. 2005. The Enterococcus faecalis sigV
protein is an extracytoplasmic function sigma factor contributing to sur- 58. Hall CL, Lytle BL, Jensen D, Hoff JS, Peterson FC, Volkman BF,
vival following heat, acid, and ethanol treatments. J Bacteriol 187:1022– Kristich CJ. 2017. Structure and dimerization of IreB, a negative regulator
1035 http://dx.doi.org/10.1128/JB.187.3.1022-1035.2005. of cephalosporin resistance in Enterococcus faecalis. J Mol Biol 429:
43. Le Jeune A, Torelli R, Sanguinetti M, Giard J-C, Hartke A, Auffray Y, 2324–2336 http://dx.doi.org/10.1016/j.jmb.2017.05.019.
Benachour A. 2010. The extracytoplasmic function sigma factor SigV 59. Hall CL, Tschannen M, Worthey EA, Kristich CJ. 2013. IreB, a Ser/
plays a key role in the original model of lysozyme resistance and virulence Thr kinase substrate, influences antimicrobial resistance in Enterococcus
of Enterococcus faecalis. PLoS One 5:e9658 http://dx.doi.org/10.1371 faecalis. Antimicrob Agents Chemother 57:6179–6186 http://dx.doi.org
/journal.pone.0009658. /10.1128/AAC.01472-13.
44. Varahan S, Iyer VS, Moore WT, Hancock LE. 2013. Eep confers ly- 60. Kristich CJ, Little JL, Hall CL, Hoff JS. 2011. Reciprocal regulation
sozyme resistance to Enterococcus faecalis via the activation of the extra- of cephalosporin resistance in Enterococcus faecalis. MBio 2:e00199-11
cytoplasmic function sigma factor SigV. J Bacteriol 195:3125–3134 http:// http://dx.doi.org/10.1128/mBio.00199-11.
dx.doi.org/10.1128/JB.00291-13. 61. Labbe BD, Kristich CJ. 2017. Growth- and stress-induced PASTA
45. Hoch JA, Silhavy TJ. 1995. Two-Component Signal Transduction. kinase phosphorylation in Enterococcus faecalis. J Bacteriol 199:e00363-
ASM Press, Washington, DC. http://dx.doi.org/10.1128/9781555818319 17 http://dx.doi.org/10.1128/JB.00363-17.
46. Hancock L, Perego M. 2002. Two-component signal transduction in 62. Churchward G. 2007. The two faces of Janus: virulence gene regu-
Enterococcus faecalis. J Bacteriol 184:5819–5825 http://dx.doi.org/10.1128 lation by CovR/S in group A streptococci. Mol Microbiol 64:34–41 http://
/JB.184.21.5819-5825.2002. dx.doi.org/10.1111/j.1365-2958.2007.05649.x.
47. Engelbert M, Mylonakis E, Ausubel FM, Calderwood SB, Gilmore MS. 63. Teng F, Wang L, Singh KV, Murray BE, Weinstock GM. 2002.
2004. Contribution of gelatinase, serine protease, and fsr to the pathogenesis Involvement of PhoP-PhoS homologs in Enterococcus faecalis virulence.
of Enterococcus faecalis endophthalmitis. Infect Immun 72:3628–3633 Infect Immun 70:1991–1996 http://dx.doi.org/10.1128/IAI.70.4.1991
http://dx.doi.org/10.1128/IAI.72.6.3628-3633.2004. -1996.2002.
48. Qin X, Singh KV, Weinstock GM, Murray BE. 2000. Effects of 64. Roh JH, Singh KV, La Rosa SL, Cohen ALV, Murray BE. 2015. The
Enterococcus faecalis fsr genes on production of gelatinase and a serine two-component system GrvRS (EtaRS) regulates ace expression in En-
protease and virulence. Infect Immun 68:2579–2586 http://dx.doi.org/10 terococcus faecalis OG1RF. Infect Immun 83:389–395 http://dx.doi.org
.1128/IAI.68.5.2579-2586.2000. /10.1128/IAI.02587-14.
49. Qin X, Singh KV, Weinstock GM, Murray BE. 2001. Characterization 65. Baureder M, Hederstedt L. 2012. Genes important for catalase activity
of fsr, a regulator controlling expression of gelatinase and serine protease in Enterococcus faecalis. PLoS One 7:e36725 http://dx.doi.org/10.1371
in Enterococcus faecalis OG1RF. J Bacteriol 183:3372–3382 http://dx /journal.pone.0036725.
.doi.org/10.1128/JB.183.11.3372-3382.2001. 66. Le Breton Y, Boël G, Benachour A, Prévost H, Auffray Y, Rincé A.
50. Thomas VC, Thurlow LR, Boyle D, Hancock LE. 2008. Regulation of 2003. Molecular characterization of Enterococcus faecalis two-component
autolysis-dependent extracellular DNA release by Enterococcus faecalis signal transduction pathways related to environmental stresses. Environ
extracellular proteases influences biofilm development. J Bacteriol 190: Microbiol 5:329–337 http://dx.doi.org/10.1046/j.1462-2920.2003.00405.x.
5690–5698 http://dx.doi.org/10.1128/JB.00314-08. 67. O’Keeffe T, Hill C, Ross RP. 1999. Characterization and heterologous
51. Thurlow LR, Thomas VC, Narayanan S, Olson S, Fleming SD, Hancock expression of the genes encoding enterocin a production, immunity, and
LE. 2010. Gelatinase contributes to the pathogenesis of endocarditis caused regulation in Enterococcus faecium DPC1146. Appl Environ Microbiol
by Enterococcus faecalis. Infect Immun 78:4936–4943 http://dx.doi.org/10 65:1506–1515.
.1128/IAI.01118-09. 68. Guzmán Prieto AM, Wijngaarden J, Braat JC, Rogers MRC, Majoor
52. Nakayama J, Cao Y, Horii T, Sakuda S, Akkermans ADL, de Vos E, Brouwer EC, Zhang X, Bayjanov JR, Bonten MJM, Willems RJL, van
WM, Nagasawa H. 2001. Gelatinase biosynthesis-activating pheromone: Schaik W. 2017. The two-component system ChtRS contributes to chlor-
a peptide lactone that mediates a quorum sensing in Enterococcus faecalis. hexidine tolerance in Enterococcus faecium. Antimicrob Agents Chemo-
Mol Microbiol 41:145–154 http://dx.doi.org/10.1046/j.1365-2958.2001 ther 61:e02122-16 http://dx.doi.org/10.1128/AAC.02122-16.
.02486.x. 69. Haas W, Shepard BD, Gilmore MS. 2002. Two-component regulator
53. Nakayama J, Chen S, Oyama N, Nishiguchi K, Azab EA, Tanaka E, of Enterococcus faecalis cytolysin responds to quorum-sensing autoinduc-
Kariyama R, Sonomoto K. 2006. Revised model for Enterococcus faecalis tion. Nature 415:84–87 http://dx.doi.org/10.1038/415084a.
fsr quorum-sensing system: the small open reading frame fsrD encodes the 70. Rumpel S, Razeto A, Pillar CM, Vijayan V, Taylor A, Giller K, Gilmore
gelatinase biosynthesis-activating pheromone propeptide corresponding MS, Becker S, Zweckstetter M. 2004. Structure and DNA-binding prop-
to staphylococcal AgrD. J Bacteriol 188:8321–8326 http://dx.doi.org/10 erties of the cytolysin regulator CylR2 from Enterococcus faecalis. EMBO
.1128/JB.00865-06. J 23:3632–3642 http://dx.doi.org/10.1038/sj.emboj.7600367.

24 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

71. Coburn PS, Pillar CM, Jett BD, Haas W, Gilmore MS. 2004. En- 87. DebRoy S, Gebbie M, Ramesh A, Goodson JR, Cruz MR, van Hoof A,
terococcus faecalis senses target cells and in response expresses cytolysin. Winkler WC, Garsin DA. 2014. Riboswitches: a riboswitch-containing
Science 306:2270–2272 http://dx.doi.org/10.1126/science.1103996. sRNA controls gene expression by sequestration of a response regulator.
72. Leboeuf C, Leblanc L, Auffray Y, Hartke A. 2000. Characterization Science 345:937–940 http://dx.doi.org/10.1126/science.1255091.
of the ccpA gene of Enterococcus faecalis: identification of starvation- 88. Fox KA, Ramesh A, Stearns JE, Bourgogne A, Reyes-Jara A, Winkler
inducible proteins regulated by ccpA. J Bacteriol 182:5799–5806 http:// WC, Garsin DA. 2009. Multiple posttranscriptional regulatory mecha-
dx.doi.org/10.1128/JB.182.20.5799-5806.2000. nisms partner to control ethanolamine utilization in Enterococcus faecalis.
73. Suárez CA, Blancato VS, Poncet S, Deutscher J, Magni C. 2011. CcpA Proc Natl Acad Sci U S A 106:4435–4440 http://dx.doi.org/10.1073/pnas
represses the expression of the divergent cit operons of Enterococcus .0812194106.
faecalis through multiple cre sites. BMC Microbiol 11:227 http://dx.doi 89. Ramesh A, DebRoy S, Goodson JR, Fox KA, Faz H, Garsin DA,
.org/10.1186/1471-2180-11-227. Winkler WC. 2012. The mechanism for RNA recognition by ANTAR
74. Mortera P, Espariz M, Suárez C, Repizo G, Deutscher J, Alarcón S, regulators of gene expression. PLoS Genet 8:e1002666 http://dx.doi.org
Blancato V, Magni C. 2012. Fine-tuned transcriptional regulation of malate /10.1371/journal.pgen.1002666.
operons in Enterococcus faecalis. Appl Environ Microbiol 78:1936–1945 90. Ghim S-Y, Kim CC, Bonner ER, D’Elia JN, Grabner GK, Switzer RL.
http://dx.doi.org/10.1128/AEM.07280-11. 1999. The Enterococcus faecalis pyr operon is regulated by autogenous
75. Suárez C, Espariz M, Blancato VS, Magni C. 2013. Expression of the transcriptional attenuation at a single site in the 5′ leader. J Bacteriol 181:
agmatine deiminase pathway in Enterococcus faecalis is activated by the 1324–1329.
AguR regulator and repressed by CcpA and PTS(Man) systems. PLoS One 91. Panina EM, Vitreschak AG, Mironov AA, Gelfand MS. 2003. Regu-
8:e76170 http://dx.doi.org/10.1371/journal.pone.0076170. lation of biosynthesis and transport of aromatic amino acids in low-GC
76. Somarajan SR, Roh JH, Singh KV, Weinstock GM, Murray BE. 2014. Gram-positive bacteria. FEMS Microbiol Lett 222:211–220 http://dx.doi
CcpA is important for growth and virulence of Enterococcus faecium. .org/10.1016/S0378-1097(03)00303-3.
Infect Immun 82:3580–3587 http://dx.doi.org/10.1128/IAI.01911-14. 92. Su YA, He P, Clewell DB. 1992. Characterization of the tet(M) deter-
77. Solioz M, Stoyanov JV. 2003. Copper homeostasis in Enterococcus minant of Tn916: evidence for regulation by transcription attenuation.
hirae. FEMS Microbiol Rev 27:183–195 http://dx.doi.org/10.1016/S0168 Antimicrob Agents Chemother 36:769–778 http://dx.doi.org/10.1128/AAC
-6445(03)00053-6. .36.4.769.
78. Low YL, Jakubovics NS, Flatman JC, Jenkinson HF, Smith AW. 2003. 93. Gao P, Pinkston KL, Nallapareddy SR, van Hoof A, Murray BE,
Manganese-dependent regulation of the endocarditis-associated virulence Harvey BR. 2010. Enterococcus faecalis rnjB is required for pilin gene
factor EfaA of Enterococcus faecalis. J Med Microbiol 52:113–119 http:// expression and biofilm formation. J Bacteriol 192:5489–5498 http://dx
dx.doi.org/10.1099/jmm.0.05039-0. .doi.org/10.1128/JB.00725-10.
79. Abrantes MC, Kok J, Lopes MF. 2013. EfaR is a major regulator of 94. Gao P, Pinkston KL, Bourgogne A, Murray BE, van Hoof A, Harvey
Enterococcus faecalis manganese transporters and influences processes BR. 2017. Functional studies of E. faecalis RNase J2 and its role in vir-
involved in host colonization and infection. Infect Immun 81:935–944 ulence and fitness. PLoS One 12:e0175212 http://dx.doi.org/10.1371
http://dx.doi.org/10.1128/IAI.06377-11. /journal.pone.0175212.
80. Ross RP, Claiborne A. 1997. Evidence for regulation of the NADH 95. Innocenti N, Golumbeanu M, Fouquier d’Hérouël A, Lacoux C,
peroxidase gene (npr) from Enterococcus faecalis by OxyR. FEMS Microbiol Bonnin RA, Kennedy SP, Wessner F, Serror P, Bouloc P, Repoila F, Aurell
Lett 151:177–183 http://dx.doi.org/10.1111/j.1574-6968.1997.tb12567.x. E. 2015. Whole-genome mapping of 5′ RNA ends in bacteria by tagged
sequencing: a comprehensive view in Enterococcus faecalis. RNA 21:1018–
81. Verneuil N, Rincé A, Sanguinetti M, Posteraro B, Fadda G, Auffray Y,
1030 http://dx.doi.org/10.1261/rna.048470.114.
Hartke A, Giard J-C. 2005. Contribution of a PerR-like regulator to the
oxidative-stress response and virulence of Enterococcus faecalis. Micro- 96. Fouquier d’Hérouel A, Wessner F, Halpern D, Ly-Vu J, Kennedy SP,
biology 151:3997–4004 http://dx.doi.org/10.1099/mic.0.28325-0. Serror P, Aurell E, Repoila F. 2011. A simple and efficient method to
82. Kajfasz JK, Mendoza JE, Gaca AO, Miller JH, Koselny KA, Giambiagi- search for selected primary transcripts: non-coding and antisense RNAs
in the human pathogen Enterococcus faecalis. Nucleic Acids Res 39:e46
Demarval M, Wellington M, Abranches J, Lemos JA. 2012. The Spx reg-
ulator modulates stress responses and virulence in Enterococcus faecalis. http://dx.doi.org/10.1093/nar/gkr012.
Infect Immun 80:2265–2275 http://dx.doi.org/10.1128/IAI.00026-12. 97. Shioya K, Michaux C, Kuenne C, Hain T, Verneuil N, Budin-Verneuil
83. Verneuil N, Sanguinetti M, Le Breton Y, Posteraro B, Fadda G, Auffray A, Hartsch T, Hartke A, Giard J-C. 2011. Genome-wide identification of
Y, Hartke A, Giard J-C. 2004. Effects of the Enterococcus faecalis hypR small RNAs in the opportunistic pathogen Enterococcus faecalis V583.
gene encoding a new transcriptional regulator on oxidative stress response PLoS One 6:e23948 http://dx.doi.org/10.1371/journal.pone.0023948.
and intracellular survival within macrophages. Infect Immun 72:4424– 98. Michaux C, Hartke A, Martini C, Reiss S, Albrecht D, Budin-Verneuil
4431 http://dx.doi.org/10.1128/IAI.72.8.4424-4431.2004. A, Sanguinetti M, Engelmann S, Hain T, Verneuil N, Giard J-C. 2014.
84. La Carbona S, Sauvageot N, Giard J-C, Benachour A, Posteraro B, Involvement of Enterococcus faecalis small RNAs in stress response
Auffray Y, Sanguinetti M, Hartke A. 2007. Comparative study of the phys- and virulence. Infect Immun 82:3599–3611 http://dx.doi.org/10.1128/IAI
iological roles of three peroxidases (NADH peroxidase, alkyl hydroperoxide .01900-14.
reductase and thiol peroxidase) in oxidative stress response, survival inside 99. Sinel C, Augagneur Y, Sassi M, Bronsard J, Cacaci M, Guérin F,
macrophages and virulence of Enterococcus faecalis. Mol Microbiol 66: Sanguinetti M, Meignen P, Cattoir V, Felden B. 2017. Small RNAs in
1148–1163 http://dx.doi.org/10.1111/j.1365-2958.2007.05987.x. vancomycin-resistant Enterococcus faecium involved in daptomycin re-
85. Verneuil N, Rincé A, Sanguinetti M, Auffray Y, Hartke A, Giard J-C. sponse and resistance. Sci Rep 7:11067 http://dx.doi.org/10.1038/s41598
2005. Implication of hypR in the virulence and oxidative stress response -017-11265-2.
of Enterococcus faecalis. FEMS Microbiol Lett 252:137–141 http://dx.doi 100. Kristich CJ, Rice LB, Arias CA. 2014. Enterococcal infection: treat-
.org/10.1016/j.femsle.2005.08.043. ment and antibiotic resistance. In Gilmore MS, Clewell DB, Ike Y, Shankar
86. Papadimitriou K, Alegría Á, Bron PA, de Angelis M, Gobbetti M, N (ed), Enterococci: from Commensals to Leading Causes of Drug Resis-
Kleerebezem M, Lemos JA, Linares DM, Ross P, Stanton C, Turroni F, tant Infection. Massachusetts Eye and Ear Infirmary, Boston, MA. https://
van Sinderen D, Varmanen P, Ventura M, Zúñiga M, Tsakalidou E, Kok www.ncbi.nlm.nih.gov/books/NBK190420/.
J. 2016. Stress physiology of lactic acid bacteria. Microbiol Mol Biol Rev 101. Weigel LM, Clewell DB, Gill SR, Clark NC, McDougal LK,
80:837–890 http://dx.doi.org/10.1128/MMBR.00076-15. Flannagan SE, Kolonay JF, Shetty J, Killgore GE, Tenover FC. 2003.

ASMscience.org/MicrobiolSpectrum 25
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

Genetic analysis of a high-level vancomycin-resistant isolate of Staphylo- faecium BM4147. Biochemistry 32:5057–5063 http://dx.doi.org/10.1021
coccus aureus. Science 302:1569–1571 http://dx.doi.org/10.1126/science /bi00070a013.
.1090956. 118. Holman TR, Wu Z, Wanner BL, Walsh CT. 1994. Identification of
102. Flannagan SE, Chow JW, Donabedian SM, Brown WJ, Perri MB, the DNA-binding site for the phosphorylated VanR protein required for
Zervos MJ, Ozawa Y, Clewell DB. 2003. Plasmid content of a vancomycin- vancomycin resistance in Enterococcus faecium. Biochemistry 33:4625–
resistant Enterococcus faecalis isolate from a patient also colonized by 4631 http://dx.doi.org/10.1021/bi00181a024.
Staphylococcus aureus with a VanA phenotype. Antimicrob Agents Che- 119. Arthur M, Depardieu F, Gerbaud G, Galimand M, Leclercq R,
mother 47:3954–3959 http://dx.doi.org/10.1128/AAC.47.12.3954-3959 Courvalin P. 1997. The VanS sensor negatively controls VanR-mediated
.2003. transcriptional activation of glycopeptide resistance genes of Tn1546 and
103. Zhu W, Murray PR, Huskins WC, Jernigan JA, McDonald LC, Clark related elements in the absence of induction. J Bacteriol 179:97–106 http://
NC, Anderson KF, McDougal LK, Hageman JC, Olsen-Rasmussen M, dx.doi.org/10.1128/jb.179.1.97-106.1997.
Frace M, Alangaden GJ, Chenoweth C, Zervos MJ, Robinson-Dunn B, 120. Arthur M, Depardieu F, Courvalin P. 1999. Regulated interactions
Schreckenberger PC, Reller LB, Rudrik JT, Patel JB. 2010. Dissemination between partner and non-partner sensors and response regulators that
of an Enterococcus Inc18-Like vanA plasmid associated with vancomycin- control glycopeptide resistance gene expression in enterococci. Microbi-
resistant Staphylococcus aureus. Antimicrob Agents Chemother 54:4314– ology 145:1849–1858 http://dx.doi.org/10.1099/13500872-145-8-1849.
4320 http://dx.doi.org/10.1128/AAC.00185-10.
121. Baptista M, Depardieu F, Reynolds P, Courvalin P, Arthur M. 1997.
104. McGuinness WA, Malachowa N, DeLeo FR. 2017. Vancomycin re- Mutations leading to increased levels of resistance to glycopeptide anti-
sistance in Staphylococcus aureus. Yale J Biol Med 90:269–281. biotics in VanB-type enterococci. Mol Microbiol 25:93–105 http://dx.doi
105. Barna JCJ, Williams DH. 1984. The structure and mode of action of .org/10.1046/j.1365-2958.1997.4401812.x.
glycopeptide antibiotics of the vancomycin group. Annu Rev Microbiol 122. Hughes CS, Longo E, Phillips-Jones MK, Hussain R. 2017. Characteri-
38:339–357 http://dx.doi.org/10.1146/annurev.mi.38.100184.002011. sation of the selective binding of antibiotics vancomycin and teicoplanin
106. Arthur M, Courvalin P. 1993. Genetics and mechanisms of glyco- by the VanS receptor regulating type A vancomycin resistance in the en-
peptide resistance in enterococci. Antimicrob Agents Chemother 37:1563– terococci. Biochim Biophys Acta Gen Subj 1861:1951–1959 http://dx.doi
1571 http://dx.doi.org/10.1128/AAC.37.8.1563. .org/10.1016/j.bbagen.2017.05.011.
107. Depardieu F, Podglajen I, Leclercq R, Collatz E, Courvalin P. 2007. 123. Heaton MP, Discotto LF, Pucci MJ, Handwerger S. 1996. Mobili-
Modes and modulations of antibiotic resistance gene expression. Clin zation of vancomycin resistance by transposon-mediated fusion of a VanA
Microbiol Rev 20:79–114 http://dx.doi.org/10.1128/CMR.00015-06. plasmid with an Enterococcus faecium sex pheromone-response plasmid.
108. Boyd DA, Willey BM, Fawcett D, Gillani N, Mulvey MR. 2008. Gene 171:9–17 http://dx.doi.org/10.1016/0378-1119(96)00022-4.
Molecular characterization of Enterococcus faecalis N06-0364 with low- 124. Tomita H, Tanimoto K, Hayakawa S, Morinaga K, Ezaki K, Oshima
level vancomycin resistance harboring a novel D-Ala-D-Ser gene cluster, H, Ike Y. 2003. Highly conjugative pMG1-like plasmids carrying Tn1546-
vanL. Antimicrob Agents Chemother 52:2667–2672 http://dx.doi.org/10 like transposons that encode vancomycin resistance in Enterococcus faecium.
.1128/AAC.01516-07. J Bacteriol 185:7024–7028 http://dx.doi.org/10.1128/JB.185.23.7024-7028
109. Xu X, Lin D, Yan G, Ye X, Wu S, Guo Y, Zhu D, Hu F, Zhang Y, .2003.
Wang F, Jacoby GA, Wang M. 2010. vanM, a new glycopeptide resistance 125. Evers S, Quintiliani R Jr, Courvalin P. 1996. Genetics of glycopeptide
gene cluster found in Enterococcus faecium. Antimicrob Agents Chemo- resistance in enterococci. Microb Drug Resist 2:219–223 http://dx.doi
ther 54:4643–4647 http://dx.doi.org/10.1128/AAC.01710-09. .org/10.1089/mdr.1996.2.219.
110. Lebreton F, Depardieu F, Bourdon N, Fines-Guyon M, Berger P, 126. Woodford N, Morrison D, Johnson AP, Bateman AC, Hastings JG,
Camiade S, Leclercq R, Courvalin P, Cattoir V. 2011. D-Ala-D-Ser VanN-type Elliott TS, Cookson B. 1995. Plasmid-mediated vanB glycopeptide resis-
transferable vancomycin resistance in Enterococcus faecium. Antimicrob tance in enterococci. Microb Drug Resist 1:235–240 http://dx.doi.org/10
Agents Chemother 55:4606–4612 http://dx.doi.org/10.1128/AAC.00714-11. .1089/mdr.1995.1.235.
111. Arias CA, Courvalin P, Reynolds PE. 2000. vanC cluster of vancomycin- 127. Carias LL, Rudin SD, Donskey CJ, Rice LB. 1998. Genetic link-
resistant Enterococcus gallinarum BM4174. Antimicrob Agents Chemother age and cotransfer of a novel, vanB-containing transposon (Tn5382) and
44:1660–1666 http://dx.doi.org/10.1128/AAC.44.6.1660-1666.2000. a low-affinity penicillin-binding protein 5 gene in a clinical vancomycin-
112. Dutta I, Reynolds PE. 2002. Biochemical and genetic characteriza- resistant Enterococcus faecium isolate. J Bacteriol 180:4426–4434.
tion of the vanC-2 vancomycin resistance gene cluster of Enterococcus 128. Garnier F, Taourit S, Glaser P, Courvalin P, Galimand M. 2000.
casseliflavus ATCC 25788. Antimicrob Agents Chemother 46:3125–3132 Characterization of transposon Tn1549, conferring VanB-type resistance
http://dx.doi.org/10.1128/AAC.46.10.3125-3132.2002. in Enterococcus spp. Microbiology 146:1481–1489 http://dx.doi.org/10
113. Abadía Patiño L, Courvalin P, Perichon B. 2002. vanE gene cluster .1099/00221287-146-6-1481.
of vancomycin-resistant Enterococcus faecalis BM4405. J Bacteriol 184: 129. García-Solache M, Lebreton F, McLaughlin RE, Whiteaker JD,
6457–6464 http://dx.doi.org/10.1128/JB.184.23.6457-6464.2002. Gilmore MS, Rice LB. 2016. Homologous recombination within large
114. Depardieu F, Bonora MG, Reynolds PE, Courvalin P. 2003. The chromosomal regions facilitates acquisition of β-lactam and vancomycin
vanG glycopeptide resistance operon from Enterococcus faecalis revisited. resistance in Enterococcus faecium. Antimicrob Agents Chemother 60:
Mol Microbiol 50:931–948 http://dx.doi.org/10.1046/j.1365-2958.2003 5777–5786 http://dx.doi.org/10.1128/AAC.00488-16.
.03737.x. 130. Manson JM, Hancock LE, Gilmore MS. 2010. Mechanism of chro-
115. Sahm DF, Marsilio MK, Piazza G. 1999. Antimicrobial resistance in mosomal transfer of Enterococcus faecalis pathogenicity island, capsule,
key bloodstream bacterial isolates: electronic surveillance with the Sur- antimicrobial resistance, and other traits. Proc Natl Acad Sci U S A 107:
veillance Network Database–USA. Clin Infect Dis 29:259–263 http://dx 12269–12274 http://dx.doi.org/10.1073/pnas.1000139107.
.doi.org/10.1086/520195. 131. Boyd DA, Conly J, Dedier H, Peters G, Robertson L, Slater E, Mulvey
116. Arthur M, Quintiliani R Jr. 2001. Regulation of VanA- and VanB- MR. 2000. Molecular characterization of the vanD gene cluster and a
type glycopeptide resistance in enterococci. Antimicrob Agents Chemother novel insertion element in a vancomycin-resistant Enterococcus isolated in
45:375–381 http://dx.doi.org/10.1128/AAC.45.2.375-381.2001. Canada. J Clin Microbiol 38:2392–2394.
117. Wright GD, Holman TR, Walsh CT. 1993. Purification and char- 132. Depardieu F, Reynolds PE, Courvalin P. 2003. VanD-type vancomycin-
acterization of VanR and the cytosolic domain of VanS: a two-component resistant Enterococcus faecium 10/96A. Antimicrob Agents Chemother 47:
regulatory system required for vancomycin resistance in Enterococcus 7–18 http://dx.doi.org/10.1128/AAC.47.1.7-18.2003.

26 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

133. Casadewall B, Reynolds PE, Courvalin P. 2001. Regulation of ex- 150. Weaver KE, Rice LR, Churchward G. 2002. Plasmids and trans-
pression of the vanD glycopeptide resistance gene cluster from Entero- posons, p 219–263. In Gilmore MS, Clewell DB, Courvalin P, Dunny G,
coccus faecium BM4339. J Bacteriol 183:3436–3446 http://dx.doi.org/10 Murray BE, Rice LB (ed), The Enterococci: Pathogenesis, Molecular Bi-
.1128/JB.183.11.3436-3446.2001. ology, and Antibiotic Resistance. ASM Press, Washington, DC. http://dx
134. Perichon B, Casadewall B, Reynolds P, Courvalin P. 2000. .doi.org/10.1128/9781555817923.ch6
Glycopeptide-resistant Enterococcus faecium BM4416 is a VanD-type strain 151. Clewell DB, An FY, Flannagan SE, Antiporta M, Dunny GM. 2000.
with an impaired D-alanine:D-alanine ligase. Antimicrob Agents Chemother Enterococcal sex pheromone precursors are part of signal sequences for
44:1346–1348 http://dx.doi.org/10.1128/AAC.44.5.1346-1348.2000. surface lipoproteins. Mol Microbiol 35:246–247 http://dx.doi.org/10.1046
135. Marshall CG, Lessard IAD, Park I, Wright GD. 1998. Glycopeptide /j.1365-2958.2000.01687.x.
antibiotic resistance genes in glycopeptide-producing organisms. Antimi- 152. An FY, Clewell DB. 2002. Identification of the cAD1 sex pheromone
crob Agents Chemother 42:2215–2220 http://dx.doi.org/10.1128/AAC precursor in Enterococcus faecalis. J Bacteriol 184:1880–1887 http://dx
.42.9.2215. .doi.org/10.1128/JB.184.7.1880-1887.2002.
136. Marshall CG, Broadhead G, Leskiw BK, Wright GD. 1997. D-Ala- 153. Antiporta MH, Dunny GM. 2002. ccfA, the genetic determinant for
D-Ala ligases from glycopeptide antibiotic-producing organisms are highly the cCF10 peptide pheromone in Enterococcus faecalis OG1RF. J Bacteriol
homologous to the enterococcal vancomycin-resistance ligases VanA and 184:1155–1162 http://dx.doi.org/10.1128/jb.184.4.1155-1162.2002.
VanB. Proc Natl Acad Sci U S A 94:6480–6483 http://dx.doi.org/10.1073 154. Flannagan SE, Clewell DB. 2002. Identification and characterization
/pnas.94.12.6480. of genes encoding sex pheromone cAM373 activity in Enterococcus
137. Clewell DB, Weaver KE, Ike Y, Coque TM, Francia MV, Hayes F. faecalis and Staphylococcus aureus. Mol Microbiol 44:803–817 http://dx
2014. Extrachromosomal and mobile elements in enterococci: transmis- .doi.org/10.1046/j.1365-2958.2002.02922.x.
sion, maintenance, and epidemiology. In Gilmore MS, Clewell DB, Ike 155. An FY, Sulavik MC, Clewell DB. 1999. Identification and char-
Y, Shankar N (ed), Enterococci: from Commensals to Leading Causes of acterization of a determinant (eep) on the Enterococcus faecalis chromo-
Drug Resistant Infection. Massachusetts Eye and Ear Infirmary, Boston, some that is involved in production of the peptide sex pheromone cAD1.
MA. https://www.ncbi.nlm.nih.gov/books/NBK190430/. J Bacteriol 181:5915–5921.
138. Jensen LB, Garcia-Migura L, Valenzuela AJS, Løhr M, Hasman H, 156. Brown MS, Ye J, Rawson RB, Goldstein JL. 2000. Regulated intra-
Aarestrup FM. 2010. A classification system for plasmids from entero- membrane proteolysis: a control mechanism conserved from bacteria
cocci and other Gram-positive bacteria. J Microbiol Methods 80:25–43 to humans. Cell 100:391–398 http://dx.doi.org/10.1016/S0092-8674(00)
http://dx.doi.org/10.1016/j.mimet.2009.10.012. 80675-3.
139. Lioy VS, Pratto F, de la Hoz AB, Ayora S, Alonso JC. 2010. Plasmid 157. Clewell DB, Pontius LT, An FY, Ike Y, Suzuki A, Nakayama J. 1990.
pSM19035, a model to study stable maintenance in Firmicutes. Plasmid Nucleotide sequence of the sex pheromone inhibitor (iAD1) determinant
64:1–17 http://dx.doi.org/10.1016/j.plasmid.2010.04.002. of Enterococcus faecalis conjugative plasmid pAD1. Plasmid 24:156–161
140. Ruiz-Masó JA, MachóN C, Bordanaba-Ruiseco L, Espinosa M, Coll http://dx.doi.org/10.1016/0147-619X(90)90019-9.
M, Del Solar G. 2015. Plasmid rolling-circle replication. Microbiol Spectr 158. Chandler JR, Dunny GM. 2008. Characterization of the sequence
3:PLAS-0035-2014. doi:10.1128/microbiolspec.PLAS-0035-2014. specificity determinants required for processing and control of sex phero-
141. del Solar G, Espinosa M. 2000. Plasmid copy number control: an mone by the intramembrane protease Eep and the plasmid-encoded protein
ever-growing story. Mol Microbiol 37:492–500 http://dx.doi.org/10.1046 PrgY. J Bacteriol 190:1172–1183 http://dx.doi.org/10.1128/JB.01327-07.
/j.1365-2958.2000.02005.x. 159. Berg T, Firth N, Apisiridej S, Hettiaratchi A, Leelaporn A, Skurray
142. del Solar G, Giraldo R, Ruiz-Echevarría MJ, Espinosa M, Díaz- RA. 1998. Complete nucleotide sequence of pSK41: evolution of staphy-
Orejas R. 1998. Replication and control of circular bacterial plasmids. lococcal conjugative multiresistance plasmids. J Bacteriol 180:4350–4359.
Microbiol Mol Biol Rev 62:434–464. 160. Varahan S, Harms N, Gilmore MS, Tomich JM, Hancock LE.
143. Weaver KE, Kwong SM, Firth N, Francia MV. 2009. The RepA_N 2014. An ABC transporter, PptAB, is required for secretion of peptide sex
replicons of Gram-positive bacteria: a family of broadly distributed but pheromones in Enterococcus faecalis. MBio 5:e01726-14.
narrow host range plasmids. Plasmid 61:94–109 http://dx.doi.org/10 161. Leonard BAB, Podbielski A, Hedberg PJ, Dunny GM. 1996. En-
.1016/j.plasmid.2008.11.004. terococcus faecalis pheromone binding protein, PrgZ, recruits a chromo-
144. Fuqua WC, Winans SC. 1994. A LuxR-LuxI type regulatory system somal oligopeptide permease system to import sex pheromone cCF10 for
activates Agrobacterium Ti plasmid conjugal transfer in the presence of a induction of conjugation. Proc Natl Acad Sci U S A 93:260–264 http://dx
plant tumor metabolite. J Bacteriol 176:2796–2806 http://dx.doi.org/10 .doi.org/10.1073/pnas.93.1.260.
.1128/jb.176.10.2796-2806.1994. 162. Nakayama J, Takanami Y, Horii T, Sakuda S, Suzuki A. 1998.
145. Clewell DB, Flannagan SE, Zitzow LA, Su YA, He P, Senghas E, Molecular mechanism of peptide-specific pheromone signaling in Entero-
Weaver KE. 1991. Properties of conjugative transposon Tn916, p 39–44. coccus faecalis: functions of pheromone receptor TraA and pheromone-
In Dunny GM, Cleary P, McKay L (ed), Genetics and Molecular Biology binding protein TraC encoded by plasmid pPD1. J Bacteriol 180:449–456.
of Streptococci, Lactococci, and Enterococci. ASM Press, Washington, 163. Ruhfel RE, Manias DA, Dunny GM. 1993. Cloning and character-
DC. ization of a region of the Enterococcus faecalis conjugative plasmid, pCF10,
146. Cook LC, Federle MJ. 2014. Peptide pheromone signaling in Strep- encoding a sex pheromone-binding function. J Bacteriol 175:5253–5259
tococcus and Enterococcus. FEMS Microbiol Rev 38:473–492 http://dx http://dx.doi.org/10.1128/jb.175.16.5253-5259.1993.
.doi.org/10.1111/1574-6976.12046. 164. Tanimoto K, An FY, Clewell DB. 1993. Characterization of the traC
147. Dunny GM. 2013. Enterococcal sex pheromones: signaling, social determinant of the Enterococcus faecalis hemolysin-bacteriocin plasmid
behavior, and evolution. Annu Rev Genet 47:457–482 http://dx.doi.org pAD1: binding of sex pheromone. J Bacteriol 175:5260–5264 http://dx
/10.1146/annurev-genet-111212-133449. .doi.org/10.1128/jb.175.16.5260-5264.1993.
148. Dunny GM, Berntsson RP-A. 2016. Enterococcal sex pheromones: 165. Berntsson RPA, Schuurman-Wolters GK, Dunny G, Slotboom D-J,
evolutionary pathways to complex, two-signal systems. J Bacteriol 198: Poolman B. 2012. Structure and mode of peptide binding of pheromone
1556–1562 http://dx.doi.org/10.1128/JB.00128-16. receptor PrgZ. J Biol Chem 287:37165–37170 http://dx.doi.org/10.1074
149. Dunny GM, Johnson CM. 2011. Regulatory circuits controlling en- /jbc.M112.386334.
terococcal conjugation: lessons for functional genomics. Curr Opin Micro- 166. Fujimoto S, Clewell DB. 1998. Regulation of the pAD1 sex phero-
biol 14:174–180 http://dx.doi.org/10.1016/j.mib.2011.01.008. mone response of Enterococcus faecalis by direct interaction between the

ASMscience.org/MicrobiolSpectrum 27
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

cAD1 peptide mating signal and the negatively regulating, DNA-binding 181. Tanimoto K, Clewell DB. 1993. Regulation of the pAD1-encoded
TraA protein. Proc Natl Acad Sci U S A 95:6430–6435 http://dx.doi.org sex pheromone response in Enterococcus faecalis: expression of the pos-
/10.1073/pnas.95.11.6430. itive regulator TraE1. J Bacteriol 175:1008–1018 http://dx.doi.org/10
167. Neiditch MB, Capodagli GC, Prehna G, Federle MJ. 2017. Genetic .1128/jb.175.4.1008-1018.1993.
and structural analyses of RRNPP intercellular peptide signaling of Gram- 182. Bensing BA, Manias DA, Dunny GM. 1997. Pheromone cCF10 and
positive bacteria. Annu Rev Genet 51:311–333 http://dx.doi.org/10.1146 plasmid pCF10-encoded regulatory molecules act post-transcriptionally
/annurev-genet-120116-023507. to activate expression of downstream conjugation functions. Mol Micro-
168. Folli C, Mangiarotti L, Folloni S, Alfieri B, Gobbo M, Berni R, biol 24:285–294 http://dx.doi.org/10.1046/j.1365-2958.1997.3301710.x.
Rivetti C. 2008. Specificity of the TraA-DNA interaction in the regu- 183. Chung JW, Bensing BA, Dunny GM. 1995. Genetic analysis of a
lation of the pPD1-encoded sex pheromone response in Enterococcus region of the Enterococcus faecalis plasmid pCF10 involved in positive
faecalis. J Mol Biol 380:932–945 http://dx.doi.org/10.1016/j.jmb.2008 regulation of conjugative transfer functions. J Bacteriol 177:2107–2117
.05.058. http://dx.doi.org/10.1128/jb.177.8.2107-2117.1995.
169. Kozlowicz BK, Shi K, Gu Z-Y, Ohlendorf DH, Earhart CA, Dunny 184. Chung JW, Dunny GM. 1995. Transcriptional analysis of a region
GM. 2006. Molecular basis for control of conjugation by bacterial pher- of the Enterococcus faecalis plasmid pCF10 involved in positive regulation
omone and inhibitor peptides. Mol Microbiol 62:958–969 http://dx.doi of conjugative transfer functions. J Bacteriol 177:2118–2124 http://dx
.org/10.1111/j.1365-2958.2006.05434.x. .doi.org/10.1128/jb.177.8.2118-2124.1995.
170. Chen Y, Bandyopadhyay A, Kozlowicz BK, Haemig HAH, Tai A, 185. An FY, Clewell DB. 1994. Characterization of the determinant (traB)
Hu WS, Dunny GM. 2017. Mechanisms of peptide sex pheromone reg- encoding sex pheromone shutdown by the hemolysin/bacteriocin plasmid
ulation of conjugation in Enterococcus faecalis. MicrobiologyOpen 6: pAD1 in Enterococcus faecalis. Plasmid 31:215–221 http://dx.doi.org/10
e00492 http://dx.doi.org/10.1002/mbo3.492. .1006/plas.1994.1023.
171. Bae T, Kozlowicz B, Dunny GM. 2002. Two targets in pCF10 DNA 186. Ike Y, Clewell DB. 1984. Genetic analysis of the pAD1 pheromone
for PrgX binding: their role in production of Qa and prgX mRNA and in response in Streptococcus faecalis, using transposon Tn917 as an inser-
regulation of pheromone-inducible conjugation. J Mol Biol 315:995–1007 tional mutagen. J Bacteriol 158:777–783.
http://dx.doi.org/10.1006/jmbi.2001.5294.
187. Weaver KE, Clewell DB. 1990. Regulation of the pAD1 sex phero-
172. Tomita H, Clewell DB. 2000. A pAD1-encoded small RNA mole- mone response in Enterococcus faecalis: effects of host strain and traA,
cule, mD, negatively regulates Enterococcus faecalis pheromone response traB, and C region mutants on expression of an E region pheromone-
by enhancing transcription termination. J Bacteriol 182:1062–1073 http:// inducible lacZ fusion. J Bacteriol 172:2633–2641 http://dx.doi.org/10
dx.doi.org/10.1128/JB.182.4.1062-1073.2000. .1128/jb.172.5.2633-2641.1990.
173. Bae T, Kozlowicz BK, Dunny GM. 2004. Characterization of cis- 188. Chandler JR, Flynn AR, Bryan EM, Dunny GM. 2005. Specific control
acting prgQ mutants: evidence for two distinct repression mechanisms by of endogenous cCF10 pheromone by a conserved domain of the pCF10-
Qa RNA and PrgX protein in pheromone-inducible enterococcal plasmid encoded regulatory protein PrgY in Enterococcus faecalis. J Bacteriol 187:
pCF10. Mol Microbiol 51:271–281 http://dx.doi.org/10.1046/j.1365-2958 4830–4843 http://dx.doi.org/10.1128/JB.187.14.4830-4843.2005.
.2003.03832.x.
189. De Boever EH, Clewell DB, Fraser CM. 2000. Enterococcus faecalis
174. Johnson CM, Manias DA, Haemig HAH, Shokeen S, Weaver conjugative plasmid pAM373: complete nucleotide sequence and genetic
KE, Henkin TM, Dunny GM. 2010. Direct evidence for control of the
analyses of sex pheromone response. Mol Microbiol 37:1327–1341 http://
pheromone-inducible prgQ operon of Enterococcus faecalis plasmid pCF10 dx.doi.org/10.1046/j.1365-2958.2000.02072.x.
by a countertranscript-driven attenuation mechanism. J Bacteriol 192:1634–
1642 http://dx.doi.org/10.1128/JB.01525-09. 190. Pontius LT, Clewell DB. 1991. A phase variation event that acti-
vates conjugation functions encoded by the Enterococcus faecalis plas-
175. Shokeen S, Johnson CM, Greenfield TJ, Manias DA, Dunny GM,
mid pAD1. Plasmid 26:172–185 http://dx.doi.org/10.1016/0147-619X
Weaver KE. 2010. Structural analysis of the anti-Q-Qs interaction: RNA-
(91)90041-T.
mediated regulation of E. faecalis plasmid pCF10 conjugation. Plasmid
64:26–35 http://dx.doi.org/10.1016/j.plasmid.2010.03.002. 191. Hirt H, Wirth R, Muscholl A. 1996. Comparative analysis of 18 sex
pheromone plasmids from Enterococcus faecalis: detection of a new in-
176. de Freire Bastos MC, Tanimoto K, Clewell DB. 1997. Regulation
sertion element on pPD1 and implications for the evolution of this plasmid
of transfer of the Enterococcus faecalis pheromone-responding plasmid
family. Mol Gen Genet 252:640–647.
pAD1: temperature-sensitive transfer mutants and identification of a new
regulatory determinant, traD. J Bacteriol 179:3250–3259 http://dx.doi 192. Olmsted SB, Erlandsen SL, Dunny GM, Wells CL. 1993. High-
.org/10.1128/jb.179.10.3250-3259.1997. resolution visualization by field emission scanning electron microscopy
of Enterococcus faecalis surface proteins encoded by the pheromone-
177. do Carmo de Freire Bastos M, Tomita H, Tanimoto K, Clewell DB.
1998. Regulation of the Enterococcus faecalis pAD1-related sex phero- inducible conjugative plasmid pCF10. J Bacteriol 175:6229–6237 http://
dx.doi.org/10.1128/jb.175.19.6229-6237.1993.
mone response: analyses of traD expression and its role in controlling
conjugation functions. Mol Microbiol 30:381–392 http://dx.doi.org/10 193. Dunny GM, Zimmerman DL, Tortorello ML. 1985. Induction of
.1046/j.1365-2958.1998.01074.x. surface exclusion (entry exclusion) by Streptococcus faecalis sex phero-
178. Bae T, Dunny GM. 2001. Dominant-negative mutants of prgX: evi- mones: use of monoclonal antibodies to identify an inducible surface an-
dence for a role for PrgX dimerization in negative regulation of pheromone- tigen involved in the exclusion process. Proc Natl Acad Sci U S A 82:
inducible conjugation. Mol Microbiol 39:1307–1320 http://dx.doi.org/10 8582–8586 http://dx.doi.org/10.1073/pnas.82.24.8582.
.1111/j.1365-2958.2001.02319.x. 194. Weidlich G, Wirth R, Galli D. 1992. Sex pheromone plasmid pAD1-
179. Chatterjee A, Johnson CM, Shu C-C, Kaznessis YN, Ramkrishna D, encoded surface exclusion protein of Enterococcus faecalis. Mol Gen
Dunny GM, Hu W-S. 2011. Convergent transcription confers a bistable Genet 233:161–168 http://dx.doi.org/10.1007/BF00587575.
switch in Enterococcus faecalis conjugation. Proc Natl Acad Sci U S A 195. Clewell DB, Weaver KE. 1989. Sex pheromones and plasmid transfer
108:9721–9726 http://dx.doi.org/10.1073/pnas.1101569108. in Enterococcus faecalis. Plasmid 21:175–184 http://dx.doi.org/10.1016
180. Johnson CM, Haemig HHA, Chatterjee A, Wei-Shou H, Weaver /0147-619X(89)90041-3.
KE, Dunny GM. 2011. RNA-mediated reciprocal regulation between two 196. De Boever EH, Clewell DB. 2001. The Enterococcus faecalis
bacterial operons is RNase III dependent. MBio 2:e00189-11 http://dx.doi pheromone-responsive plasmid pAM373 does not encode an entry exclu-
.org/10.1128/mBio.00189-11. sion function. Plasmid 45:57–60 http://dx.doi.org/10.1006/plas.2000.1494.

28 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

197. Ehrenfeld EE, Kessler RE, Clewell DB. 1986. Identification of 211. Alvarez-Martinez CE, Christie PJ. 2009. Biological diversity of pro-
pheromone-induced surface proteins in Streptococcus faecalis and evi- karyotic type IV secretion systems. Microbiol Mol Biol Rev 73:775–808
dence of a role for lipoteichoic acid in formation of mating aggregates. http://dx.doi.org/10.1128/MMBR.00023-09.
J Bacteriol 168:6–12 http://dx.doi.org/10.1128/jb.168.1.6-12.1986. 212. Chen Y, Zhang X, Manias D, Yeo H-J, Dunny GM, Christie PJ.
198. Trotter KM, Dunny GM. 1990. Mutants of Enterococcus fae- 2008. Enterococcus faecalis PcfC, a spatially localized substrate receptor
calisdeficient as recipients in mating with donors carrying pheromone- for type IV secretion of the pCF10 transfer intermediate. J Bacteriol 190:
inducible plasmids. Plasmid 24:57–67 http://dx.doi.org/10.1016/0147-619X 3632–3645 http://dx.doi.org/10.1128/JB.01999-07.
(90)90025-8. 213. Whitaker N, Chen Y, Jakubowski SJ, Sarkar MK, Li F, Christie PJ.
199. Waters CM, Dunny GM. 2001. Analysis of functional domains of 2015. The all-alpha domains of coupling proteins from the Agrobacterium
the Enterococcus faecalis pheromone-induced surface protein aggregation tumefaciens VirB/VirD4 and Enterococcus faecalis pCF10-encoded type IV
substance. J Bacteriol 183:5659–5667 http://dx.doi.org/10.1128/JB.183 secretion systems confer specificity to binding of cognate DNA substrates.
.19.5659-5667.2001. J Bacteriol 197:2335–2349 http://dx.doi.org/10.1128/JB.00189-15.
200. Waters CM, Hirt H, McCormick JK, Schlievert PM, Wells CL, Dunny 214. Li F, Alvarez-Martinez C, Chen Y, Choi K-J, Yeo H-J, Christie PJ.
GM. 2004. An amino-terminal domain of Enterococcus faecalis aggre- 2012. Enterococcus faecalis PrgJ, a VirB4-like ATPase, mediates pCF10
gation substance is required for aggregation, bacterial internalization by conjugative transfer through substrate binding. J Bacteriol 194:4041–
epithelial cells and binding to lipoteichoic acid. Mol Microbiol 52:1159– 4051 http://dx.doi.org/10.1128/JB.00648-12.
1171 http://dx.doi.org/10.1111/j.1365-2958.2004.04045.x. 215. Laverde Gomez JA, Bhatty M, Christie PJ. 2014. PrgK, a multido-
201. Chow JW, Thal LA, Perri MB, Vazquez JA, Donabedian SM, Clewell main peptidoglycan hydrolase, is essential for conjugative transfer of the
DB, Zervos MJ. 1993. Plasmid-associated hemolysin and aggregation pheromone-responsive plasmid pCF10. J Bacteriol 196:527–539 http://
substance production contribute to virulence in experimental enterococcal dx.doi.org/10.1128/JB.00950-13.
endocarditis. Antimicrob Agents Chemother 37:2474–2477 http://dx.doi 216. Francia MV, Clewell DB. 2002. Transfer origins in the conjugative
.org/10.1128/AAC.37.11.2474.
Enterococcus faecalis plasmids pAD1 and pAM373: identification of the
202. Chuang-Smith ON, Wells CL, Henry-Stanley MJ, Dunny GM. 2010. pAD1 nic site, a specific relaxase and a possible TraG-like protein. Mol Mi-
Acceleration of Enterococcus faecalis biofilm formation by aggregation crobiol 45:375–395 http://dx.doi.org/10.1046/j.1365-2958.2002.03007.x.
substance expression in an ex vivo model of cardiac valve colonization.
217. Garcillán-Barcia MP, Francia MV, de la Cruz F. 2009. The diver-
PLoS One 5:e15798 http://dx.doi.org/10.1371/journal.pone.0015798.
sity of conjugative relaxases and its application in plasmid classification.
203. Rakita RM, Vanek NN, Jacques-Palaz K, Mee M, Mariscalco MM, FEMS Microbiol Rev 33:657–687 http://dx.doi.org/10.1111/j.1574-6976
Dunny GM, Snuggs M, Van Winkle WB, Simon SI. 1999. Enterococ- .2009.00168.x.
cus faecalis bearing aggregation substance is resistant to killing by human
218. Francia MV, Clewell DB, de la Cruz F, Moncalián G. 2013. Catalytic
neutrophils despite phagocytosis and neutrophil activation. Infect Immun
domain of plasmid pAD1 relaxase TraX defines a group of relaxases re-
67:6067–6075.
lated to restriction endonucleases. Proc Natl Acad Sci U S A 110:13606–
204. Schlievert PM, Chuang-Smith ON, Peterson ML, Cook LCC, Dunny 13611 http://dx.doi.org/10.1073/pnas.1310037110.
GM. 2010. Enterococcus faecalis endocarditis severity in rabbits is re-
duced by IgG Fabs interfering with aggregation substance. PLoS One 5: 219. Staddon JH, Bryan EM, Manias DA, Chen Y, Dunny GM. 2006.
Genetic characterization of the conjugative DNA processing system of
e13194 http://dx.doi.org/10.1371/journal.pone.0013194.
enterococcal plasmid pCF10. Plasmid 56:102–111 http://dx.doi.org/10
205. Schlievert PM, Gahr PJ, Assimacopoulos AP, Dinges MM, Stoehr .1016/j.plasmid.2006.05.001.
JA, Harmala JW, Hirt H, Dunny GM. 1998. Aggregation and binding
substances enhance pathogenicity in rabbit models of Enterococcus fae- 220. Staddon JH, Bryan EM, Manias DA, Dunny GM. 2004. Conserved
calis endocarditis. Infect Immun 66:218–223. target for group II intron insertion in relaxase genes of conjugative ele-
ments of Gram-positive bacteria. J Bacteriol 186:2393–2401 http://dx.doi
206. Chandler JR, Hirt H, Dunny GM. 2005. A paracrine peptide sex .org/10.1128/JB.186.8.2393-2401.2004.
pheromone also acts as an autocrine signal to induce plasmid transfer and
virulence factor expression in vivo. Proc Natl Acad Sci U S A 102:15617– 221. Chen Y, Staddon JH, Dunny GM. 2007. Specificity determinants of
15622 http://dx.doi.org/10.1073/pnas.0505545102. conjugative DNA processing in the Enterococcus faecalis plasmid pCF10
and the Lactococcus lactis plasmid pRS01. Mol Microbiol 63:1549–1564
207. Bhatty M, Cruz MR, Frank KL, Gomez JA, Andrade F, Garsin DA,
http://dx.doi.org/10.1111/j.1365-2958.2007.05610.x.
Dunny GM, Kaplan HB, Christie PJ. 2015. Enterococcus faecalis pCF10-
encoded surface proteins PrgA, PrgB (aggregation substance) and PrgC 222. Fujimoto S, Tomita H, Wakamatsu E, Tanimoto K, Ike Y. 1995.
contribute to plasmid transfer, biofilm formation and virulence. Mol Mi- Physical mapping of the conjugative bacteriocin plasmid pPD1 of En-
crobiol 95:660–677 http://dx.doi.org/10.1111/mmi.12893. terococcus faecalis and identification of the determinant related to the
pheromone response. J Bacteriol 177:5574–5581 http://dx.doi.org/10.1128
208. Bhatty M, Camacho MI, Gonzalez-Rivera C, Frank KL, Dale JL,
/jb.177.19.5574-5581.1995.
Manias DA, Dunny GM, Christie PJ. 2017. PrgU: a suppressor of sex
pheromone toxicity in Enterococcus faecalis. Mol Microbiol 103:398–412 223. Hedberg PJ, Leonard BAB, Ruhfel RE, Dunny GM. 1996. Identifi-
http://dx.doi.org/10.1111/mmi.13563. cation and characterization of the genes of Enterococcus faecalis plas-
209. Francia MV, Haas W, Wirth R, Samberger E, Muscholl-Silberhorn mid pCF10 involved in replication and in negative control of pheromone-
A, Gilmore MS, Ike Y, Weaver KE, An FY, Clewell DB. 2001. Completion inducible conjugation. Plasmid 35:46–57 http://dx.doi.org/10.1006/plas
of the nucleotide sequence of the Enterococcus faecalis conjugative viru- .1996.0005.
lence plasmid pAD1 and identification of a second transfer origin. Plasmid 224. Weaver KE, Clewell DB, An F. 1993. Identification, characterization,
46:117–127 http://dx.doi.org/10.1006/plas.2001.1533. and nucleotide sequence of a region of Enterococcus faecalis pheromone-
210. Hirt H, Manias DA, Bryan EM, Klein JR, Marklund JK, Staddon responsive plasmid pAD1 capable of autonomous replication. J Bacteriol
JH, Paustian ML, Kapur V, Dunny GM. 2005. Characterization of the 175:1900–1909 http://dx.doi.org/10.1128/jb.175.7.1900-1909.1993.
pheromone response of the Enterococcus faecalis conjugative plasmid 225. Francia MV, Fujimoto S, Tille P, Weaver KE, Clewell DB. 2004.
pCF10: complete sequence and comparative analysis of the transcriptional Replication of Enterococcus faecalis pheromone-responding plasmid pAD1:
and phenotypic responses of pCF10-containing cells to pheromone in- location of the minimal replicon and oriV site and RepA involvement in
duction. J Bacteriol 187:1044–1054 http://dx.doi.org/10.1128/JB.187.3 initiation of replication. J Bacteriol 186:5003–5016 http://dx.doi.org/10
.1044-1054.2005. .1128/JB.186.15.5003-5016.2004.

ASMscience.org/MicrobiolSpectrum 29
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

226. Gering M, Götz F, Brückner R. 1996. Sequence and analysis of the expression of a 33-amino-acid toxic peptide by an unusual mechanism.
replication region of the Staphylococcus xylosus plasmid pSX267. Gene Mol Microbiol 37:652–660 http://dx.doi.org/10.1046/j.1365-2958.2000
182:117–122 http://dx.doi.org/10.1016/S0378-1119(96)00526-4. .02035.x.
227. Kwong SM, Skurray RA, Firth N. 2004. Staphylococcus aureus 245. Greenfield TJ, Weaver KE. 2000. Antisense RNA regulation of the
multiresistance plasmid pSK41: analysis of the replication region, initiator pAD1 par post-segregational killing system requires interaction at the
protein binding and antisense RNA regulation. Mol Microbiol 51:497– 5′ and 3′ ends of the RNAs. Mol Microbiol 37:661–670 http://dx.doi.org
509 http://dx.doi.org/10.1046/j.1365-2958.2003.03843.x. /10.1046/j.1365-2958.2000.02034.x.
228. Tanaka T, Ogura M. 1998. A novel Bacillus natto plasmid pLS32 246. Greenfield TJ, Franch T, Gerdes K, Weaver KE. 2001. Antisense RNA
capable of replication in Bacillus subtilis. FEBS Lett 422:243–246 http:// regulation of the par post-segregational killing system: structural analysis
dx.doi.org/10.1016/S0014-5793(98)00015-5. and mechanism of binding of the antisense RNA, RNAII and its target,
229. Dunny GM, Leonard BAB. 1997. Cell-cell communication in Gram- RNAI. Mol Microbiol 42:527–537 http://dx.doi.org/10.1046/j.1365-2958
positive bacteria. Annu Rev Microbiol 51:527–564 http://dx.doi.org/10 .2001.02663.x.
.1146/annurev.micro.51.1.527. 247. Shokeen S, Patel S, Greenfield TJ, Brinkman C, Weaver KE. 2008.
230. Schumacher MA, Tonthat NK, Kwong SM, Chinnam NB, Liu MA, Translational regulation by an intramolecular stem-loop is required for
Skurray RA, Firth N. 2014. Mechanism of staphylococcal multiresistance intermolecular RNA regulation of the par addiction module. J Bacteriol
plasmid replication origin assembly by the RepA protein. Proc Natl Acad 190:6076–6083 http://dx.doi.org/10.1128/JB.00660-08.
Sci U S A 111:9121–9126 http://dx.doi.org/10.1073/pnas.1406065111. 248. Shokeen S, Greenfield TJ, Ehli EA, Rasmussen J, Perrault BE, Weaver
231. Paulsson J, Chattoraj DK. 2006. Origin inactivation in bacterial DNA KE. 2009. An intramolecular upstream helix ensures the stability of a
replication control. Mol Microbiol 61:9–15 http://dx.doi.org/10.1111/j toxin-encoding RNA in Enterococcus faecalis. J Bacteriol 191:1528–1536
.1365-2958.2006.05229.x. http://dx.doi.org/10.1128/JB.01316-08.
232. Kwong SM, Firth N. 2015. Structural and sequence requirements 249. Weaver KE, Ehli EA, Nelson JS, Patel S. 2004. Antisense RNA reg-
for the antisense RNA regulating replication of staphylococcal multire- ulation by stable complex formation in the Enterococcus faecalis plasmid
sistance plasmid pSK41. Plasmid 78:17–25 http://dx.doi.org/10.1016/j pAD1 par addiction system. J Bacteriol 186:6400–6408 http://dx.doi.org
.plasmid.2015.01.002. /10.1128/JB.186.19.6400-6408.2004.
233. Gerdes K, Møller-Jensen J, Bugge Jensen R. 2000. Plasmid and chro- 250. Patel S, Weaver KE. 2006. Addiction toxin Fst has unique effects on
mosome partitioning: surprises from phylogeny. Mol Microbiol 37:455–466 chromosome segregation and cell division in Enterococcus faecalis and
http://dx.doi.org/10.1046/j.1365-2958.2000.01975.x. Bacillus subtilis. J Bacteriol 188:5374–5384 http://dx.doi.org/10.1128/JB
234. Francia MV, Weaver KE, Goicoechea P, Tille P, Clewell DB. 2007. .00513-06.
Characterization of an active partition system for the Enterococcus fae- 251. Weaver KE, Reddy SG, Brinkman CL, Patel S, Bayles KW, Endres JL.
calis pheromone-responding plasmid pAD1. J Bacteriol 189:8546–8555 2009. Identification and characterization of a family of toxin-antitoxin
http://dx.doi.org/10.1128/JB.00719-07. systems related to the Enterococcus faecalis plasmid pAD1 par addiction
235. Heath DG, An FY, Weaver KE, Clewell DB. 1995. Phase variation module. Microbiology 155:2930–2940 http://dx.doi.org/10.1099/mic.0
of Enterococcus faecalis pAD1 conjugation functions relates to changes in .030932-0.
iteron sequence region. J Bacteriol 177:5453–5459 http://dx.doi.org/10 252. Weaver KE, Weaver DM, Wells CL, Waters CM, Gardner ME,
.1128/jb.177.19.5453-5459.1995. Ehli EA. 2003. Enterococcus faecalis plasmid pAD1-encoded Fst toxin
236. Hayes F. 2003. Toxins-antitoxins: plasmid maintenance, programmed affects membrane permeability and alters cellular responses to lantibiotics.
cell death, and cell cycle arrest. Science 301:1496–1499 http://dx.doi.org J Bacteriol 185:2169–2177 http://dx.doi.org/10.1128/JB.185.7.2169-2177
/10.1126/science.1088157. .2003.
237. Brantl S, Jahn N. 2015. sRNAs in bacterial type I and type III toxin- 253. Göbl C, Kosol S, Stockner T, Rückert HM, Zangger K. 2010. Solu-
antitoxin systems. FEMS Microbiol Rev 39:413–427 http://dx.doi.org/10 tion structure and membrane binding of the toxin fst of the par addiction
.1093/femsre/fuv003. module. Biochemistry 49:6567–6575 http://dx.doi.org/10.1021/bi1005128.
238. Gerdes K. 2013. Prokaryotic Toxin-Antitoxins. Springer-Verlag, Berlin, 254. Fozo EM, Hemm MR, Storz G. 2008. Small toxic proteins and the
Germany. http://dx.doi.org/10.1007/978-3-642-33253-1. antisense RNAs that repress them. Microbiol Mol Biol Rev 72:579–589
239. Weaver KE. 2012. The par toxin-antitoxin system from Enterococ- http://dx.doi.org/10.1128/MMBR.00025-08.
cus faecalis plasmid pAD1 and its chromosomal homologs. RNA Biol 255. Fozo EM, Makarova KS, Shabalina SA, Yutin N, Koonin EV, Storz
9:1498–1503 http://dx.doi.org/10.4161/rna.22311. G. 2010. Abundance of type I toxin-antitoxin systems in bacteria: searches
240. Weaver KE. 2013. Type I toxin-antitoxin loci: hok/soc and fst, for new candidates and discovery of novel families. Nucleic Acids Res
p 9–26. In Gerdes K (ed), Prokaryotic Toxin-Antitoxins. Springer-Verlag, 38:3743–3759 http://dx.doi.org/10.1093/nar/gkq054.
Berlin, Germany. http://dx.doi.org/10.1007/978-3-642-33253-1_2 256. Kwong SM, Jensen SO, Firth N. 2010. Prevalence of Fst-like toxin-
241. Weaver KE. 2015. The type I toxin-antitoxin par locus from En- antitoxin systems. Microbiology 156:975–977, discussion 977 http://dx
terococcus faecalis plasmid pAD1: RNA regulation by both cis- and trans- .doi.org/10.1099/mic.0.038323-0.
acting elements. Plasmid 78:65–70 http://dx.doi.org/10.1016/j.plasmid.2014 257. Kawano M, Oshima T, Kasai H, Mori H. 2002. Molecular charac-
.10.001. terization of long direct repeat (LDR) sequences expressing a stable mRNA
242. Weaver KE, Tritle DJ. 1994. Identification and characterization of an encoding for a 35-amino-acid cell-killing peptide and a cis-encoded small
Enterococcus faecalis plasmid pAD1-encoded stability determinant which antisense RNA in Escherichia coli. Mol Microbiol 45:333–349 http://dx
produces two small RNA molecules necessary for its function. Plasmid 32: .doi.org/10.1046/j.1365-2958.2002.03042.x.
168–181 http://dx.doi.org/10.1006/plas.1994.1053. 258. Weaver KE, Chen Y, Miiller EM, Johnson JN, Dangler AA, Manias
243. Weaver KE, Jensen KD, Colwell A, Sriram SI. 1996. Functional DA, Clem AM, Schjodt DJ, Dunny GM. 2017. Examination of Enterococcus
analysis of the Enterococcus faecalis plasmid pAD1-encoded stability de- faecalis toxin-antitoxin system toxin Fst function utilizing a pheromone-
terminant par. Mol Microbiol 20:53–63 http://dx.doi.org/10.1111/j.1365 inducible expression vector with tight repression and broad dynamic range.
-2958.1996.tb02488.x. J Bacteriol 199:199 http://dx.doi.org/10.1128/JB.00065-17.
244. Greenfield TJ, Ehli E, Kirshenmann T, Franch T, Gerdes K, Weaver 259. Brinkman CL, Bumgarner R, Kittichotirat W, Dunman PM,
KE. 2000. The antisense RNA of the par locus of pAD1 regulates the Kuechenmeister LJ, Weaver KE. 2013. Characterization of the effects of an

30 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

rpoC mutation that confers resistance to the Fst peptide toxin-antitoxin sys- recombinases. EMBO J 8:2425–2433 http://dx.doi.org/10.1002/j.1460
tem toxin. J Bacteriol 195:156–166 http://dx.doi.org/10.1128/JB.01597-12. -2075.1989.tb08373.x.
260. Arthur M, Molinas C, Depardieu F, Courvalin P. 1993. Charac- 277. Marra D, Scott JR. 1999. Regulation of excision of the conjugative
terization of Tn1546, a Tn3-related transposon conferring glycopeptide transposon Tn916. Mol Microbiol 31:609–621 http://dx.doi.org/10.1046
resistance by synthesis of depsipeptide peptidoglycan precursors in En- /j.1365-2958.1999.01201.x.
terococcus faecium BM4147. J Bacteriol 175:117–127 http://dx.doi.org 278. Grainge I, Jayaram M. 1999. The integrase family of recombinase:
/10.1128/jb.175.1.117-127.1993. organization and function of the active site. Mol Microbiol 33:449–456
261. Perkins JB, Youngman PJ. 1984. A physical and functional analy- http://dx.doi.org/10.1046/j.1365-2958.1999.01493.x.
sis of Tn917, a Streptococcus transposon in the Tn3 family that functions 279. Lu F, Churchward G. 1994. Conjugative transposition: Tn916 in-
in Bacillus. Plasmid 12:119–138 http://dx.doi.org/10.1016/0147-619X tegrase contains two independent DNA binding domains that recognize
(84)90058-1. different DNA sequences. EMBO J 13:1541–1548 http://dx.doi.org/10
262. Hodel-Christian SL, Murray BE. 1991. Characterization of the .1002/j.1460-2075.1994.tb06416.x.
gentamicin resistance transposon Tn5281 from Enterococcus faecalis and 280. Wojciak JM, Connolly KM, Clubb RT. 1999. NMR structure of the
comparison to staphylococcal transposons Tn4001 and Tn4031. Anti- Tn916 integrase-DNA complex. Nat Struct Biol 6:366–373 http://dx.doi
microb Agents Chemother 35:1147–1152 http://dx.doi.org/10.1128/AAC .org/10.1038/7603.
.35.6.1147. 281. Connolly KM, Ilangovan U, Wojciak JM, Iwahara M, Clubb RT.
263. Quintiliani R Jr, Courvalin P. 1996. Characterization of Tn1547, a 2000. Major groove recognition by three-stranded β-sheets: affinity de-
composite transposon flanked by the IS16 and IS256-like elements, that terminants and conserved structural features. J Mol Biol 300:841–856
confers vancomycin resistance in Enterococcus faecalis BM4281. Gene http://dx.doi.org/10.1006/jmbi.2000.3888.
172:1–8 http://dx.doi.org/10.1016/0378-1119(96)00110-2. 282. Taylor KL, Churchward G. 1997. Specific DNA cleavage mediated
264. Rice LB, Carias LL. 1998. Transfer of Tn5385, a composite, multi- by the integrase of conjugative transposon Tn916. J Bacteriol 179:1117–
resistance chromosomal element from Enterococcus faecalis. J Bacteriol 1125 http://dx.doi.org/10.1128/jb.179.4.1117-1125.1997.
180:714–721. 283. Caparon MG, Scott JR. 1989. Excision and insertion of the conju-
265. Shoemaker NB, Smith MD, Guild WR. 1980. DNase-resistant trans- gative transposon Tn916 involves a novel recombination mechanism. Cell
fer of chromosomal cat and tet insertions by filter mating in Pneumococcus. 59:1027–1034 http://dx.doi.org/10.1016/0092-8674(89)90759-9.
Plasmid 3:80–87 http://dx.doi.org/10.1016/S0147-619X(80)90036-0. 284. Jaworski DD, Clewell DB. 1994. Evidence that coupling sequences
266. Shoemaker NB, Barber RD, Salyers AA. 1989. Cloning and charac- play a frequency-determining role in conjugative transposition of Tn916
terization of a Bacteroides conjugal tetracycline-erythromycin resistance in Enterococcus faecalis. J Bacteriol 176:3328–3335 http://dx.doi.org/10
element by using a shuttle cosmid vector. J Bacteriol 171:1294–1302 http:// .1128/jb.176.11.3328-3335.1994.
dx.doi.org/10.1128/jb.171.3.1294-1302.1989. 285. Pethel B, Churchward G. 2000. Coupling sequences flanking Tn916
267. Burrus V, Pavlovic G, Decaris B, Guédon G. 2002. Conjugative do not determine the affinity of binding of integrase to the transposon ends
transposons: the tip of the iceberg. Mol Microbiol 46:601–610 http://dx and adjacent bacterial DNA. Plasmid 43:123–129 http://dx.doi.org/10
.doi.org/10.1046/j.1365-2958.2002.03191.x. .1006/plas.1999.1440.
268. Burrus V. 2017. Mechanisms of stabilization of integrative and con- 286. Marra D, Pethel B, Churchward GG, Scott JR. 1999. The frequency
jugative elements. Curr Opin Microbiol 38:44–50 http://dx.doi.org/10 of conjugative transposition of Tn916 is not determined by the frequency
.1016/j.mib.2017.03.014. of excision. J Bacteriol 181:5414–5418.
269. Delavat F, Miyazaki R, Carraro N, Pradervand N, van der Meer 287. Rudy CK, Scott JR, Churchward G. 1997. DNA binding by the Xis
JR. 2017. The hidden life of integrative and conjugative elements. FEMS protein of the conjugative transposon Tn916. J Bacteriol 179:2567–2572
Microbiol Rev 41:512–537 http://dx.doi.org/10.1093/femsre/fux008. http://dx.doi.org/10.1128/jb.179.8.2567-2572.1997.
270. Johnson CM, Grossman AD. 2015. Integrative and conjugative 288. Su YA, Clewell DB. 1993. Characterization of the left 4 kb of
elements (ICEs): what they do and how they work. Annu Rev Genet 49: conjugative transposon Tn916: determinants involved in excision. Plas-
577–601 http://dx.doi.org/10.1146/annurev-genet-112414-055018. mid 30:234–250 http://dx.doi.org/10.1006/plas.1993.1055.
271. Wozniak RAF, Waldor MK. 2010. Integrative and conjugative ele- 289. Marra D, Smith JG, Scott JR. 1999. Excision of the conjugative
ments: mosaic mobile genetic elements enabling dynamic lateral gene flow. transposon Tn916 in Lactococcus lactis. Appl Environ Microbiol 65:
Nat Rev Microbiol 8:552–563 http://dx.doi.org/10.1038/nrmicro2382. 2230–2231.
272. Roberts AP, Mullany P. 2009. A modular master on the move: the 290. Rudy C, Taylor KL, Hinerfeld D, Scott JR, Churchward G. 1997.
Tn916 family of mobile genetic elements. Trends Microbiol 17:251–258 Excision of a conjugative transposon in vitro by the Int and Xis proteins of
http://dx.doi.org/10.1016/j.tim.2009.03.002. Tn916. Nucleic Acids Res 25:4061–4066 http://dx.doi.org/10.1093/nar
273. Roberts AP, Mullany P. 2011. Tn916-like genetic elements: a diverse /25.20.4061.
group of modular mobile elements conferring antibiotic resistance. FEMS 291. Connolly KM, Iwahara M, Clubb RT. 2002. Xis protein binding to the
Microbiol Rev 35:856–871 http://dx.doi.org/10.1111/j.1574-6976.2011 left arm stimulates excision of conjugative transposon Tn916. J Bacteriol
.00283.x. 184:2088–2099 http://dx.doi.org/10.1128/JB.184.8.2088-2099.2002.
274. Lu F, Churchward G. 1995. Tn916 target DNA sequences bind the 292. Hinerfeld D, Churchward G. 2001. Xis protein of the conjuga-
C-terminal domain of integrase protein with different affinities that cor- tive transposon Tn916 plays dual opposing roles in transposon excision.
relate with transposon insertion frequency. J Bacteriol 177:1938–1946 Mol Microbiol 41:1459–1467 http://dx.doi.org/10.1046/j.1365-2958.2001
http://dx.doi.org/10.1128/jb.177.8.1938-1946.1995. .02626.x.
275. Hosking SL, Deadman ME, Moxon ER, Peden JF, Saunders NJ, 293. Jaworski DD, Clewell DB. 1995. A functional origin of transfer
High NJ. 1998. An in silico evaluation of Tn916 as a tool for generalized (oriT) on the conjugative transposon Tn916. J Bacteriol 177:6644–6651
mutagenesis in Haemophilus influenzae Rd. Microbiology 144:2525– http://dx.doi.org/10.1128/jb.177.22.6644-6651.1995.
2530 http://dx.doi.org/10.1099/00221287-144-9-2525. 294. Rocco JM, Churchward G. 2006. The integrase of the conjugative
276. Poyart-Salmeron C, Trieu-Cuot P, Carlier C, Courvalin P. 1989. transposon Tn916 directs strand- and sequence-specific cleavage of the
Molecular characterization of two proteins involved in the excision of origin of conjugal transfer, oriT, by the endonuclease Orf20. J Bacteriol
the conjugative transposon Tn1545: homologies with other site-specific 188:2207–2213 http://dx.doi.org/10.1128/JB.188.6.2207-2213.2006.

ASMscience.org/MicrobiolSpectrum 31
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Weaver

295. Flannagan SE, Zitzow LA, Su YA, Clewell DB. 1994. Nucleotide 310. Llosa M, Bolland S, Grandoso G, de la Cruz F. 1994. Conjugation-
sequence of the 18-kb conjugative transposon Tn916 from Enterococcus independent, site-specific recombination at the oriT of the IncW plasmid
faecalis. Plasmid 32:350–354 http://dx.doi.org/10.1006/plas.1994.1077. R388 mediated by TrwC. J Bacteriol 176:3210–3217 http://dx.doi.org/10
296. Senghas E, Jones JM, Yamamoto M, Gawron-Burke C, Clewell .1128/jb.176.11.3210-3217.1994.
DB. 1988. Genetic organization of the bacterial conjugative transposon 311. Francia MV, Clewell DB. 2002. Amplification of the tetracycline
Tn916. J Bacteriol 170:245–249 http://dx.doi.org/10.1128/jb.170.1.245 resistance determinant of pAMalpha1 in Enterococcus faecalis requires
-249.1988. a site-specific recombination event involving relaxase. J Bacteriol 184:
297. Celli J, Trieu-Cuot P. 1998. Circularization of Tn916 is required for 5187–5193 http://dx.doi.org/10.1128/JB.184.18.5187-5193.2002.
expression of the transposon-encoded transfer functions: characterization 312. Yagi Y, Clewell DB. 1980. Amplification of the tetracycline resis-
of long tetracycline-inducible transcripts reading through the attachment tance determinant of plasmid pAM α 1 in Streptococcus faecalis: depen-
site. Mol Microbiol 28:103–117 http://dx.doi.org/10.1046/j.1365-2958 dence on host recombination machinery. J Bacteriol 143:1070–1072.
.1998.00778.x.
313. Ike Y, Tanimoto K, Tomita H, Takeuchi K, Fujimoto S. 1998.
298. Scornec H, Bellanger X, Guilloteau H, Groshenry G, Merlin C. 2017. Efficient transfer of the pheromone-independent Enterococcus faecium
Inducibility of Tn916 conjugative transfer in Enterococcus faecalis by plasmid pMG1 (Gmr) (65.1 kilobases) to Enterococcus strains during
subinhibitory concentrations of ribosome-targeting antibiotics. J Antimi- broth mating. J Bacteriol 180:4886–4892.
crob Chemother 72:2722–2728 http://dx.doi.org/10.1093/jac/dkx202.
314. Tomita H, Pierson C, Lim SK, Clewell DB, Ike Y. 2002. Possible
299. Bringel F, Van Alstine GL, Scott JR. 1992. Conjugative transposition connection between a widely disseminated conjugative gentamicin resis-
of Tn916: the transposon int gene is required only in the donor. J Bacteriol tance (pMG1-like) plasmid and the emergence of vancomycin resistance in
174:4036–4041 http://dx.doi.org/10.1128/jb.174.12.4036-4041.1992. Enterococcus faecium. J Clin Microbiol 40:3326–3333 http://dx.doi.org
300. Norgren M, Scott JR. 1991. The presence of conjugative transposon /10.1128/JCM.40.9.3326-3333.2002.
Tn916 in the recipient strain does not impede transfer of a second copy of 315. Tanimoto K, Ike Y. 2008. Complete nucleotide sequencing and
the element. J Bacteriol 173:319–324 http://dx.doi.org/10.1128/jb.173.1 analysis of the 65-kb highly conjugative Enterococcus faecium plasmid
.319-324.1991. pMG1: identification of the transfer-related region and the minimum re-
301. Flannagan SE, Clewell DB. 1991. Conjugative transfer of Tn916 gion required for replication. FEMS Microbiol Lett 288:186–195 http://
in Enterococcus faecalis: trans activation of homologous transposons. dx.doi.org/10.1111/j.1574-6968.2008.01342.x.
J Bacteriol 173:7136–7141 http://dx.doi.org/10.1128/jb.173.22.7136-7141
316. Tomita H, Ike Y. 2005. Genetic analysis of transfer-related regions of
.1991.
the vancomycin resistance Enterococcus conjugative plasmid pHTbeta:
302. Burrus V, Pavlovic G, Decaris B, Guédon G. 2002. The ICESt1 ele- identification of oriT and a putative relaxase gene. J Bacteriol 187:7727–
ment of Streptococcus thermophilus belongs to a large family of integrative 7737 http://dx.doi.org/10.1128/JB.187.22.7727-7737.2005.
and conjugative elements that exchange modules and change their spe-
317. Tanimoto K, Ike Y. 2002. Analysis of the conjugal transfer system
cificity of integration. Plasmid 48:77–97 http://dx.doi.org/10.1016/S0147
of the pheromone-independent highly transferable Enterococcus plasmid
-619X(02)00102-6.
pMG1: identification of a tra gene (traA) up-regulated during conjugation.
303. Roberts AP, Johanesen PA, Lyras D, Mullany P, Rood JI. 2001. J Bacteriol 184:5800–5804 http://dx.doi.org/10.1128/JB.184.20.5800-5804
Comparison of Tn5397 from Clostridium difficile, Tn916 from Ente- .2002.
rococcus faecalis and the CW459tet(M) element from Clostridium per-
318. Grady R, Hayes F. 2003. Axe-Txe, a broad-spectrum proteic toxin-
fringens shows that they have similar conjugation regions but different
antitoxin system specified by a multidrug-resistant, clinical isolate of En-
insertion and excision modules. Microbiology 147:1243–1251 http://dx
terococcus faecium. Mol Microbiol 47:1419–1432 http://dx.doi.org/10
.doi.org/10.1099/00221287-147-5-1243.
.1046/j.1365-2958.2003.03387.x.
304. Osborn AM, Böltner D. 2002. When phage, plasmids, and transposons
collide: genomic islands, and conjugative- and mobilizable-transposons as 319. Halvorsen EM, Williams JJ, Bhimani AJ, Billings EA, Hergenrother
a mosaic continuum. Plasmid 48:202–212 http://dx.doi.org/10.1016/S0147 PJ. 2011. Txe, an endoribonuclease of the enterococcal Axe-Txe toxin-
-619X(02)00117-8. antitoxin system, cleaves mRNA and inhibits protein synthesis. Microbi-
ology 157:387–397 http://dx.doi.org/10.1099/mic.0.045492-0.
305. Mikalsen T, Pedersen T, Willems R, Coque TM, Werner G, Sadowy
E, van Schaik W, Jensen LB, Sundsfjord A, Hegstad K. 2015. Investigating 320. Condon C. 2006. Shutdown decay of mRNA. Mol Microbiol 61:
the mobilome in clinically important lineages of Enterococcus faecium and 573–583 http://dx.doi.org/10.1111/j.1365-2958.2006.05270.x.
Enterococcus faecalis. BMC Genomics 16:282 http://dx.doi.org/10.1186 321. Hayes F, Van Melderen L. 2011. Toxins-antitoxins: diversity, evo-
/s12864-015-1407-6. lution and function. Crit Rev Biochem Mol Biol 46:386–408 http://dx.doi
306. Duerkop BA, Palmer KL, Horsburgh MJ. 2014. Enterococcal .org/10.3109/10409238.2011.600437.
bacteriophages and genome defense. In Gilmore MS, Clewell DB, Ike Y, 322. Rosvoll TCS, Pedersen T, Sletvold H, Johnsen PJ, Sollid JE, Simonsen
Shankar N (ed), Enterococci: from Commensals to Leading Causes of GS, Jensen LB, Nielsen KM, Sundsfjord A. 2010. PCR-based plasmid
Drug Resistant Infection [Internet]. Massachusetts Eye and Ear Infirmary, typing in Enterococcus faecium strains reveals widely distributed pRE25-,
Boston, MA. https://www.ncbi.nlm.nih.gov/books/NBK190419/. pRUM-, pIP501- and pHTbeta-related replicons associated with glyco-
307. Perkins JB, Youngman P. 1983. Streptococcus plasmid pAM α 1 is a peptide resistance and stabilizing toxin-antitoxin systems. FEMS Immunol
composite of two separable replicons, one of which is closely related to Med Microbiol 58:254–268 http://dx.doi.org/10.1111/j.1574-695X.2009
Bacillus plasmid pBC16. J Bacteriol 155:607–615. .00633.x.
308. Clewell DB, Yagi Y, Dunny GM, Schultz SK. 1974. Characterization 323. Sletvold H, Johnsen PJ, Hamre I, Simonsen GS, Sundsfjord A,
of three plasmid deoxyribonucleic acid molecules in a strain of Strepto- Nielsen KM. 2008. Complete sequence of Enterococcus faecium pVEF3
coccus faecalis: identification of a plasmid determining erythromycin re- and the detection of an ω-ε-ζ toxin-antitoxin module and an ABC trans-
sistance. J Bacteriol 117:283–289. porter. Plasmid 60:75–85 http://dx.doi.org/10.1016/j.plasmid.2008.04
309. Broome-Smith J. 1980. RecA independent, site-specific recombina- .002.
tion between ColE1 or ColK and a miniplasmid they complement for 324. Lim S-K, Tanimoto K, Tomita H, Ike Y. 2006. Pheromone-responsive
mobilization and relaxation: implications for the mechanism of DNA conjugative vancomycin resistance plasmids in Enterococcus faecalis isolates
transfer during mobilization. Plasmid 4:51–63 http://dx.doi.org/10.1016 from humans and chicken feces. Appl Environ Microbiol 72:6544–6553
/0147-619X(80)90082-7. http://dx.doi.org/10.1128/AEM.00749-06.

32 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00
Enterococcal Genetics

325. de la Hoz AB, Ayora S, Sitkiewicz I, Fernández S, Pankiewicz R, ysis of the nosocomial pathogen Enterococcus faecium and identification
Alonso JC, Ceglowski P. 2000. Plasmid copy-number control and better- of a large transferable pathogenicity island. BMC Genomics 11:239–239
than-random segregation genes of pSM19035 share a common regulator. http://dx.doi.org/10.1186/1471-2164-11-239.
Proc Natl Acad Sci U S A 97:728–733 http://dx.doi.org/10.1073/pnas.97 341. Duerkop BA, Clements CV, Rollins D, Rodrigues JLM, Hooper LV.
.2.728. 2012. A composite bacteriophage alters colonization by an intestinal com-
326. Moreno-Del Álamo M, Tabone M, Lioy VS, Alonso JC. 2017. Toxin mensal bacterium. Proc Natl Acad Sci U S A 109:17621–17626 http://dx
ζ triggers a survival response to cope with stress and persistence. Front .doi.org/10.1073/pnas.1206136109.
Microbiol 8:1130 http://dx.doi.org/10.3389/fmicb.2017.01130. 342. Matos RC, Lapaque N, Rigottier-Gois L, Debarbieux L, Meylheuc T,
327. Soheili S, Ghafourian S, Sekawi Z, Neela VK, Sadeghifard N, Gonzalez-Zorn B, Repoila F, Lopes MF, Serror P. 2013. Enterococcus
Taherikalani M, Khosravi A, Ramli R, Hamat RA. 2015. The mazEF faecalis prophage dynamics and contributions to pathogenic traits. PLoS
toxin-antitoxin system as an attractive target in clinical isolates of En- Genet 9:e1003539 http://dx.doi.org/10.1371/journal.pgen.1003539.
terococcus faecium and Enterococcus faecalis. Drug Des Devel Ther 9: 343. Lepage E, Brinster S, Caron C, Ducroix-Crepy C, Rigottier-Gois L,
2553–2561. Dunny G, Hennequet-Antier C, Serror P. 2006. Comparative genomic
328. Wessner F, Lacoux C, Goeders N, Fouquier d’Hérouel A, Matos R, hybridization analysis of Enterococcus faecalis: identification of genes
Serror P, Van Melderen L, Repoila F. 2015. Regulatory crosstalk between absent from food strains. J Bacteriol 188:6858–6868 http://dx.doi.org/10
type I and type II toxin-antitoxin systems in the human pathogen En- .1128/JB.00421-06.
terococcus faecalis. RNA Biol 12:1099–1108 http://dx.doi.org/10.1080 344. Solheim M, Brekke MC, Snipen LG, Willems RJL, Nes IF, Brede
/15476286.2015.1084465. DA. 2011. Comparative genomic analysis reveals significant enrichment of
329. Cataudella I, Trusina A, Sneppen K, Gerdes K, Mitarai N. 2012. mobile genetic elements and genes encoding surface structure-proteins in
Conditional cooperativity in toxin-antitoxin regulation prevents random hospital-associated clonal complex 2 Enterococcus faecalis. BMC Micro-
toxin activation and promotes fast translational recovery. Nucleic Acids biol 11:3 http://dx.doi.org/10.1186/1471-2180-11-3.
Res 40:6424–6434 http://dx.doi.org/10.1093/nar/gks297. 345. Bourgogne A, Garsin DA, Qin X, Singh KV, Sillanpaa J, Yerrapragada
330. Hacker J, Kaper JB. 2000. Pathogenicity islands and the evolution S, Ding Y, Dugan-Rocha S, Buhay C, Shen H, Chen G, Williams G, Muzny
of microbes. Annu Rev Microbiol 54:641–679 http://dx.doi.org/10.1146 D, Maadani A, Fox KA, Gioia J, Chen L, Shang Y, Arias CA, Nallapareddy
/annurev.micro.54.1.641. SR, Zhao M, Prakash VP, Chowdhury S, Jiang H, Gibbs RA, Murray BE,
331. Lesic B, Carniel E. 2005. Horizontal transfer of the high-pathogenicity Highlander SK, Weinstock GM. 2008. Large scale variation in Enterococ-
island of Yersinia pseudotuberculosis. J Bacteriol 187:3352–3358 http:// cus faecalis illustrated by the genome analysis of strain OG1RF. Genome
dx.doi.org/10.1128/JB.187.10.3352-3358.2005. Biol 9:R110–R110 http://dx.doi.org/10.1186/gb-2008-9-7-r110.
332. Ruzin A, Lindsay J, Novick RP. 2001. Molecular genetics of SaPI1: a 346. Stevens RH, Ektefaie MR, Fouts DE. 2011. The annotated com-
mobile pathogenicity island in Staphylococcus aureus. Mol Microbiol plete DNA sequence of Enterococcus faecalis bacteriophage φEf11 and its
41:365–377 http://dx.doi.org/10.1046/j.1365-2958.2001.02488.x. comparison with all available phage and predicted prophage genomes.
333. Shankar N, Baghdayan AS, Gilmore MS. 2002. Modulation of vir- FEMS Microbiol Lett 317:9–26 http://dx.doi.org/10.1111/j.1574-6968
ulence within a pathogenicity island in vancomycin-resistant Enterococcus .2010.02203.x.
faecalis. Nature 417:746–750 http://dx.doi.org/10.1038/nature00802. 347. Mazaheri Nezhad Fard R, Barton MD, Heuzenroeder MW. 2011.
334. McBride SM, Coburn PS, Baghdayan AS, Willems RJL, Grande MJ, Bacteriophage-mediated transduction of antibiotic resistance in entero-
Shankar N, Gilmore MS. 2009. Genetic variation and evolution of the cocci. Lett Appl Microbiol 52:559–564 http://dx.doi.org/10.1111/j.1472
pathogenicity island of Enterococcus faecalis. J Bacteriol 191:3392–3402 -765X.2011.03043.x.
http://dx.doi.org/10.1128/JB.00031-09. 348. Yasmin A, Kenny JG, Shankar J, Darby AC, Hall N, Edwards C,
335. McBride SM, Fischetti VA, Leblanc DJ, Moellering RC Jr, Gilmore Horsburgh MJ. 2010. Comparative genomics and transduction potential
MS. 2007. Genetic diversity among Enterococcus faecalis. PLoS One 2: of Enterococcus faecalis temperate bacteriophages. J Bacteriol 192:1122–
e582 http://dx.doi.org/10.1371/journal.pone.0000582. 1130 http://dx.doi.org/10.1128/JB.01293-09.
336. Coburn PS, Baghdayan AS, Dolan GT, Shankar N. 2007. Horizontal 349. Rath D, Amlinger L, Rath A, Lundgren M. 2015. The CRISPR-Cas
transfer of virulence genes encoded on the Enterococcus faecalis patho- immune system: biology, mechanisms and applications. Biochimie 117:
genicity island. Mol Microbiol 63:530–544 http://dx.doi.org/10.1111/j 119–128 http://dx.doi.org/10.1016/j.biochi.2015.03.025.
.1365-2958.2006.05520.x. 350. Lyons C, Raustad N, Bustos MA, Shiaris M. 2015. Incidence of type
337. Oancea C, Klare I, Witte W, Werner G. 2004. Conjugative transfer II CRISPR1-Cas systems in Enterococcus is species-dependent. PLoS One
of the virulence gene, esp, among isolates of Enterococcus faecium and 10:e0143544 http://dx.doi.org/10.1371/journal.pone.0143544.
Enterococcus faecalis. J Antimicrob Chemother 54:232–235 http://dx.doi 351. Price VJ, Huo W, Sharifi A, Palmer KL. 2016. CRISPR-Cas and
.org/10.1093/jac/dkh249. restriction-modification act additively against conjugative antibiotic re-
338. Laverde Gomez JA, Hendrickx APA, Willems RJ, Top J, Sava I, sistance plasmid transfer in Enterococcus faecalis. MSphere 1:e00064-16
Huebner J, Witte W, Werner G. 2011. Intra- and interspecies genomic http://dx.doi.org/10.1128/mSphere.00064-16.
transfer of the Enterococcus faecalis pathogenicity island. PLoS One 6: 352. Hullahalli K, Rodrigues M, Palmer KL. 2017. Exploiting CRISPR-
e16720 http://dx.doi.org/10.1371/journal.pone.0016720. Cas to manipulate Enterococcus faecalis populations. eLife 6:e26664
339. Leavis HL, Willems RJL, Top J, Spalburg E, Mascini EM, Fluit AC, http://dx.doi.org/10.7554/eLife.26664.
Hoepelman A, de Neeling AJ, Bonten MJM. 2003. Epidemic and non- 353. Palmer KL, Godfrey P, Griggs A, Kos VN, Zucker J, Desjardins
epidemic multidrug-resistant Enterococcus faecium. Emerg Infect Dis 9: C, Cerqueira G, Gevers D, Walker S, Wortman J, Feldgarden M, Haas
1108–1115 http://dx.doi.org/10.3201/eid0909.020383. B, Birren B, Gilmore MS. 2012. Comparative genomics of enterococci:
340. van Schaik W, Top J, Riley DR, Boekhorst J, Vrijenhoek JEP, variation in Enterococcus faecalis, clade structure in E. faecium, and de-
Schapendonk CME, Hendrickx APA, Nijman IJ, Bonten MJM, Tettelin fining characteristics of E. gallinarum and E. casseliflavus. MBio 3:e00318-
H, Willems RJL. 2010. Pyrosequencing-based comparative genome anal- 11 http://dx.doi.org/10.1128/mBio.00318-11.

ASMscience.org/MicrobiolSpectrum 33
Downloaded from www.asmscience.org by
IP: 129.110.242.50
On: Sat, 09 Mar 2019 17:17:00

Das könnte Ihnen auch gefallen