Download as pdf or txt
Download as pdf or txt
You are on page 1of 466

GENÉTICA

MICROBIANA

MMCR 7003 – 1
Segundo Semestre
2022-2023
Gabriel Trueba
Instructions to Authors

and the text and an appropriate statement concerning the no-


menclatural status of the name should be made in the text.
Nomenclature of Microorganisms “Candidatus” species should always be set in quotation marks.
Microorganisms, viruses, and plasmids should be given des-
Binary names, consisting of a generic name and a specific epi- ignations consisting of letters and serial numbers. It is gener-
thet (e.g., Escherichia coli), must be used for all microorgan- ally advisable to include a worker’s initials or a descriptive sym-
isms. Names of categories at or above the genus level may be bol of locale or laboratory, etc., in the designation. Each new
used alone, but specific and subspecific epithets may not. A strain, mutant, isolate, or derivative should be given a new
specific epithet must be preceded by a generic name, written (serial) designation. This designation should be distinct from
out in full the first time it is used in a paper. Thereafter, the those of the genotype and phenotype, and genotypic and phe-
generic name should be abbreviated to the initial capital letter notypic symbols should not be included. Plasmids are named
(e.g., E. coli), provided there can be no confusion with other with a lowercase “p” followed by the designation in uppercase
genera used in the paper. Names of all taxa (kingdoms, phyla, letters and numbers. To avoid the use of the same designation
classes, orders, families, genera, species, and subspecies) are as that of a widely used strain or plasmid, check the designation
printed in italics and should be italicized in the manuscript; against a publication database such as Medline.
strain designations and numbers are not. Vernacular (com-
mon) names should be in lowercase roman type (e.g., strepto- Genetic Nomenclature
coccus, brucella). For Salmonella, genus, species, and subspe-
cies names should be rendered in standard form: Salmonella To facilitate accurate communication, it is important that
enterica at first use, S. enterica thereafter; Salmonella enterica standard genetic nomenclature be used whenever possible
subsp. arizonae at first use, S. enterica subsp. arizonae thereaf- and that deviations or proposals for new naming systems be
ter. Names of serovars should be in roman type with the first endorsed by an appropriate authoritative body. Review
letter capitalized: Salmonella enterica serovar Typhimurium. and/or publication of submitted manuscripts that contain new
After the first use, the serovar may also be given without a or nonstandard nomenclature may be delayed by the editor or
species name: Salmonella Typhimurium, S. Typhimurium, or the Journals Department so that they may be reviewed.
Salmonella serovar Typhimurium. For other information re-
garding serovar designations, see Antigenic Formulae of the Bacteria. The genetic properties of bacteria are described in
Sal-monella Serovars, 9th ed. (P. A. D. Grimont and F.-X. terms of phenotypes and genotypes. The phenotype describes
Weill, WHO Collaborating Centre for Reference and the observable properties of an organism. The genotype refers
Research on Salmonella, Institut Pasteur, Paris, France, 2007; to the genetic constitution of an organism, usually in reference
see http: //www.pasteur.fr/ip/portal/action/ to some standard wild type. The guidelines that follow are
WebdriveActionEvent/oid/ 01s-000036-089). For a summary based on the recommendations of Demerec et al. (Genetics
of the current standards for Salmonella nomenclature and the 54:61–76, 1966).
Kaufmann-White criteria, see the article by Brenner et al. (J. (i) Phenotypic designations must be used when mutant loci
Clin. Microbiol. 38:2465–2467, 2000), the opinion of the have not been identified or mapped. They can also be used to
Judicial Commission of the International Committee on identify the protein product of a gene, e.g., the OmpA protein.
Systematics of Prokaryotes (Int. J. Syst. Evol. Microbiol. Phenotypic designations generally consist of three-letter sym-
55:519 –520, 2005), and the article by Tindall et al. (Int. J. bols; these are not italicized, and the first letter of the symbol is
Syst. Evol. Microbiol. 55:521–524, 2005). The spelling of capitalized. It is preferable to use Roman or Arabic numerals
bacterial names should follow the Approved Lists of Bacterial (instead of letters) to identify a series of related phenotypes.
Names (Amended) & Index of the Bacterial and Yeast Thus, nucleic acid polymerase mutants might be designated
Nomenclatural Changes (V. B. D. Skerman et al., ed., Pol1, Pol2, and Pol3, etc. Wild-type characteristics can be des-
American Society for Microbiology, Washington, DC, 1989) ignated with a superscript plus (Pol⫹), and, when necessary for
and the validation lists and notification lists published in the clarity, negative superscripts (Pol⫺) can be used to designate
International Journal of Systematic and Evolutionary Microbiol- mutant characteristics. Lowercase superscript letters may be
ogy (formerly the International Journal of Systematic Bacteriol- used to further delineate phenotypes (e.g., Strr for streptomy-
ogy) since January 1989. In addition, two sites on the World cin resistance). Phenotypic designations should be defined.
Wide Web list current approved bacterial names: Prokaryotic (ii) Genotypic designations are also indicated by three-letter
No-menclature Up-to-Date (http://www.dsmz.de/bacterial- locus symbols. In contrast to phenotypic designations, these
diversity /prokaryotic-nomenclature-up-to-date.html) and List are lowercase italic (e.g., ara his rps). If several loci govern
of Pro-karyotic Names with Standing in Nomenclature related functions, these are distinguished by italicized capital
(http://www .bacterio.net/). If there is reason to use a name letters following the locus symbol (e.g., araA araB araC). Pro-
that does not have standing in nomenclature, the name moter, terminator, and operator sites should be indicated as
should be enclosed in quotation marks in the title and at its described by Bachmann and Low (Microbiol. Rev. 44:1–56,
first use in the abstract 1980), e.g., lacZp, lacAt, and lacZo.
(iii) Wild-type alleles are indicated with a superscript plus
(ara⫹ his⫹). A superscript minus is not used to indicate a mu-
August 2014, Instructions to Authors tant locus; thus, one refers to an ara mutant rather than an
ara⫺ strain.
(iv) Mutation sites are designated by placing serial isolation

Journal of Bacteriology jb.asm.org 15


Instructions to Authors

numbers (allele numbers) after the locus symbol (e.g., araA1 parentheses (or brackets) are used (␭, F⫹). Reference to an
araA2). If only a single such locus exists or if it is not known in integrated episome is indicated as described for inserted ele-
which of several related loci the mutation has occurred, a hy- ments, and an exogenote is shown as, for example, W3110/
phen is used instead of the capital letter (e.g., ara-23). It is F⬘8(gal⫹).
essential in papers reporting the isolation of new mutants that For information about the symbols in current use, consult
allele numbers be given to the mutations. For Escherichia coli, Berlyn (Microbiol. Mol. Biol. Rev. 62:814 –984, 1998) for E.
there is a registry of such numbers: the Coli Genetic Stock coli K-12, Sanderson and Roth (Microbiol. Rev. 52:485–532,
Center (http://cgsc.biology.yale.edu/). For the genus Salmo- 1988) for Salmonella serovar Typhimurium, Holloway et al.
nella, the registry is the Salmonella Genetic Stock Centre (http: (Microbiol. Rev. 43:73–102, 1979) for the genus Pseudomonas,
//people.ucalgary.ca/~kesander/). For the genus Bacillus, the and Piggot and Hoch (Microbiol. Rev. 49:158 –179, 1985) for
registry is the Bacillus Genetic Stock Center (http://www.bgsc Bacillus subtilis.
.org/).
(v) The use of superscripts with genotypes (other than ⫹ to Conventions for naming genes. It is recommended that
indicate wild-type alleles) should be avoided. Designations in- (entirely) new genes be given names that are mnemonics of
dicating amber mutations (Am), temperature-sensitive muta- their function, avoiding names that are already assigned and
tions (Ts), constitutive mutations (Con), cold-sensitive muta- earlier or alternative gene names, irrespective of the bacterium
tions (Cs), production of a hybrid protein (Hyb), and other for which such assignments have been made. Similarly, it is
important phenotypic properties should follow the allele num- recommended that, whenever possible, orthologous genes
ber [e.g., araA230(Am) hisD21(Ts)]. All other such designa- present in different organisms receive the same name. When
tions of phenotype must be defined at the first occurrence. If homology is not apparent or the function of a new gene has not
superscripts must be used, they must be approved by the editor been established, a provisional name may be given by one of
and defined at the first occurrence in the text. the following methods. (i) The gene may be named on the basis
Subscripts may be used in two situations. Subscripts may be of its map location in the style yaaA, analogous to the style used
used to distinguish between genes (having the same name) for recording transposon insertions (zef) as discussed below. A
from different organisms or strains; e.g., hisE. coli or hisK-12 for list of such names in use for E. coli has been published by Rudd
the his gene of E. coli or strain K-12, respectively, may be used (Microbiol. Mol. Biol. Rev. 62:985–1019, 1998). (ii) A provi-
to distinguish this gene from the his gene in another species or sional name may be given in the style described by Demerec et
strain. An abbreviation may also be used if it is explained. Sim- al. (e.g., usg, gene upstream of folC). Such names should be
ilarly, a subscript is also used to distinguish between genetic unique, and names such as orf or genX should not be used. For
elements that have the same name. For example, the promoters reference, the Coli Genetic Stock Center’s database includes an
of the gln operon can be designated glnAp1 and glnAp2. This updated listing of E. coli gene names and gene products. It is
form departs slightly from that recommended by Bachmann accessible on the Internet (http://cgsc.biology.yale.edu/index
and Low (e.g., desC1p). .php). A list can also be found in the work of Riley (Microbiol.
(vi) Deletions are indicated by the symbol ⌬ placed before Rev. 57:862–952, 1993). For the genes of other bacteria, con-
the deleted gene or region, e.g., ⌬trpA432, ⌬(aroP-aceE)419, or sult the references given above.
⌬(hisQ-hisJo)1256. Similarly, other symbols can be used (with For prokaryotes, gene names should not begin with prefixes
appropriate definition). Thus, a fusion of the ara and lac oper- indicating the genus and species from which the gene is derived
ons can be shown as ⌽(ara-lac)95. Likewise, ⌽(araB⬘- (for example, do not use EcmecA for the mecA gene from E.
lacZ⫹)96 indicates that the fusion results in a truncated araB coli). However, subscripts may be used where necessary to dis-
gene fused to an intact lacZ gene, and ⌽(malE-lacZ)97(Hyb) tinguish between genes from different organisms or strains as
shows that a hybrid protein is synthesized. An inversion is described in section v of “Bacteria” above.
shown as IN(rrnD-rrnE)1. An insertion of an E. coli his gene
into plasmid pSC101 at zero kilobases (0 kb) is shown as Locus tags. Locus tags are systematic, unique identifiers
pSC101 ⍀(0kb::K-12hisB)4. An alternative designation of an that are assigned to each gene in GenBank. All genes men-
insertion can be used in simple cases, e.g., galT236::Tn5. The tioned in a manuscript should be traceable to their sequences
number 236 refers to the locus of the insertion, and if the strain by the reader, and locus tags may be used for this purpose in
carries an additional gal mutation, it is listed separately. Addi- manuscripts to identify uncharacterized genes. In addition,
tional examples, which utilize a slightly different format, can be authors should check GenBank to make sure that they are us-
found in the papers by Campbell et al. and Novick et al. cited ing the correct, up-to-date format for locus tags (e.g., upper-
below. It is important in reporting the construction of strains case versus lowercase letters and the presence or absence of an
in which a mobile element was inserted and subsequently de- underscore, etc.). Locus tag formats vary between different or-
leted that this fact be noted in the strain table. This can be done ganisms and also may be updated for a given organism, so it is
by listing the genotype of the strain used as an intermediate in important to check GenBank at the time of manuscript prep-
a table footnote or by making a direct or parenthetical remark aration.
in the genotype, e.g., (F⫺), ⌬Mu cts, or mal::⌬Mu cts::lac. In
setting parenthetical remarks within the genotype or dividing “Mutant” versus “mutation.” Keep in mind the distinc-
the genotype into constituent elements, parentheses and tion between a mutation (an alteration of the primary se-
brackets are used without special meaning; brackets are used quence of the genetic material) and a mutant (a strain carrying
outside parentheses. To indicate the presence of an episome, one or more mutations). One may speak about the mapping of

16 August 2014, Instructions to Authors Journal of Bacteriology jb.asm.org


Instructions to Authors

a mutation, but one cannot map a mutant. Likewise, a mutant tors, and of Roberts et al. (Nucleic Acids Res. 31:1805–1812,
has no genetic locus, only a phenotype. 2003) for restriction enzymes, DNA methyltransferases, hom-
ing endonucleases, and their genes should be used. The no-
“Homology” versus “similarity.” For use of terms that de- menclature for recombinant DNA molecules constructed in
scribe relationships between genes, consult the articles by vitro follows the nomenclature for insertions in general. DNA
Theissen (Nature 415:741, 2002) and Fitch (Trends Genet. 16: inserted into recombinant DNA molecules should be de-
227–231, 2000). “Homology” implies a relationship between scribed by using the gene symbols and conventions for the
genes that have a common evolutionary origin; partial homol- organism from which the DNA was obtained.
ogy is not recognized. When sequence comparisons are dis-
cussed, it is more appropriate to use the term “percent se- Tetracycline resistance determinants. The nomenclature
quence similarity” or “percent sequence identity,” as appropri- for tetracycline resistance determinants is based on the pro-
ate. posal of Levy et al. (Antimicrob. Agents Chemother. 43:1523–
1524, 1999). The style for such determinants is, e.g., Tet B; the
Strain designations. Do not use the genotype as a name space helps distinguish the determinant designation from that
(e.g., “subsequent use of leuC6 for transduction”). If a strain for phenotypes and proteins (TetB). The above-referenced ar-
designation has not been chosen, select an appropriate word ticle also gives the correct format for genes, proteins, and de-
combination (e.g., “another strain containing the leuC6 muta- terminants in this family
tion”).

Bacteriophages. The genetic nomenclature for phages dif-


fers from that for bacteria and tends to have separate conven-
tions for each phage. Genetic symbols may be one, two, or
three letters and are italicized. For example, a mutant strain of
␭ might be designated ␭ Aam11 int2 red114 cI857; this strain
carries mutations in genes cI, int, and red and an amber-sup-
pressible (am) mutation in gene A. Phenotypic symbols and
designations of gene products are not italicized (e.g., “the Spi
phenotype” or “Int protein”), and superscript plus and minus
symbols can be used to indicate wild-type and mutant pheno-
types, respectively. Host DNA insertions into phages should be
delineated by square brackets, and the genetic symbols and
designations for such inserted DNA should conform to those
used for the host genome. Lists of gene symbols for several
phages can be found in Genetic Maps, 6th ed. (S. J. O’Brien, ed.,
Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,
1993). Relevant references for some of the more widely studied
phages are as follows: for phage ␭, Daniels et al. (p. 469 –515, in
R. W. Hendrix, J. W. Roberts, F. W. Stahl, and R. A. Weisberg,
ed., Lambda II, Cold Spring Harbor Laboratory, Cold
Spring Harbor, NY, 1983); for phage T4, Kutter et al. (p.
491–519, in J. D. Karam, ed., Molecular Biology of Bacterio-
phage T4, American Society for Microbiology, Washington,
DC, 1994); and for phage T7, Dunn and Studier (J. Mol.
Biol. 166:477–535, 1983).

Transposable elements, plasmids, and restriction en-


zymes. Nomenclature of transposable elements (insertion se-
quences, transposons, and phage Mu, etc.) should follow the
recommendations of Campbell et al. (Gene 5:197–206, 1979),
with the modifications given in section vi of “Bacteria,” above.
The Internet site where insertion sequences of eubacteria and
archaea are described and new sequences can be recorded is
http://www-is.biotoul.fr/.
The system of designating transposon insertions at sites
where there are no known loci, e.g., zef-123::Tn5, has been
described by Chumley et al. (Genetics 91:639 – 655, 1979). The
nomenclature recommendations of Novick et al. (Bacteriol.
Rev. 40:168 –189, 1976) for plasmids and plasmid-specified ac-
tivities, of Low (Bacteriol. Rev. 36:587– 607, 1972) for F⬘ fac-

August 2014, Instructions to Authors Journal of Bacteriology jb.asm.org 17


n

100 CHAPTER 4· 0 GENES IN POPU-LATIONS 101

rates (see Chapter 7), while 95% of the mammalian Y chromosome does not alleles at a single locus make- enough difference to phenotype to have a
recombine. measurable effect on fitness.
An important example of linkage disequilibrium at the molecular level In most cases mutations will be deleterious and so lower fimess. These
occurs in the genes which make the inajor histocompatibility complex (MHC), mutations will be removed fairly quickly from the population by purifying or
a vital part of the vertebrate inunune system (the evolution of this gene complex negative selection. At other times a new mutation might have a higher füness
is discussed in detail in Chapter .7): Alleles at different MHC genes, called than ali others in the population and so is able to multiply. This is usually
haplotypes;· are óften inherited in combination; with . different haplotypes called positive selection in molecular evolution, and will lead to.the eventual
found in different·populations. For example, the alleles AL B8 and DR3 show fixation of the selectively favoured allele (see below).. For example, it may
significant.linkage disequilibriulTl in Caucasian populations. Unfortunately, this be that allele A has a higher fitness than. allele a. This m·eans .(assuming A is
haplotype has also been associated with a faster progression to AIDS in mv­ dominan! to a) that selection will favour the AA andAa genotypes over the aa
infected individuals. genotype. In terms of selective coefficients, we can think of this as:
Genotype AA Aa ªª
4.2.2 Natural selection Fitness 1 ¡ -s
Natural selectionis at the heart of the evolutionary process. Because of the On the other hand, if there is codorninance, so that the Aa het_erozygote l;J.as a
'struggle for existence' that faces organisms as they compete for resources, fitness intermediate to those of the two . homozygotes, then:
those with genes that better adapt them to their environment have a greater
probability of surviVing this struggle, so that their favourable genes áre
Genotyp� AA Aa ªª
Fitness l 1-s I -2s
preferentially passed on and will increase in frequency in the population.
Although natural selection is synonymous with the name of Charles Darwin, As well as Iooking at a single Iocus, it is also possible to think of natural
it was .R.A. Fisher who <lid much to show the power of this process at the selection acting on a phenotypic character which is under the control .of many
gen�tic leveL different loci, such as height or body weight. The genetics of these quantitative
Natural -selection can act in a variety of ways and at different points during characters is discussed more fully in section 4.3; 'fypically,: characters · of this
an otganism's life-cycle. For example, genotypes rnay differ in their viability son have a bell-shaped normal disnibuti.on of phenotypes, with most ·indivi.du.als
of producing ·adult organisms from zygotes. La ter in the life-cycle, individuals somewhere near the middle (the mean) of the distrib.ution,.and-decreasing
may·differ·in their·fecundity-the numbers of offspring they produce­ numbers ar more extreme values (represented by a variance). Positive selection
which:wil1 also lead to natural selection. However, before this takes place there on qua:f1:titative characters; more often referred to as directional selection,
may also be-sexual selection in which organisms differ in their mating success. will move the mean·of the distribution in one direction (-Fig. 4.3a).
·Finally, gametes may have different probabilities of achieving fertilisation Sorne of the best examples of positive sele'ction_ involve the eyolution..of
leading to a fom1 of gamétic selection. resistance to antibiotics, drugs and pesticides, such as that built up by mosquitoes
, The simplest way of thinking about whether one organism is better adapted to the chernical DDT once used to control them. A spectacular example of this,
than another is to-describe its fitness. In population genetic terms, fitness can and.one that has been of. great practica! importance in recent years, is- the
be défined as the ·capability-that any particular génotype has to survive and resistan.ce developed by lllV-1 to the drug AZ�. AZT inhibits viral replication
reproduce. This is usually expressed in relative terms, such as whether the by terminating transcription of the enzyme reverse transcriptase. In the absence
heterozygote Aa has a higher fitness than the homozygotes AA and aa. Fitness of AZT, the wild-type allele for reverse transcriptase was found to be between
is also specifi.c to each environment, because a genotype which is beneficia! in 0.4 and 2.3.% more fit than alleles adapte:d for dnig resistance (s = 0.004.-
one location might be deleterious in another (and vice versa). 0.023 ). Although the drug works extremely well for the first few inon�s, -and
In molecular evolution, fim•esses are most often expressed _in terms of the greatly reduces .the amount of circulating virns, the benefit is only shorHived
selection coefficient, · denoted · s, which is a measure of the reduction in and within six months or so the virus is able to evolve resistance through a
fitness comPared with the best genotype in the population: for instance, an s small number of mutations. The frequency of the wild-type allele then declines
of 0.01 rn:eansthat a genotype has a I % less chance ohurvival than the Qest rapidly. Current apprbaches to IllV therapy therefore involve combinat.ions
genotype-it is 99% as fit. Despite this very .simple notation, fitness is a of ·drugs because the virus has a much smaller chance of developing multiple
complex entity which may change through time, and is difficult to measure in resistance.
nature. Indeed, there ai'e very few • cases in which the differences between In other cases.natural s.election wilJ favour the hetero�ygote over either of
102 CHAPTER 4 GENES IN POPULATIONS 103

whkh takes exactly the samc fnrm as 1he equilíbrtum achieved under forward
(a)
and backward mutatiort pressure {eguatíon 4.7}.
Phenotypic
distributlon Directíonal The most ce[ebrated exampies of overdomlnance are the sickle-ce!l poJy•
in the _selec;tlon morphísm in human fi-haemogiobin and the alcohol dehydrogenase {Adh)
populatían
polymorphism in Drosophila (see Chapter 7). Overdominant selectíon is also
likely to be opcrating on the MHC beca use a heterozygote witb dlfíerent Jv1HC
molecules will recognise more parasites than a homozygote with only one
type of MHC molecule. The analogo1.1s process to overdominant selection for

(b)
, quanti1ative characters, in that genetk variafüm is actively mainudne<l, is
usually called stabilising selection and meaos that indlviduaJs in the mean
oí the dístributlon wm benefü over those at thc extremes (Fig. 4.3b).
St.i:bHí$lng The alternative to overdominance is where the hcterozygote has a hnver
selectíon Fig. 4.3 Differcnt types nf fii.ness rhan either hornozygote:
natural selectlon and how
they cffec1 the phenotypk Genotype AA Aa "ª
dl.stributicm of a dwracter Fitness 1 1-s
controllcd by many genes
(Le. a qunn:i!a:ive d aracter This is called underdominance and although homozygous genotypes are
with a nDnnal dis1rib111ion) favoured, it still preserves both the A anda allcles although this polymorphisrn
and fht' frequende o( alleles
will he unstable, wlth a change in aJlele frequency away from the equilibrium
iH a single lorus where each
Disruptive shaded elilpse represents a leading to the fixation of A or a, For quantitatíve cha.ractcrs an anafogous
se!ection population of individuals process is cafled disruptlve selection whereby individuais at the extremes
(cirdes:) with an allele of a of the distribution have a higher füness than thos.e with mean values (Fig.
pan.icu!ar colour-black ,,r 4.3c).
whitc. Thc open and shaded Underdominant selection appears to be controiling bill -size in the African
bells represen! the phe:notypic
dlsufüutiom before and after finch Pyrenestes ostrinus studie<l by Thomas Bates Smith. Bill sízes in this species
selection. respectively, and are either large or small. but not of intermedíate dimensions. The basis for this
the arrows signiiy the differem:e appears to be the hardness of the seeds eaten by the birds: duríng
dircct!on of sele.::tion. the reproductive season both the large• and srnaU-biHed morphs preíer soft
seeds but durlng the dry season, when food abundance is low, the farge-billed
morph tends to leed on hará seeds while the smail-biUed morph expands
its diet to indude other foods. No seeds of íntermediate hardness are found 1n
the homozygotes. This is caUed overdominance (sorne-times heterozygous
the Jocafüies where these birds líve so that selection acts agalnst those wlth
advantage) and wc can think of it in tenns of fitnesses as follows:
intermedíate bill sizes.
Gen-0type AA Aa ª" Polymorphísms can also be rnaintained in populations through frequency-
Fitness ¡-s 1 1- t dependent selection (Fig, 4.4). Thjs occurs when the fitness of a gcnotype
depends on its frequency in the population. Por example, the genotype at
where t is simply the selection coeffident for the aa genotype. Overdominant
the !owest frequency may have thc highest fitness {negative frequency-
selection wH! produce high heterozygosíty (Le. over that expected under tbe
dependent selectiún). Thís may occur tri host-parasite sys'tems where a
Hardy-Weinberg eguilihrium) and beca use it preserves both the p and q alleles
parasite at low frequency is not recognlsed hy the host immune system and is
will maintain genetic varíation in populations, giving rise to a balanced
therefore able to increase in frequency, whereas a strain at higher frequency
polymorphism. The eqtiilibrium allele frequencies' {i.e. fa) whkh reflect
may elicit a stronger hnmune response agalnst it and so be more easily deared.
Lhis bá.fance depcnd only on the relative fitncsses of the three genotypes, so
This wíll cause an oscillation in the frequcncy of different strains as they evade
1hat:
or are recognised by the immune system, leading to a polymorphism in thc
population. In other circumstances t.he genotype vvith the híghcst frcqucncy
(4.9) may be íavoured (positive frequency..<fependent s.eiection), whicb wiU
104 CHAPTER 4 GENES 1N POPULAT!ONS 105

allele} will be after a generation of this seiective process. We can denote this
aa new frcquency of p as p', and can cakulate it by:

il, p'=_P_2 (4. 10)

J,__
_________
1l l-sq
;.M The change in allele frequency (tlp} can then be simply computed as p' - por
O 1 directly by:
Frequencyof allele A
'
Fig. 4.4 AUele and genotype frequendes under frequency•dependcnt selecrlon. Genotypes Ap=...!E'L (4.11)
AA and ha are suhjttt to n tlve frequency-dependent sele<tion whereas the fitness oí Aa 1 - s q "'
L frequency-independent. To the rlght of the po\m where tbe lines cn) s. the A allele- is It is worth m1.1:strating this with a numera eiample. Let's assume p arid·q i
favourt"d and ilu:rea.,i¡es in irequency althoilgh the fitness oí thé AA genotype decreases, To
thc left of this pohlt, tbc fre{}uency Of the A·anetc declines aud the a1lelé is favoured. the _population are initially 0.25 and O. 75, respectively, and the sclection
Althoúgh the relationship be1weCil íimess and írequency Is shown here as a stratght line, coeffícient against aa is at the top end of what we observed in the case oí AZT
in reallty it can take on any shape. Adapted Jrom Rid.ley (1996). resistance, say 0.025. Applying equation 4.11 to get Ap we discover that-

spq 2 0.025 X 0,25 X (0.75)'


prevent polymorphisms from heing established. Ftequency dependent selecrton
may also ocrur if a population occupies a heterogenem.rn envíronment,. so that
1- sq2 = 1-0.025 X (0.75)'
dlfferent alleles are favoured in different patches (mkro-environments) or at 0.0035
different times. =0.9854
Unfortunately, detecting natural selection at the molecular leve! is a =0.0035
notoriously difficult thing to do because its effects can be subtle and becaus.c which means· that p V\'ill have íncreased fróm 0.25 to 0.2535. Although thís
different evoltHionary processes can leave similar signatUres in sequences (see seems Jike a tiny íncrease ít has happened in a single generation, and represents
section 4,.5 and Chapter 7). An added complkation is that the agem of selection spectacular progress compared with what happe-ns under mutatíon alone. lf
may. no longer· be presen:t in the population. For instance, the chemokine we run this process for long enough:rhen the A allele wilI eventually achleve
receptor 5 (CC 5) gene ts an importan t. factor in aUowíng HIV-1 to infect cells fixation. However, the rate at which this occurs is not constant, befng fastest
called macr,ophages. About 10% of Caucasian people have a 32-bp deletion when lhere are many heterozygotes in the population and slówest ·When
(called A32} which inactiva tes this gene and those homozygous for this deletion there are mostly homozygotes (plug in sorne different values for p and q and
(about 1% of individua)s) have a greatly reduced susceptibility to BIV infection, see for .yoursdf}. The same will aiso -be true lf thfie •is codominance" Jn
whil thosc that an; heterózygous may become HIV infected but seem to which case the change in allele freqUencies takes: on ai1 S shaped· (s.ymmetrkal)
progress more slowly to AIDS. Whatis puzzling is that this mutation is at such form (Fig, 45). In su:m, we can see that natural selection operares most
hi.gh frequency in Caucasians {see Fig. 4.13) but; is very rare in African and efficiently when there are large amo-unts of genetic variation for it to Work
Asían populations, Because HJV is so recentan invader it cannot be the selective with, Furthermore, the efíidency o( natura} selection is also detennined by
agent responsible for the high frequency of this mntation, which raises the the size of the population, working best when it is large, When popuiation
posslbility that a past infectious disease may have acted as a strong selectíve
force favoming individuals 1-'Vith this deletion.
Despite chis complexity, it is still possible to show how natural selection
changes g ne frequendes \"ilith some simple mathematks. Taking the simplest Fig. 4.5· The change in al!ele
example, of positive selection favouring AA and Aa over aa, so that: frequendes under posltive
natural sele.:tion for a
Genotype AA Aa ªª oodominant advamageoll5
Fitness l l 1-,. A ailek that has entered
a population of na
it ís easy to see that frequenCf of the aa genotyJ}e, or q2 if we rccall our Hardy- homwygotes.. Frmn Griffiths
Weinberg fo1mula, will be. reduced by 1 - s in a given generatton, The next eta!. (1993), wilhpemtlsslon Number.of generatlom. - - - ►
thing we would like to do is calculate what p {the frequency of lhe favoured írom WH Freeman.
106 CHAPTER 4 GENES IN POPULATI'ONS 107

sizes are small, mutations are more under the control oI chance lhOcesses.
This is the subject of the nexr section.
límeto
füation
4.2.3 Genetic drift
Once a mutation has arisen in a popu1ation it can experlence one of two
evolutionary fates: it cat1 be fixed and so come to domínate the population, or •A!lelernpy
dies out
ít can be ]ost. \Vhích outcome a new allele faces ls not ahvays down to how because it
much betrer or worse it is compared to those alleles already present in the faih to
population. Instead it may simp-ly be down to dlance. For example, sorne reproduce
ali eles may reside in individuals who leave no offspring and even if an ir1dívidual
does produce offsprlng only a rnndom sample of their genes. those present in
the successfui egg or sperm, will be inherited. Because of bad luck, most gametes
wil.l not make it to the next generation. This means that alleles are in effect
randornly s.ampled in,every generation, and tbis random sampHng can change
Flg. 4.6 Fixation oI an allele
altete frequendes. This is caHed genetíc ddft. (bfack circle) by genclíc drift.
To understand how allele frequendes change wlth genetk drift we need to The shaded cltipse represems
devefop a stochastic model of evolution, fn which chance plays the leadjng a population observcd at
role. Thís differs from our prevíous discussions of mutation and selection wbere difiermt time points, The
we dealt with determinist:k models, which ignored random effects, First othcr ailde in the poJJUlatlon
(individuals. with whit¡;
imagine a diploid population of size N, so that there are a total of 2N allelic
drdes) dies out becaose ft
copícs of cach gene. Because of the random sampling of gamctes there is a fatls to reproduce. On average
chance that some of these alJeles will contrtbute no copies of themselves to it takes an allí'lí' 4iV Odg!nof
thc ncxt generation, whilst others will colltribute many. Sorne a.Beles wlll Henerations t) gí't to fixation. afte!e
.at1hough the probabUity of (generation 1)
therefore be Iost from the population ín each generation because the gamctes
carrying them have not been passed on. This will cause aUele frequendes to this happcniug is oµly l/2N.
fh.1ctuate slightly from generation to generation as sorne alleles are lost and
others reproduce. Ifthis stochastk variation in reproductive sucress continues advantage, most of whkh 'WiH also be lost by chance unless their benefü is
for long eno11gh tl1ere wilI eventually come a time when all 2N alleles wmbe substantiaL For example, R,A. Fisher cakulated that a mutant with a l %
descended from a slngie of our odginaI 2N alleles because all others have failed selecüve advantage only has about a '2% chance of being fixed (So a 9íP'lo
to reproduce at some stage along the way (Fig. 4.6}. This aHeie wíll then have chance of being Jost). Loss by genetic drift is espedally Bkely soon after thc
hecn ffxed by the process oí genetic drift alone, The probability oí fixation of mutation has appeared and so is still at low frequency.
an alleJe by random gcnetk drift is simply 1/(2N}, which is its frequency in tbe Usins a matbematical modcl it has been shown that H takes an average of
Jlopulation after it has arisen by mutation. Thls also means rhat the probability 4N generations for a neutral altele to get to fixation through genetic driit,
of fixatíon is greater when the population slze is small. because l/2N is larger, aJrhough the random naturc of the process rneans that tbere is a large vatiance
Not only will genetic drift work better in small populations, bul it will also a.round thh time. Mutations with a selective advantage will bC fixC'd qukker
be most important for those mmations which have no effect on phenotype, rhan neutral alleles (that is, in less than 4N generations) because indíviduals
and so are neithe advar.t..1geous nordeleterious compared to theirpredecessors. with the better gene "vlll produce more offspring than those with other alleles,
These mútations, whlch are free from the rigours of natural selecfüm, are Conversely, when natural selection is actively maintaining a polymorphism,
called neutral mutations and their evolution is díscussed in detaíl in Chapter .alleles can coexist in the popuíarJon wilhout any going to fixation far as long
7. Beca use an individual carrying a neutral rnutation will not beata selective as the selection p:ressure exists, so that they may survive for Ionger than 4N
advantage, the only way they can he fixed is through the chance action generations.
of genetic drift (tbe one exception-genetic hitchhiking-is discussed in During the time it takes for an allele ro be Jixed by genetic drift, thc
Chapter 7). However, genetíc drift wiH aiso affect new mutations wíth a selective population will exhibit polymorphísm at the locus in question. Obviously, the
FEMS Microbiology Reviews 26 (2002) 355^374
www.fems-microbiology.org

The bacterial replication initiator DnaA. DnaA and oriC,


the bacterial mode to initiate DNA replication

Walter Messer
Max-Planck-Institut fu«r molekulare Genetik, Ihnestrasse 73, D-14195 Berlin-Dahlem, Germany

Received 2 June 2002; received in revised form 1 July 2002; accepted 2 July 2002

First published online 3 October 2002

Abstract

The initiation of replication is the central event in the bacterial cell cycle. Cells control the rate of DNA synthesis by modulating the
frequency with which new chains are initiated, like all macromolecular synthesis. The end of the replication cycle provides a checkpoint
that must be executed for cell division to occur. This review summarizes recent insight into the biochemistry, genetics and control of the
initiation of replication in bacteria, and the central role of the initiator protein DnaA.
, 2002 Federation of European Microbiological Societies. Published by Elsevier Science B.V. All rights reserved.

Keywords : Protein domain ; DnaB helicase ; ATP-binding DnaA box; Plasmid

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 356
2. The di¡erent functions of DnaA protein, an overview . . . . . . . . . . . . . . . . . . . . . . . . . . . 356
3. Steps in the initiation of E. coli DNA replication . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 356
4. Domains of DnaA and the homology between DnaA proteins . . . . . . . . . . . . . . . . . . . . . 357
5. Domain functions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 358
6. The role of ATP binding on DnaA structure and function . . . . . . . . . . . . . . . . . . . . . . . . 359
7. DnaA and membranes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 360
8. DnaA boxes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 360
9. DnaA oligomerization and cooperativity. Rules for DnaA binding . . . . . . . . . . . . . . . . . . 360
10. DnaA-mediated origin unwinding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 361
11. DnaA-mediated helicase loading: the DnaA primosome . . . . . . . . . . . . . . . . . . . . . . . . . . 362
12. Variations of the theme: DnaA and oriC in other bacteria . . . . . . . . . . . . . . . . . . . . . . . . 362
12.1. B. subtilis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 362
12.2. Streptomyces . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 363
12.3. T. thermophilus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 364
12.4. H. pylori . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 364
12.5. Synechocystis sp. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 364
12.6. Caulobacter crescentus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 364
13. DnaA as a transcription factor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 365
14. Joint action of DnaA and plasmid initiators in plasmid replication . . . . . . . . . . . . . . . . . . 365
14.1. P1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 365
14.2. F . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 365
14.3. RK2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 365
14.4. R6K . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 366
14.5. R1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 366
14.6. pSC101 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 366

* Tel. : +49 (30) 8173788; Fax: +49 (30) 8413 1385. E-mail address: messer@molgen.mpg.de (W. Messer).

0168-6445 / 02 / $22.00 , 2002 Federation of European Microbiological Societies. Published by Elsevier Science B.V. All rights reserved.
PII: S 0 1 6 8 - 6 4 4 5 ( 0 2 ) 0 0 1 2 7 - 4

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


356 W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374

15. The special case of DnaA and V plasmids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 366


16. How to limit initiation to once per generation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 367
16.1. Sequestration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 367
16.2. Titration of DnaA to the datA locus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 367
17. DnaA and oriC in the cell cycle, control of initiation . . . . . . . . . . . . . . . . . . . . . . . . . . . . 367
18. Perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 368
19. Note added in proof . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 368

Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 368

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 368

1. Introduction E. coli. Especially the elegant work over many years from
the Kornberg laboratory using the DnaA- and oriC-de-
The initiator protein DnaA is found in all eubacteria pendent in vitro replication system is the basis of our
analyzed so far. The name derives from the fact that understanding [10].
dnaA was the ¢rst mutant isolated that is a¡ected in oriC of E. coli contains in 260 bp ¢ve DnaA boxes, and
DNA replication. In fact, mutants from this series were at its left end an AT-rich region consisting of three 13-mer
the ¢rst temperature-sensitive mutants in bacteria and es- repeats and the so-called AT cluster (see Fig. 1). In addi-
tablished the use of this type of conditional lethal muta- tion, there are binding sites for accessory proteins like IHF
tions for cellular functions [1,2]. The replicon hypothesis and FIS, and control factors like IciA, Rob, H-NS whose
by Jacob et al. [3] postulates two basic elements for the precise function is not known (see [5] and references there-
initiation of replication : the initiator, a trans-acting sub- in). The importance of 11 GATC sites, recognition sequen-
stance, DnaA protein, and the cis-acting replicator, which ces for the Dam methyltransferase, will be discussed in
we now call replication origin, oriC. Most bacteria possess Section 16.
a unique replication origin on the usually circular chromo- DnaA protein binds to its ¢ve binding sites in oriC as a
some. Several aspects of the initiation of bacterial replica- monomer, introducing a 40‡ bend at each site [11]. Only
tion are covered in a number of recent reviews [4^7]. DnaA complexed with ATP is active in initiation [12].
ATP-DnaA binds to additional sites, 6-mer ATP-DnaA
boxes with the sequence 5P-AGATCT or a close match
2. The di¡erent functions of DnaA protein, an overview [13,14]. These sites are predominant in the AT-rich region
which is unwound, and subsequently stabilized due to spe-
All functions of DnaA protein depend on its ability to ci¢c binding of ATP-DnaA to single-stranded ATP-DnaA
bind speci¢cally to an asymmetric 9-bp recognition se- boxes. In this sequence of interactions binding to 9-mer
quence, the DnaA box: 5P-TTATNCACA. A replication DnaA boxes in oriC is a high-a⁄nity interaction (KD =
origin, oriC, usually consists of an array of several DnaA 1 nM), binding to 6-mer ATP-DnaA boxes in double-
boxes. Binding of DnaA to such an array, origin recogni- stranded DNA is a low-a⁄nity interaction that requires
tion, is the ¢rst step in the assembly of a specialized nu- cooperativity among the proteins, and binding to 6-mer
cleoprotein complex [8], the initiation complex. Structural ATP-DnaA boxes in single-stranded DNA is again a
distortions of the DNA within such a complex are the high-a⁄nity interaction [14]. A threshold level of DnaA
prerequisite for the second function of DnaA in the ini- protein is required for the conversion of the initial to the
tiation process. It acts as a DnaA primosome. Protein^ open complex. It is presumably because of these graded
protein interaction results in the loading of the replicative a⁄nities and the cooperativity between the proteins that a
helicase, DnaB in the case of Escherichia coli. precise timing of initiation in the cell cycle is possible. The
DnaA protein is also a transcription factor (for a review net result is unwinding of the AT-rich origin region [15^
see [9]). Binding to one or two DnaA boxes in a promoter 17]. FIS protein has a negative e¡ect on the reaction
region may result in repression. Most importantly, the [18,19], HU and IHF proteins [20,21], a high ATP concen-
dnaA gene itself is repressed by DnaA, it is autoregulated. tration ( s 2 mM), high temperature (38‡C) [22], and tran-
Other promoters are activated by DnaA binding. If DnaA scriptional activation enhance unwinding [21,23^25]. The
boxes are within a transcription unit, DnaA binding may replicatively active complex contains 20^30 DnaA mono-
result in transcription termination. mers, as determined by electron microscopy [26,27]. Direct
measurement by surface plasmon resonance gives a stoi-
chiometry of 18 DnaA monomers per oriC in the initial
3. Steps in the initiation of E. coli DNA replication complex [7]. The open complex with six more binding sites
in the single-stranded AT-rich region thus must contain 24
Most of our knowledge on DnaA and oriC is based on DnaA monomers, corroborating the electron microscopic

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374 357

Fig. 1. Schematic replication origin of E. coli.

results. However, lower stoichiometries have also been re- However, the high number of proteins to compare and
ported, and the in£uence of binding conditions on DnaA the possibility to relate the predicted structure to experi-
stoichiometries has been emphasized [28]. mentally determined structures of other members of this
The unwound region spans 28 bp without and 44^46 bp protein family, the AAAþ family, make this prediction
with SSB present [17]. Since single-stranded DNA covered pretty reliable. The AAAþ class of ATPases is a large
with SSB is a poor substrate for DnaB helicase, it has to family of proteins with common sequence motifs. Many
be loaded with the help of DnaA. Two double hexamers of of them are involved in the initiation of replication and
DnaB and the helicase loader DnaC, one double hexamer other functions of DNA metabolism throughout all king-
for each replication direction, are positioned by DnaA doms with a presumptive role in remodeling and loading
into the loop [28,29]. DnaC leaves the complex immedi- of proteins. DnaA and also DnaC are members of this
ately after or during loading, accompanied by ATP hydro- family [41].
lysis. This activates the helicase activity of DnaB [30]. The Grouping of DnaA sequences from eubacteria on the
two DnaB hexamers slide past each other in 5P-3P direc- basis of similarity reproduced closely the phylogenetic re-
tion, and expand the bubble to about 65 nucleotides [29]. lations. DnaA domains are shown in Fig. 3. For domain 1
Now primase can enter the complex and synthesize two (86 residues) a consensus sequence is not very pronounced,
leading strand primers. Synthesis of opposed and overlap- except for a few patches of conserved residues. Domain 2
ping leading strands has been postulated before in order to is highly variable in sequence and length, between 6 and
ensure complete synthesis of oriC [31]. 247 (Streptomyces) amino acids. Therefore we assume it to
The sliding clamp, a ring-shaped dimer of the L-subunit form a £exible region. The C-terminal two-thirds are quite
of DNA polymerase III, is loaded onto each primed tem- similar and make it possible to derive a reasonable con-
plate by the clamp loader, the polymerase III Q complex sensus sequence. Domain 3 contains an ATP-binding site,
[32]. This activates the intrinsic ATPase activity of DnaA i.e. Walker A and B motifs, and other motifs found in the
[33,34], in cooperation with a protein with sequence ho- AAAþ protein family. Domain 4 is also well conserved
mology to DnaA, Hda [35]. Inactivation of DnaA by ATP and lacks homologues in the archaea as well as in the
hydrolysis prevents further initiations. The E. coli initia- eukarya. It contains the DnaA signature, a de¢nition
tion cycle is shown schematically in Fig. 2. used in the SWISS PROSITE online search facility,
which has been updated on the basis of the now known
104 DnaA sequences: [STPQ]-x(3)-[IL]-[GA]-x(2)-[FLIM]-
4. Domains of DnaA and the homology between DnaA x(1,2)-[RK]-[DTSNER]-[HA]-[TSPA]-[TSVA]-[VILM] (C.
proteins Weigel and W. Messer, http://www.molgen.mpg.de/
Vmesser/).
Sequence homology based on four species [36,37] and Secondary structure prediction was done by the PHD
later on 30 species [6] made it possible to subdivide DnaA server (http://www.embl-heidelberg.de/predictprotein/pre-
protein into four domains. These domains were later dictprotein.html) [42]. Results were virtually identical for
shown to correspond to functional domains, with minor di¡erent prediction methods. The secondary structure pre-
correction of their borders [38,39]. The functional studies diction showed a much higher conservation for the struc-
were inspired by earlier attempts to combine sequence tural elements than for primary sequence. It is shown in
comparisons with secondary structure predictions [40]. Fig. 3. Domain 3 shows an [KL]5 Rossman fold-type ATP-
These results were recently corroborated and extended ase motif [43] suggesting a tight structure which aligns
by a thorough study that compares 104 di¡erent DnaA perfectly with the known structure of polymerase IIINP
proteins (C. Weigel and W. Messer, http://www.molgen. [44]. This illustrates the reliability of the prediction. The
mpg.de/Vmesser/). Despite considerable e¡ort no three- prediction for the DNA-binding domain 4 shows four
dimensional structure of a DnaA protein is available. K-helices, the C-terminal long one is divided in two in

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


358 W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374

some DnaA proteins. K-Helices 12 and 13 (Fig. 3) with a


basic loop in between were suggested to represent the
DNA-binding motif [45].

5. Domain functions

The N-terminal domain 1 mediates protein^protein in-


teractions, DnaA oligomerization and interaction with
DnaB protein. Several techniques were used to demon-
strate oligomerization. Domains 1 of E. coli [46] and of
Streptomyces lividans [47] were able to functionally replace
the oligomerization domain of VcI repressor. Physical in-
teraction of isolated domain 1 was demonstrated with a
solid phase binding assay in vitro, showing that dimeriza-
tion via domain 1 does not require other parts of DnaA
protein [46]. Cooperative binding of DnaA to two adja-
cent DnaA boxes depended on domain 1 [48]. Expression
of isolated domain 1 was inhibitory to initiation, as mea-
sured by suppression of the overinitiation phenotype of
dnaA219 (see Section 6) [46], showing that dimerization
via domain 1 is an essential step in initiation in vivo.
The larger part of domain 1, amino acids 24^86, is also
involved in the interaction of DnaA with DnaB, as shown
in vitro with a solid phase binding assay [49]. In in vivo
experiments domain 1, or amino acids 24^86, was required
for the loading of DnaB [49,50]. There is a second site for
DnaA^DnaB interaction in the N-terminal region of do-
main 3 (see below). In summary, physical interaction be-
tween DnaA and DnaB requires amino acids 24^86 of
DnaA, whereas DnaA dimerization requires the complete
domain 1, amino acids 1^86 ; in later experiments amino
acids 1^77 were found to be su⁄cient. Surprisingly, do-
mains 1 and 2 were found not to be required for DNA
unwinding of the AT-rich region in vitro [50]. However, it
has also been reported that mutants with altered residues
26 or 40 within domain 1 were unable to unwind the AT-
rich region [51]. Additional experiments are required to
clarify this point.
Domain 2 is very di¡erent in size in di¡erent bacteria, as
mentioned above. In E. coli DnaA all of domain 2, amino
acids 87^134, can be deleted without loss of function. A
spontaneous mutant, dnaA216, ‘DnaA light’ lacking resi-
dues 87^104, was functionally indistinguishable from wild-
type [38]. However, long domains 2, e.g. in Streptomyces,
have been conserved in evolution with respect to length
not sequence. Therefore the length of domain 2 may well
have an important function in certain conditions.
Domain 3 is organized as an open twisted KL-structure
[11] and contains the ATP-binding region of AAAþ ATP-
ase-type proteins, a P-loop or Walker A motif [41,43,52^
54]. The presence of an ATPase is shown by the Walker B
motif as well as by the isolation of mutants a¡ected in
ATPase activity, E204Q and R334H [55,56]. The impor-
Fig. 2. The initiation cycle of E. coli. tance of ATP binding and hydrolysis will be discussed in
Section 6.

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374 359

Fig. 3. Secondary structure prediction for E. coli DnaA protein with sequence motifs and interaction domains.

In the N-terminal part of domain 3 is a region, amino 6. The role of ATP binding on DnaA structure and function
acids 130^148, that comprises a second interaction site
with DnaB [49,50,57,58]. DnaA thus contains two sites Binding of ATP is required for the unwinding of the
for interaction with DnaB, amino acids 24^86, interacting AT-rich region by DnaA [12]. In fact, this is the only
with DnaB amino acids 154^210 (C-terminal L Q~ fragment reaction that requires ATP-complexed DnaA, since heli-
[59]), and DnaA amino acids 130^148 interacting with the case loading occurs in mutants that are de¢cient in ATP
DnaB N-terminus (K fragment) [49]. binding [49]. ATP promotes an allosteric modi¢cation and
Domain 3 also contains a second region that promotes does not provide energy for the unwinding reaction since
oligomerization. This has been shown for Streptomyces non-hydrolyzable ATP analogues are equally e¡ective [12].
DnaA by kinetic analysis [48] and by gel retardation Several of the ‘classical’ dnaA mutants, e.g. dnaA5 and
that even showed the existence of mixed dimers containing dnaA46, carry a mutation A184V close to the ATP-bind-
full-length and truncated DnaA [47]. Likewise, in E. coli ing site in the Walker A motif [65]. These mutants are
DnaA the presence of a region from the C-terminal part of temperature-sensitive and do not bind ATP or ADP at
domain 3 gave a strongly reduced dissociation rate in sur- any temperature [66^68], but can be activated to ATP
face plasmon resonance analysis, suggesting cooperativity binding by DnaK and GrpE proteins [68]. All A184V
between monomers that did not contain domain 1 [7]. This mutants carry secondary mutations in dnaA [65,69]. How-
same region is conserved in the NtrC family of transcrip- ever, when the A184V mutation is separated from the sec-
tion factors [45]. ondary mutations, it confers similar temperature sensitiv-
The DNA-binding domain 4 is represented by the 94 ity [68]. When A184V is present on multicopy plasmids,
C-terminal amino acids [45]. The DNA-binding motif such plasmids confer cold sensitivity to dnaAþ or dnaA46
probably consists of K-helices 12 and 13 (see Fig. 3) with hosts [65,68,70]. Intragenic suppressors of dnaA46 have
a basic loop between the helices. In fact, mutation of some been isolated, dnaAcos [71] which carries two additional
basic amino acids in the loop abolishes DNA binding mutations (Q156L and Y271H) [69], and dnaA219 with a
[60,61]. A thorough mutational analysis of this domain R342C mutation [46]. Both suppressor strains are cold-
revealed that mutations with impaired binding were found sensitive due to DNA overinitiation at 30‡C [46,71]. The
throughout domain 4 [60]. Mutations that a¡ect sequence dnaA219-carrying strain has been successfully used to
speci¢city are clustered in the beginning of helix 15 [60,62]. monitor for conditions that are detrimental to initiation
Suppressors of temperature-sensitive dnaX mutants [63] and thereby relieve the cold sensitivity and provide a pos-
also map to this speci¢city region. Binding of ATP to its itive selection for impaired initiation [46,49].
site in domain 3 modi¢es the sequence speci¢city of DnaA The active structure of ATP-complexed DnaA is sensed
protein [13] (see Section 9). In concert with this property, by a region close to or in DNA-binding domain 4, since
bound ATP is located close to the DNA-binding domain ATP-DnaA acquires an additional sequence speci¢city.
in the three-dimensional structure, as found by cross-link- Mutations that a¡ect ATP hydrolysis were found just up-
ing the Q-phosphate of ATP to Lys-415 in domain 4 [64]. stream of domain 4, R334H [56], as well as at a position in

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


360 W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374

domain 3, E204Q, that is involved in hydrolysis in similar was found to be indispensable for membrane-mediated
ATPases [55]. In addition, physical proximity between release of nucleotides [92], and a peptide between amino
ATP bound in the P-loop to the DNA-binding region acids 373^381, adjacent to K-helix 11 on the C-terminal
was demonstrated by crosslinking the Q-phosphate posi- side, was found to bind to phospholipids [92]. So far, it is
tion of ATP with K415 in domain 4 (see Section 5) [64]. not known whether the physiological relevance for DnaA^
The intrinsic ATPase of DnaA is weak. As outlined in membrane interaction lies in DnaA-mediated binding of
Section 3 it can be stimulated and DnaA thereby inacti- the initiation complex to the membrane or in the ATP-
vated. Since DnaA protein with the A184V mutation does ADP ‘rejuvenation’ reaction.
not bind ATP (or ADP) we must assume that the sup-
pressing mutations confer an active conformation onto
the protein without nucleotide binding. Of course, such a 8. DnaA boxes
protein cannot be inactivated by ATP hydrolysis, which
may explain the phenotype of overinitiation [72,73]. ATP-DnaA and ADP-DnaA bind with the same a⁄nity
The intrinsic ATPase activity of DnaA is stimulated at (KD between 0.6 and 50 nM) to an asymmetric 9-mer
the end of the initiation cycle. This reaction is promoted consensus sequence, the DnaA box 5P-TTA /T TNCACA,
by the loading of the sliding clamp to single-stranded as measured by gel retardation of oligonucleotides carry-
DNA, the dimeric L-subunit of DNA polymerase III ho- ing a single DnaA box [11] and by surface plasmon reso-
loenzyme [33]. This reaction requires Hda as cofactor [35] nance [60]. UDG footprinting shows that within this se-
(see Section 3). Therefore, hda mutants have an overinitia- quence T2, T4, T7P, and T9P are of particular importance.
tion phenotype similar to dnaAcos or dnaA219. The over- In this technique speci¢c T residues are replaced by dU,
all ratio of ADP to ATP in exponentially growing cultures and their accessibility to uracil-DNA glycosylase is deter-
was 4:1. In synchronized cultures ATP-DnaA increased mined with and without DnaA [93]. Previously a more
prior to initiation and was hydrolyzed to ADP-DnaA dur- relaxed consensus sequence which allowed 1^2-bp devia-
ing initiation [74]. This also results in a £uctuation of tion from this stringent consensus sequence was found
DnaA synthesis in the cell cycle since ATP-DnaA is the using DNase I footprinting or retention on nitrocellulose
active repressor for the dnaA promoter [13]. The cycling ¢lters [94^97]. When the ability of DnaA-DNA box com-
between ATP- and ADP-bound states of initiation pro- plexes to block transcribing RNA polymerase was used
teins is apparently a ubiquitous regulatory principle [75]. to de¢ne DnaA boxes an even more relaxed consensus
It has been described that DnaA at low Mg2þ or nucle- sequence was found: 5P-(T,C)(T,C)(A,T,C)T(A,C)C(A,G)
otide-free DnaA protein binds DNA unspeci¢cally (A,C,T)(A,C) [98]. The reason for the apparent discrep-
[28,76,77]. However, this is an artifact of the Sekimizu ancy are the rules for binding of E. coli DnaA protein
protocol of DnaA preparation [78] which involves a dena- described below. A monomer of E. coli DnaA binds
turation step. Native DnaA protein is always complexed only to the stringent DnaA box. Relaxed consensus se-
with ATP or ADP. quences require the cooperation of two monomers, and
hence two DnaA boxes [7,13,99].

7. DnaA and membranes


9. DnaA oligomerization and cooperativity. Rules for DnaA
A large part of the DnaA protein in E. coli cells is found binding
in a membrane fraction [78,79]. An E. coli mutant, pgsA,
with a limited ability to synthesize acidic phospholipids The dnaA promoter region was analyzed by DNase I
leads to arrest of cell growth [80]. This can be suppressed footprinting and surface plasmon resonance for the modes
by overproduction of DnaA proteins with mutations in of DnaA binding. The surprising observation was that
the membrane interaction domain described below [81]. ATP-complexed DnaA, but not ADP-DnaA, has a second
Also, some dnaA mutants have an altered cell membrane sequence speci¢city for a 6-mer consensus sequence, the
permeability [82]. There are thus a number of indications ATP-DnaA box [13]. This is also true for the dnaA pro-
that DnaA^membrane interaction is physiologically rele- moter region of Vibrio harveyi [100]. DnaA binding to
vant. ‘non-canonical’ sites was observed before with V DNA
Acidic phospholipids decrease the a⁄nity of DnaA for [101]. A summary of these sites and the ATP-DnaA box
ATP or ADP, and therefore enhance the exchange of these consensus sequence is given in Table 1.
nucleotides and promote a ‘rejuvenation’ of inactive ADP- The rules for DnaA binding as derived from the dnaA
DnaA [83^86]. Phospholipids also decrease the a⁄nity of promoter region are as follows :
DnaA for oriC sequences [87]. The region of DnaA that 1. E. coli DnaA, both ATP-DnaA and ADP-DnaA, binds
interacts with membrane was de¢ned by mutations in to a single DnaA box as a monomer. But this box must
K-helices 10 and 11 (see Fig. 3), amino acids 327^344 conform to the stringent consensus sequence 5P-TTA /
and 357^374 [88^91]. K-helix 11 (amino acids 357^374) T TNCACA, henceforth called a ‘strong’ box.

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374 361

2. Both forms do not bind to single ‘weak’ boxes, i.e. similar but not identical, and therefore show the variabil-
DnaA boxes with one or two mismatches to the con- ity of the binding reaction, see also Section 12. Bacillus
sensus sequence, unless cooperation occurs with a sec- subtilis DnaA apparently follows similar rules, since a hy-
ond DnaA protein bound to a DnaA box close by. brid origin with the main part of oriC from E. coli and the
3. ATP-DnaA recognizes in addition 6-mer ATP-DnaA AT-rich region from B. subtilis can execute most initiation
boxes with a consensus sequence that corresponds to reactions, primarily it can be unwound [103]. DnaA from
BglII recognition sites. These constitute weak boxes, S. lividans has the same two interaction domains with
and therefore require an adjacent strong box for similar kinetic consequences [47,61,104]. However, oligo-
DnaA binding. merization via domain 3 does not require binding to a
4. ATP-DnaA also binds to single-stranded ATP-DnaA second DnaA box as in the case of E. coli, since mixed
boxes. This was observed for the unwound region in oligomers made out of full-length and truncated DnaA
E. coli oriC [14] and for a substrate with a double- proteins, respectively, can bind to a single DnaA box
stranded strong box and adjacent single-stranded [47]. Streptomyces species have a high G/C content in their
DNA with an ATP-DnaA box [102,234]. In the latter DNA, and consequently the consensus sequence of the
case the strong box was required for binding to the Streptomyces DnaA box has a G or C at position 3
single-stranded ATP-DnaA box, in oriC ATP-DnaA [105,106]. Although this is a strong box, binding a⁄nity
could bind to single-stranded ATP-DnaA boxes with- to the E. coli consensus sequence is still better, and
out the help of a strong box, presumably because of the although binding to a single box of this kind is possible,
high number of such boxes in oriC. Single-stranded binding to two boxes is preferred [48]. It is not known
ATP-DnaA boxes have an intermediate a⁄nity whether the ATP-complexed form of Streptomyces DnaA
(KD = 40 nM); strong double-stranded boxes have a recognizes a 6-mer box.
KD of about 1 nM, and weak double-stranded boxes, Thermus thermophilus is also a G/C-rich organism.
including ATP-DnaA boxes, have a KD of around However, contrary to Streptomyces, the DnaA boxes in
400 nM [14]. the Thermus origin have the stringent E. coli consensus
As discussed in Section 5, there are two regions respon- sequence. However, single DnaA boxes do not bind
sible for oligomerization of DnaA monomers, domain 1, T. thermophilus DnaA (approx. KD s 1500 nM), and
amino acids 1^77, and a region in domain 3. This second only cooperativity allows reasonable binding with an ap-
oligomerization region was localized in the C-terminal parent KD of 30^60 nM. This cooperativity requires ATP-
part of domain 3 between amino acids 334 and 373 [7]. DnaA, the a⁄nity of T. thermophilus oriC to ADP-DnaA
The domain 1 oligomerization domain can operate over a is about 10 times lower [107].
distance of at least 150 bp [46,49], and does not even
require DnaA protein to be bound to DNA, the domain
3 oligomerization region requires a precise spacing of ad- 10. DnaA-mediated origin unwinding
jacent DnaA boxes [7].
Rules for binding of DnaAs from other bacteria are The unwinding of the AT-rich region is the crucial step
in the initiation of most origins, prokaryotic and eukary-
otic ones. In the case of E. coli, the AT-rich region con-
Table 1 tains six ATP-DnaA boxes adjacent to the 9-mer DnaA
E. coli ATP-DnaA boxes box R1 (Fig. 4). The sequential binding of ATP-DnaA to
DNA background ATP-DnaA boxes these sites has been determined using DNase I footprinting
and surface plasmon resonance [14]. Initial binding is to
E. coli dnaAp [13] AGAACT
AGATCT DnaA box R1. This then serves as an anchor for the
AGTTTA cooperative binding of ATP-DnaA to the double-stranded
AGATTT ATP-DnaA boxes. As outlined above, these boxes pro-
E. coli oriC [14] AGATCT mote low-a⁄nity binding. Therefore cooperative interac-
AGATCT
tion is required. DnaA bends the DNA by 40‡ upon bind-
AGATCA
AGGATC ing [11]. The topological stress in this complex unwinds
V [101] AGAACT the DNA in the AT-rich region. ATP-DnaA then binds to
AGATCC the single-stranded ATP-DnaA boxes, providing an initial
AGTCAT stabilization of the single-stranded state. This is again a
AGTATT
high-a⁄nity interaction. It seems to be a general feature in
V. harveyi dnaAp [100] TGATCG
AGATCG the assembly of protein complexes at speci¢c sites and at
AGACTG de¢ned times that a sequence of high-low-high a⁄nity is
AGATCT followed. Cooperative binding to the double-stranded AT-
AGATCA rich region is therefore presumably the limiting step in the
Consensus AGatct
initiation reaction, followed by unwinding [14].

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


362 W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374

Initially 28 bp are unwound by DnaA protein alone,


and 44^52 when SSB is also present. This is true both
for E. coli and for B. subtilis oriC [17]. A hybrid origin
with the main part from oriC of E. coli and the AT-rich
region from B. subtilis could be unwound by E. coli DnaA,
and helicase was loaded into the bubble [103]. Therefore
we assume that the initiation process and the action of
DnaA are similar for E. coli and for B. subtilis. We also
conclude that the species speci¢city of the DnaA^oriC
interaction lies in the main part of oriC with its ¢ve
9-mer DnaA boxes.

11. DnaA-mediated helicase loading: the DnaA primosome


Fig. 4. Mechanism of unwinding of the AT-rich region. ATP-DnaA is
Two double hexamers of the replicative helicase DnaB shown in red, ATP-DnaA boxes are shown in blue, the high-a⁄nity box
R1 in magenta.
and the helicase loader protein DnaC, one for each repli-
cation direction, must be loaded into the unwound region.
Since single-stranded DNA covered with SSB is not a between the 3P-end of the DnaA box and the potentially
substrate for DnaB, helicase must be positioned by single-stranded DNA is similar in most cases. Such a
DnaA. As outlined in Section 5, DnaA and DnaB interact structure is found in several bacterial origins, e.g. B. sub-
physically. The DnaA N-terminus (amino acids 24^86) tilis [17,112], Pseudomonas putida and aeruginosa [113,114],
interacts with the DnaB LQ fragment (amino acids 154^ Coxiella burnetti [115], T. thermophilus [107], and others.
210), and the DnaB N-terminus (K fragment) with domain This also holds for some plasmid origins like P1 [116] or
3 of DnaA (amino acids 130^148) [49,50,57,58]. In an mini-F [117]. In an arti¢cial system, the ABC primosome,
elegant study, O’Donnell and co-workers could show a DnaA box is positioned in a hairpin structure within
that two DnaB6 ^DnaC6 complexes are actually introduced single-stranded phage DNA [118]. In all cases, a single
into the bubble in a reaction that does not require helicase double-stranded DnaA box is followed by a single-
activity [29]. This stoichiometry was subsequently con- stranded region to its 3P-side.
¢rmed [28]. The DnaB hexamer forms a ring around sin-
gle-stranded DNA [108,109]. The channel through the
helicase ring corresponds to about 20 nucleotides. The 12. Variations of the theme : DnaA and oriC in other
bubble can therefore accommodate just two helicase rings. bacteria
A binding activity to single-stranded DNA induced in
DnaC upon interaction with DnaB has been described. Di¡erent bacteria have replication origins with widely
This seems to facilitate the transfer of DnaB to the tem- di¡ering sizes, but all (except Synechocystis, see below)
plate [110]. DnaC dissociates from the complexes with contain several DnaA boxes and an AT-rich region (Fig.
concomitant ATP hydrolysis resulting in two head-to- 5). For example, E. coli oriC consists of 260 bp and ¢ve
head helicase hexamers. This activates DnaB as a helicase DnaA boxes, B. subtilis oriC contains three clusters of
[111]. The two helicase complexes move past each other in DnaA boxes which are separated by the dnaA gene
5P-3P direction, and only after they moved s 65 nucleoti- [119]. T. thermophilus has symmetrically arranged 12 or
des can they interact with primase [29]. 13 boxes (number 12 and 13 overlap) [107], and Strepto-
The strand in oriC to which the ¢rst helicase is posi- myces has 19 DnaA boxes in 600 bp [106,120].
tioned by DnaA was determined using an arti¢cial sub-
strate that consisted of a double-stranded DnaA box R1 12.1. B. subtilis
of oriC and single-stranded DNA from the AT-rich region
in one strand and random single-stranded DNA in the All three clusters of DnaA boxes are required for oriC
other strand. At limiting DnaA concentrations DnaB function. The intervening dnaA gene can be largely deleted
was loaded to the lower strand [102,234]. This suggests if DnaA is provided in trans. There are three AT-rich 16-
that the ¢rst helicase hexamer is positioned onto the lower mers at the rpmH-proximal border of oriC, and an AT-
strand, as illustrated in Fig. 2. This is compatible with rich cluster of 27 bp at the dnaN side [121]. The unwinding
results obtained by footprinting [29]. reaction occurs at this 27-bp AT cluster and surrounding
There is a very striking similarity between the region of sequences [122]. DnaA proteins bound to the DnaA box
E. coli oriC where DnaB is loaded into the AT-rich bubble clusters interact and form loops that can be visualized in
and structures found in other systems where DnaB heli- the electron microscope. This is also seen with E. coli oriC,
case is loaded with the help of DnaA. Even the distance but the physiological relevance is not clear [122]. The

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374 363

DnaA protein is biochemically very similar to that of sion of the dnaA gene is autoregulated, as in the case of
E. coli [123]. As described above, the structure of the re- E. coli (see Section 13) [133].
gion between the right-most DnaA box in Fig. 5 and the One of the additional controls found in B. subtilis but
AT cluster is very similar to the corresponding region in not in E. coli are two replication checkpoints 100^200 kb
oriC of E. coli, including the positions of ATP-DnaA downstream of oriC on either side. They are dependent on
boxes, and the extent and positions of unwound nucleo- the stringent response and on a RTP protein-binding site
tides are virtually identical between E. coli and B. subtilis of the type normally used for replication arrest at the
[17]. chromosomal terminus [134].
The location of oriC with DnaA box clusters upstream
of dnaA or between dnaA and dnaN is conserved in many 12.2. Streptomyces
bacteria [124], e.g. Micrococcus luteus [36], Mycoplasma
capricolum [125], Spiroplasma citri [126], Mycobacterium Streptomyces species have large linear chromosomes
[127], Helicobacter pylori [128], and Streptomyces [106]. with a centrally located oriC. This oriC can be cloned as
Likewise, many bacteria carry a ‘dnaA’ operon down- a circular, autonomously replicating minichromosome,
stream, containing besides dnaA and dnaN, recF and which also has a low copy number [120]. Each oriC con-
gyrB. It has therefore been suggested that this genomic tains 19 DnaA boxes whose location and orientation are
arrangement represents a primordial structure [104,113, conserved [106]. Streptomyces oriC contains no extended
121,124]. However, there are also many exceptions with AT-rich region, but ¢ve short AT-rich stretches. Conse-
dnaA and oriC located in di¡erent environments on the quently, unwound bases are interspersed with paired ones
genome [129,130]. (J. Majka and J. Zakrzewska-Czerwinska, personal com-
A minichromosome replicating from oriC of B. subtilis munication). Therefore it is unknown where and how the
has been isolated [131]. However, distinct from minichro- replicative helicase is loaded.
mosomes of E. coli, such plasmids have a strong incom- Streptomyces DnaA protein is similar to that from
patibility to the chromosomal origin, and consequently E. coli in many respects [7]. It can oligomerize via two
their copy number is low. Incompatibility is also observed regions, one in domain 1 and one in domain 3 [47,48,
when isolated DnaA box clusters from oriC are cloned in 61,104]. The consensus sequence of Streptomyces DnaA
high-copy-number vectors [112]. Presumably, the relaxed boxes shows a G or C at position 3 [105,106], similar to
copy number control of E. coli minichromosomes is due to M. luteus [36]. Both are organisms with a high G+C con-
the Dam-SeqA system (see Section 16) that allows the cell tent. Such a strong Streptomyces DnaA box (5P-
to discriminate between replicated and not yet replicated TTGTCCACA) can bind a DnaA monomer, and di¡erent
origins. An organism like B. subtilis without such a meth- from the situation in E. coli, also a dimer [47,61]. How-
ylation must control its replication initiation much more ever, binding to a DnaA box with the E. coli consensus
stringently. An in vitro replication system for oriC plas- sequence is still better (KD around 10 nM for TTGTCCA-
mids has also been established for B. subtilis [132]. Expres- CA and around 3 nM for TTATCCACA) [48]. Weak

Fig. 5. Replication origins of di¡erent bacteria. DnaA boxes are shown as red arrows, AT-rich regions are shown in yellow.

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


364 W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374

boxes with mismatches require cooperation between DnaA 12.4. H. pylori


proteins, and therefore require two boxes [48,135]. In an
experiment where optimal binding sites were selected by H. pylori DnaA also has similar structural properties as
evolution from random sequences, the optimal spacing the other DnaA proteins [128]. The replication origin of
was 3 bp and the boxes were facing each other [48]. At H. pylori has been de¢ned by its location close to DnaA,
higher concentrations long-range interactions between by a strand switch in the G/C composition, and by ¢ve
DnaA protein bound to distant DnaA boxes in oriC pro- DnaA boxes in a 180-bp stretch. These DnaA boxes, how-
mote loop formation [47,104]. The promoter region of ever, all have at least one mismatch to the consensus se-
dnaA contains two DnaA boxes with the same spacing quence. Despite this, the DNA-binding domain 4 of Heli-
and arrangement as found in the selection from random cobacter DnaA can bind to each isolated DnaA box as a
sequence [136]. Cooperative binding of DnaA to these monomer [128].
boxes results in repression, and thus in autoregulation of
the dnaA gene [135]. 12.5. Synechocystis sp.

12.3. T. thermophilus Cyanobacteria also have regular DnaA proteins


[129,138]. The Synechocystis DnaA protein binds to
The T. thermophilus replication origin is also located DnaA boxes, as does its C-terminal domain 4 with a⁄n-
between the dnaA and dnaN genes. It has a quite unusual, ities comparable to E. coli DnaA. The genome of Syn-
nearly symmetrical structure [107]. Six DnaA boxes in the echocystis has been completely sequenced, and there is
¢rst half of oriC point toward dnaA, six boxes in the no stretch with several DnaA boxes that could qualify as
second half point toward the dnaN gene. Box number 13 a replication origin. In addition, the dnaA gene could be
overlaps with box number 12. Like Streptomyces, T. ther- deleted without loss of viability [139]. However, DnaA has
mophilus is an organism with a high G+C content in its been conserved during evolution, and its transcription is
chromosome. Nevertheless, all DnaA boxes, except num- not autoregulated but light-dependent instead, and follows
bers 6 and 13, have the E. coli consensus sequence, 5P- the circadian rhythm of DNA synthesis [139]. The easiest
TTATCCACA. At the dnaN side is a 40-bp-long AT- way to interpret these seemingly incompatible observa-
rich stretch. tions is to assume that normal replication initiation oper-
The Thermus DnaA protein has the same overall struc- ates via DnaA and an origin that is even more di⁄cult to
ture as the other DnaA proteins. The binding properties, detect than H. pylori oriC. In addition, Synechocystis must
however, are di¡erent. A single DnaA box is nearly inert have a very e⁄cient alternative initiation pathway, possi-
to DnaA. DnaA protein binding to oriC requires cooper- bly involving R-loops or D-loops as in the case of stable
ativity between monomers, and this cooperativity is en- replication in E. coli [140].
hanced when DnaA is in the ATP-complexed form (Table
2) [107]. There are no DnaA boxes in the promoter region 12.6. Caulobacter crescentus
of the dnaA gene, and consequently, T. thermophilus dnaA
is not autoregulated [137]. This is a bacterium with a developmentally di¡erenti-
A comparison of the two G+C-rich organisms, Strepto- ated cell cycle. Asymmetric cell division results in a repli-
myces and T. thermophilus, shows that they follow di¡er- cating stalked cell and a non-replicating swarmer cell. Mo-
ent strategies to adjust the properties of their DnaA pro- tile swarmer cells must di¡erentiate into sessile stalked
teins to their origins which are both rich in DnaA boxes. cells before they can replicate [141]. An essential control
Streptomyces with a high-a⁄nity DnaA protein reduces system for the cell cycle in this bacterium is a response
the quality of its DnaA boxes. Thermus, on the other regulator, CtrA (for cell cycle transcription regulator),
hand, compensates potentially high-a⁄nity DnaA boxes which is homologous to E. coli OmpR. The Caulobacter
with a DnaA protein with low binding a⁄nity. replication origin, Cori, contains ¢ve DnaA boxes, of
which none has the E. coli consensus sequence, and ¢ve
binding sites for CtrA [142]. CtrA binds to these sites with
Table 2 a 40-fold enhanced a⁄nity if it is phosphorylated by the
Apparent dissociation constants of T. thermophilus DnaA protein with histidine kinase CckA [143]. CtrA is only present in
DnaA boxes from T. thermophilus oriC
swarmer cells and represses among many other genes rep-
DnaA box(es) [DnaA] at 50% of Rmax (nM) lication from Cori [144]. During di¡erentiation from
ADP-DnaA ATP-DnaA swarmer to stalked cells CtrA is degraded by regulated
4 1300 1500 proteolysis [145]. Four of the ¢ve binding sites at which
7 2700 2200 CtrA represses replication could, by AT-richness and by
1^3 735 82 their location to the left-most DnaA box of Cori, represent
1^6 270 31 potential sites of unwinding which have to be cleared be-
1^13 420 60
fore initiation. Whatever the precise mechanism, regula-

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374 365

tion of replication by a transcriptional regulator, that is a dual initiator proteins ; DnaA works in concert with a
response regulator which can be phosphorylated, is a very plasmid-encoded initiator protein [156]. DnaA can assist
unusual way to regulate replication in this unusual organ- in the unwinding reaction, in helicase loading, or it can
ism. However, initiation of chromosome replication is have a primary role in the structure of the initiation com-
strictly dependent on DnaA also in Caulobacter [146]. plex, e.g. in pSC101. The dependence of the unwinding
Like E. coli, Caulobacter has a DNA methyltransferase, reaction on DnaA protein is di¡erent for di¡erent plas-
CcrM with the recognition sequence GANTC, that is cell mids. It is absolutely required for some, e.g. P1, where
cycle-regulated. However, it is not clear whether it has a DnaA can unwind the AT-rich region even alone,
similar role as Dam methyltransferase in E. coli [147]. although quite ine⁄ciently. DnaA is required, in concert
with the plasmid initiator, for unwinding in mini-F, RK2
and R6K. At the other end of the list is plasmid R1 which
13. DnaA as a transcription factor requires DnaA in vitro, but not in vivo. The cooperation
between DnaA and plasmid initiator proteins extends to
DnaA boxes are found in the promoter regions of many the loading of DnaB helicase, at least in the cases that
genes where they can mediate repression, transcriptional have been analyzed.
activation, or transcription termination due to loop for-
mation between two DnaA boxes in a transcription unit 14.1. P1
and long-range DnaA^DnaA interaction [99,148]. The
property of DnaA as a transcription factor has been cov- The origin of plasmid P1 contains two groups of DnaA
ered in a review by Messer and Weigel [9]. Here I want to boxes at either end. The left box region is followed by an
limit myself to some aspects of the autoregulation of the AT-rich region containing GATC sites. These sites must
dnaA gene. E. coli dnaA is transcribed from two pro- be methylated by Dam methyltransferase for e⁄cient ini-
moters. There is a consensus DnaA box between them. tiation [157]. Adjacent to the right box region are ¢ve 19-
Inactivation of DnaA in temperature-sensitive dnaA mu- bp repeats, binding sites for the plasmid initiator protein
tants results in derepression of dnaA promoters, whereas RepA. Only one of the two DnaA box regions is required.
overproduction of dnaA cloned in a plasmid represses Even one box is su⁄cient, provided it has the consensus
dnaA transcription [149^154]. However, a dnaA gene sequence for a strong box. Optimal replication needs both
with a mutation of the DnaA box (dnaA820) was still DnaA box regions [158,159]. The phasing of DnaA boxes
subject to autoregulation [155]. The solution of this para- relative to one another and the rest of the origin is impor-
dox is the presence of additional binding sites for DnaA tant [160]. DnaA is required for the unwinding of the AT-
between the promoters. There is a second DnaA box with rich region [159]. DnaA, together with HU protein, is
a mismatch and four ATP-DnaA boxes. The dnaA pro- su⁄cient for unwinding. However, the reaction is much
moter region was the model sequence for derivation of the more e⁄cient in the presence of RepA, as measured by
rules for DnaA binding [13]. ADP-DnaA binds to the two KMnO4 footprinting [116,160].
9-mer DnaA boxes, ATP-DnaA binds to all boxes as
shown by DNase I footprinting and surface plasmon res- 14.2. F
onance. The result is that ATP-DnaA is a much better
repressor for the dnaA promoter than ADP-DnaA [13]. Plasmid mini-F possesses all necessary functions for rep-
The promoter region with the dnaA820 mutation is bound lication of F plasmids. The replication origin (ori2) has
about 60% as e⁄ciently by ATP-DnaA as the wild-type two DnaA boxes, followed by an AT-rich region and
promoter. There are additional controls for dnaA expres- four 19-bp direct repeats, iterons to which the plasmid-
sion, e.g. methylation of GATC sites present in the region encoded initiator protein RepE binds as monomer. Both
(see Section 16), or Fis and IciA proteins. These are listed DnaA and RepE proteins alone, but together with HU
in [9]. protein, are able to unwind mini-F. For e⁄cient unwind-
ing, as well as sequence speci¢city for the AT-rich region,
however, the concerted action of DnaA, RepE, and HU is
14. Joint action of DnaA and plasmid initiators in plasmid required [117].
replication
14.3. RK2
DnaA is involved in the initiation of replication of many
E. coli plasmids. These plasmids contain in their replica- The replication origin of the broad-host-range plasmid
tion origins one or several DnaA boxes and an AT-rich RK2/RP4 (393 bp) carries four (lousy) DnaA boxes, ¢ve
region for unwinding, and, in addition, several iterons, 17-bp iterons for binding of the plasmid initiator TrfA in
repeated binding sites for a plasmid-encoded initiation its monomeric form, and an AT-rich region consisting of
protein. Whereas bacterial initiation requires the single four 13-mer repeats that are similar to the 13-mers in
initiator DnaA, initiation of plasmids with iterons requires E. coli oriC. DnaA binds cooperatively to the four

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


366 W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374

DnaA boxes, in agreement with the rules for DnaA bind- initiation in vitro, the DnaA box can be mutated [172].
ing formulated above. DnaA box number 4 seems to serve Presumably, similar to loading of DnaA to a DnaA box
as an anchor point [161]. However, DnaA binding by itself with mismatches, such a loading is also possible due to
does not result in unwinding of the AT-rich region, di¡er- interaction with RepA. The role of DnaA in R1 replica-
ent from the plasmids discussed above [162]. TrfA, bound tion is, however, not clear.
to the iterons, unwinds the AT-rich region in the presence
of HU and ATP. This unwinding is enhanced by DnaA 14.6. pSC101
[162]. DnaA is indispensable for the following step, the
delivery of the helicase [163]. However, DnaA cannot by The origin of pSC101 contains a consensus DnaA box
itself activate the DnaB helicase at the RK2 origin. Both followed by an AT-rich region, a binding site for IHF, and
DnaA and TrfA are required for DnaB-induced template ¢ve iterons for RepA binding. Within the iterons is a weak
unwinding [163]. Surprisingly, it has been found that the DnaA box with two mismatches to the consensus sequence
DnaA^DnaB^DnaC complex is not assembled at the un- [174]. DnaA is required for unwinding of the AT-rich re-
wound AT-rich region but at the DnaA box region that is gion and for helicase loading, in concert with RepA and
about 200 bp away [164]. We must therefore assume a IHF [174,175]. However, since domains 2 and 3 of DnaA
physical interaction by DNA looping between the DnaA are dispensable for pSC101 replication [49,176], the in-
box region with the bound complex and the unwound AT- volvement of DnaA must be indirect. It has been sug-
rich region, presumably with the help of TrfA. gested that DnaA ful¢lls a structural role in pSC101 ini-
tiation, together with IHF, by stabilizing a loop between
14.4. R6K the DnaA box at the left end of the origin and the iteron
region [174]. Mutant DnaA protein consisting of domains
The replication of plasmid R6K is initiated at either of 1 and 4 only might dimerize with domain 1, bind with
three origins, K, L, and Q. K and L origins require the domain 4 to the left DnaA box and with domain 4 of
presence of ori Q in cis for function. Initiation from ori the second monomer to the iteron region. This might hap-
K and ori Q requires both the plasmid-encoded initiator Z pen by binding to the weak DnaA box or by interaction
and DnaA protein (reviewed in [165]. ori Q contains (in with RepA. In fact, interaction of DnaA protein domain 1
this order) a DnaA box, an AT-rich region, and seven and of domain 4 with RepA has been demonstrated [176].
iterons for binding of Z. Between the AT-rich region and An alternative anchor at the iteron side of pSC101 ori
the iterons is a binding site for IHF. It has been suggested could be a second region of DnaA that can support
that its function is to bend the origin and thereby bring pSC101 replication [39]. DnaA domains 1 and 4 do not
into contact DnaA and Z [166]. Unwinding of the AT-rich need to be covalently linked. A leucine zipper added to
region in ori Q depends on an interaction between DnaA each domain allows a non-covalent association that is suf-
and the N-terminal part (amino acids 1^116) of Z [167]. ¢cient to provide a bridge between the left DnaA box and
The ATP-complexed form of DnaA is not required, since the iteron region in the origin [176].
a mutation in the ATP-binding site (K178A) of DnaA is
as e¡ective [167].
15. The special case of DnaA and V plasmids
14.5. R1
Replication of bacteriophage V, and of plasmids derived
DnaA protein is required for the in vitro replication of from phage V, depends on the phage encoded proteins O
plasmid R1, whereas it is dispensable for replication in and P (for review see [177]). O is a functional analogue of
vivo [168,169]. Therefore, dnaA(Null) mutants can be in- DnaA, and P replaces DnaC during loading of DnaB heli-
tegratively suppressed by R1, but not by F [170]. R1 rep- case. The replication origin (ori V) is located within the O
lication initiation is di¡erent from the other plasmids dis- gene. Transcription from the rightward V promoter pR is
cussed so far. Instead of iterons it contains two partially required for expression of O and P, but also for transcrip-
palindromic 10-bp sequences at each end of a 100-bp re- tional activation of the origin (for review see [177]). Tran-
gion. This region is completely protected in DNase I foot- scriptional activation is required for early replication of
printing experiments. At one end is a DnaA box, and at phage V, and for replication of V plasmids. Replication
the other end an AT-rich region [171]. A model has been of V plasmids depends on DnaA protein [178]. This was
proposed in which a DNA loop is formed by binding and surprising, since there is no DnaA box in the V origin
interaction of two RepA molecules, probably dimers, to region. However, binding of DnaA to several 9-bp sequen-
the binding sites at the ends of ori R, and then the 100-bp ces that have similarity to DnaA boxes, as well as to 6-mer
loop is ¢lled with more RepA. The AT-rich region is then ATP-DnaA boxes, has been demonstrated [101]. Since all
unwound in this higher-order complex [156,171]. Binding these boxes represent low-a⁄nity binding sites, coopera-
of DnaA to its box is very ine⁄cient and requires DnaA^ tive binding of DnaA is required. DnaA activates tran-
RepA contacts [172,173]. Although DnaA is required for scription, i.e. transcriptional activation, from pR in vivo

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374 367

and in vitro [179,180]. A DnaA box located several base R1. From there it seems to spread by cooperative binding
pairs downstream of pR is particularly important for this to adjacent regions, competing with DnaA for binding
activation, since mutation of this box abolishes transcrip- [198^200]. This is likely to be the main reason for the
tion stimulation by DnaA [180]. It has been shown that negative e¡ect of SeqA on initiation of replication. Also
early V replication via the bidirectional a mode depends on sequestration of the dnaA promoter region a¡ects initia-
activation of pR by DnaA. It is predominantly unidirec- tion negatively [193], since methylation of GATC sites in
tional in dnaA mutants [181]. Unidirectional a type of the promoter region is required for maximum promoter
replication precedes the late c type of replication. We activity [151,201]. Hence, shortly after initiation at the
may therefore assume that in dnaAþ hosts a depletion of time when the dnaA promoter is sequestered, there is a
the DnaA pool due to binding to DnaA boxes is the rea- transient suppression of dnaA expression [202]. Other in-
son for the switch from a to c replication [181]. The acti- hibitory e¡ects could come from changes in oriC topology
vation is delicately balanced, e.g. certain dnaA(Ts) mutants made by SeqA protein [203,204].
are unable to activate pR even at permissive temperature Sequestration by Dam methylation is restricted to En-
[182]. All these results show that the role of DnaA in terobacteriaceae. It gives E. coli and its relatives the op-
V plasmid replication is a role as transcription factor, portunity to regulate initiation in a very relaxed way.
and not as a replisome organizer. Since the cells are able to tell a new origin from an old
one they do not need to keep track of individual copies
and need not control the copy number of oriC tightly.
16. How to limit initiation to once per generation Consequently, the copy numbers of E. coli minichromo-
somes are around 10 per cell, and they show a very large
All organisms have developed mechanisms that ensure variation, and therefore a high loss rate [205,206]. Quite
chromosomal replication occurs once and only once per contrary, the copy number of minichromosomes from
generation [183]. In an E. coli cell there are at least three B. subtilis [131] or Streptomyces [120] is around 1 per
systems that prevent reinitiation of origins that have al- cell, and there is strong incompatibility between minichro-
ready initiated: (i) sequestration of oriC and of the dnaA mosomes and the chromosomal origin. As expected, ini-
promoter region after replication and hence blocking of tiation of origins that are not subject to methylation is
their activities for a given time window; (ii) binding of more tightly controlled.
DnaA protein to a region close to oriC that provides a
sink for DnaA ; (iii) regulatory inactivation of ATP-DnaA 16.2. Titration of DnaA to the datA locus
at the end of the initiation cycle. This has been discussed
in Sections 3 and 6. About 300 DnaA boxes, located around the E. coli
chromosome, are able to bind DnaA protein. Five chro-
16.1. Sequestration mosomal regions show an especially high a⁄nity [207].
The locus with the highest DnaA-binding capacity is
The replication origin of E. coli contains an unusually datA, located close to oriC [208]. It can bind about eight
high number (11) of GATC sequences, recognition sites times more DnaA than the combined oriC-mioC region
for the Dam methyltransferase. These sites must be meth- with a similar number of DnaA boxes. This high capacity,
ylated for e⁄cient initiation [184,185]. Especially hemi- compared to oriC, might result from a missing competi-
methylated GATC sites, as they are present shortly after tion with SeqA since there are only few GATC sites in
replication, are detrimental to initiation [186]. Hemimethy- datA. So far, it has not been analyzed whether there are
lated GATC sites in oriC, and also in the promoter region ATP-DnaA boxes in datA or close to the other high-a⁄n-
of dnaA, remain hemimethylated for about one-third of a ity sites that might be responsible for the high binding
generation, whereas elsewhere on the chromosome they capacity.
become remethylated within about 1 min [187,188]. The There is evidence for a direct involvement of datA in the
length of this eclipse period depends on the supply of Dam regulation of initiation. Deletion of datA results in over-
methyltransferase [184,189,190]. Since in vitro initiation initiation [209]. Additional copies of datA on a plasmid
can occur on unmethylated and hemimethylated origins limit initiation or block it completely [209,210]. We can
[184,191,192], it has been concluded that within cells these therefore safely assume that datA is a sink for DnaA pro-
regions are sequestered by binding to cellular factors. The tein which regulates the availability of DnaA and hence
most prominent of these factors is SeqA [193,194]. Anoth- a¡ects initiation.
er member of the membrane-bound sequestration complex
is SeqB [195].
SeqA has a high a⁄nity for hemimethylated oriC, some- 17. DnaA and oriC in the cell cycle, control of initiation
what less for fully methylated oriC, and does not bind
speci¢cally to unmethylated DNA [196,197]. It binds spe- When E. coli grows in a rich medium it contains several
ci¢cally to two sites in oriC, one on each side of DnaA box chromosomes. These initiate synchronously within a very

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


368 W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374

short time interval [183,211,212]. oriC present on mini- measured [74]. This does not invalidate these hypotheses,
chromosomes initiate in the same interval [213]. A number but they have to be re¢ned in view of the newer results.
of conditions upset this synchrony. These could either be
suboptimal conditions of initiation, e.g. in some dnaA(Ts)
mutants even at permissive temperature [214], or interfer- 18. Perspectives
ence with the sequestration apparatus, e.g. in dam and
seqA mutants [189,212]. DnaA and oriC and the replicative helicase are the ma-
Initiation occurs at a de¢ned time in the cell cycle. Don- jor elements in bacterial replication initiation. They have
achie discovered that at the time of initiation cells have a been the topic for extensive research for close to 40 years,
more or less constant volume per replication origin, called but many questions are still unanswered. So far most of
the initiation volume [215]. Later it was discovered that the research has centered on E. coli, but this one-sided
there is some variation in this initiation volume [216]. view is slowly broadening. Following good tradition in
After initiation it takes a constant time to replicate the molecular biology, the initiation cycle in bacteria, in
chromosome, independent of growth rate as long as it is E. coli, can serve as a paradigm for other more compli-
6 60 min at 37‡C, and a constant time between termina- cated systems. Eukaryotic viruses like SV40, polyoma or
tion of replication and cell division [217,218]. The fre- papilloma have origins and initiators that are basically
quency of initiation thus determines the rate of DNA syn- comparable to E. coli [232]. Yeast origins have an AT-
thesis, and indirectly the rate of cell division. rich region and a binding site for the six-protein origin
The DNA-bending proteins Fis and IHF have speci¢c recognition complex (ORC). ORC binds like DnaA to
binding sites in oriC (Fig. 1) [219^224]. The occupation of double-stranded and to single-stranded DNA, and the
binding sites by these architectural proteins and by DnaA change between the binding speci¢cities is associated
has been determined in synchronized cells by in vivo foot- with an ATP/ADP switch [75,233]. ORCs and correspond-
printing. The result was that throughout most of the cell ing origins have been found in all metazoa that have been
cycle, DnaA was bound to DnaA boxes R1, R2, and R4, looked at. The unwinding of double-stranded origin DNA
and Fis was bound to its site, presumably inhibiting ini- and the loading of helicase into this region is one of the
tiation. At the time of initiation Fis was lost from oriC, basic reactions in the cell cycle. The E. coli way of life
DnaA bound to DnaA box R3, and IHF bound to its site gives us a good example how to think about this process
[225,226]. This results in bending of oriC and in a redis- and design experiments.
tribution of DnaA [227]. So far a correlation of these in
vivo results with the results obtained in vitro has not been
attempted. 19. Note added in proof
The molecular basis of the initiation volume discussed
above apparently is the concentration of DnaA protein. It A crystal structure of domains 3 and 4 of one DnaA
has been shown that the initiation volume can be changed protein has recently been published [Erzberger, J.P., Pir-
by modulation of the DnaA level [228]. A hypothesis has ruccello, M.M. and Berger, J.M. (2002) The structure of
been formulated [229,230] that postulates that initially all bacterial DnaA: implications for general mechanisms
newly synthesized DnaA protein is bound to DnaA boxes underlying DNA replication initiation. EMBO J. 21,
around the chromosome. These boxes increase in number 4763^4773]. It extends and corroborates many of the pre-
due to replication, and DnaA continues to be synthesized dictions given in Section 4.
until the number of DnaA molecules exceeds the number
of DnaA boxes. This is the moment of initiation. A neces-
sary condition for this hypothesis is that the binding event Acknowledgements
that triggers initiation is of lower a⁄nity than binding to
the other boxes [229]. The switch to cooperative binding at I thank Harald Seitz, Kirsten Skarstad, Grzegorz Wegr-
ATP-DnaA boxes (see Section 10) is ideally suited to ful¢ll zyn, Christoph Weigel and Jolanta Zakrzewska-Czerwin-
this condition. ska for critically reading the manuscript. I gratefully ac-
In addition, it has been suggested that DnaA protein knowledge support from the Fonds der Chemischen
molecules that become free due to initiation and replica- Industrie.
tion are immediately used for initiation at adjacent origins
in the form of an initiation cascade or an avalanche, there-
by ensuring the highly synchronous initiation of all origins References
in a cell [231].
[1] Kohiyama, M., Cousin, D., Ryter, A. and Jacob, F. (1966) Mutants
Both these hypotheses have been formulated before the
thermosensibles d’E. coli K-12. I. Isolement et characterisation ra-
discovery of inactivation of ATP-DnaA due to ATP hy- pide. Ann. Inst. Pasteur 110, 465^486.
drolysis [33], and before the drastic changes in the propor- [2] Kohiyama, M. (1968) DNA synthesis in temperature sensitive mu-
tion of ATP-DnaA before and after initiation had been tants in E. coli. Cold Spring Harbor Symp. Quant. Biol. 43, 317^324.

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374 369

[3] Jacob, F., Brenner, S. and Cuzin, F. (1963) On the regulation of [24] Lark, K.G. (1972) Evidence for direct involvement of RNA in the
DNA replication in bacteria. Cold Spring Harbor Symp. Quant. initiation of DNA replication in E. coli 15T. J. Mol. Biol. 64, 47^60.
Biol. 28, 329^348. [25] Baker, T.A. and Kornberg, A. (1988) Transcriptional activation of
[4] Skarstad, K. and Boye, E. (1994) The initiator protein DnaA: Evo- initiation of replication from the E. coli chromosomal origin: An
lution, properties and function. Biochim. Biophys. Acta 1217, 111^ RNA-DNA hybrid near oriC. Cell 55, 113^123.
130. [26] Funnell, B.E., Baker, T.A. and Kornberg, A. (1987) In vitro assembly
[5] Messer, W. and Weigel, C. (1996) Initation of chromosome replica- of a prepriming complex at the origin of the Escherichia coli chro-
tion. In: Escherichia coli and Salmonella, Cellular and Molecular mosome. J. Biol. Chem. 262, 10327^10334.
Biology (Neidhardt, F.C., Curtiss III, R., Ingraham, J., Lin, [27] Crooke, E., Thresher, R., Hwang, D.S., Gri⁄th, J. and Kornberg, A.
E.C.C., Low, K.B., Magasanik, B., Rezniko¡, W.S., Riley, M., (1993) Replicatively active complexes of DnaA protein and the Es-
Schaechter, M. and Umbarger, H.E., Eds.), pp. 1579^1601. ASM, cherichia coli chromosomal origin observed in the electron micro-
Washington, DC. scope. J. Mol. Biol. 233, 16^24.
[6] Kaguni, J.M. (1997) Escherichia coli DnaA protein: the replication [28] Carr, K.M. and Kaguni, J.M. (2001) Stoichiometry of DnaA and
initiator. Mol. Cells 7, 145^157. DnaB protein in initiation at the Escherichia coli chromosomal ori-
[7] Messer, W., Blaesing, F., Jakimowicz, D., Majka, J., Nardmann, J., gin. J. Biol. Chem. 276, 44919^44925.
Schaper, S., Seitz, H., Speck, C., Weigel, C., Wegrzyn, G., Welzeck, [29] Fang, L.H., Davey, M.J. and O’Donnell, M. (1999) Replisome as-
M. and Zakrzewska-Czerwinska, J. (2001) Bacterial replication ini- sembly at oriC, the replication origin of E. coli, reveals an explana-
tiator DnaA. Rules for DnaA binding and roles of DnaA in origin tion for initiation sites outside an origin. Mol. Cell 4, 541^553.
unwinding and helicase loading. Biochimie 83, 1^9. [30] Wahle, E., Lasken, R.S. and Kornberg, A. (1989) The dnaB-dnaC
[8] Echols, H. (1990) Nucleoprotein structures initiating DNA replica- replication protein complex of Escherichia coli. I. Formation and
tion, transcription, and site-speci¢c recombination. J. Biol. Chem. properties. J. Biol. Chem. 264, 2463^2468.
265, 14697^14700. [31] Messer, W., Seufert, W., Schaefer, C., Gielow, A., Hartmann, H. and
[9] Messer, W. and Weigel, C. (1997) DnaA initiator ^ also a transcrip- Wende, M. (1988) Functions of the DnaA protein of Escherichia coli
tion factor. Mol. Microbiol. 24, 1^6. in replication and transcription. Biochim. Biophys. Acta 951, 351^
[10] Kornberg, A. and Baker, T.A. (1992) DNA Replication. W.H. Free- 358.
man and Company, New York. [32] Kelman, Z. and O’Donnell, M. (1995) DNA polymerase III holoen-
[11] Schaper, S. and Messer, W. (1995) Interaction of the initiator protein zyme: structure and function of a chromosomal replicating machine.
DnaA of Escherichia coli with its DNA target. J. Biol. Chem. 270, Annu. Rev. Biochem. 64, 171^200.
17622^17626. [33] Katayama, T., Kubota, T., Kurokawa, K., Crooke, E. and Sekimizu,
[12] Sekimizu, K., Bramhill, D. and Kornberg, A. (1987) ATP activates K. (1998) The initiator function of DnaA protein is negatively regu-
dnaA protein in initiating replication of plasmids bearing the origin lated by the sliding clamp of the E. coli chromosomal replicase. Cell
of the E. coli chromosome. Cell 50, 259^265. 94, 61^71.
[13] Speck, C., Weigel, C. and Messer, W. (1999) ATP- and ADP-DnaA [34] Katayama, T. and Sekimizu, K. (1999) Inactivation of Escherichia
protein, a molecular switch in gene regulation. EMBO J. 18, 6169^ coli DnaA protein by DNA polymerase III and negative regulations
6176. for initiation of chromosomal replication. Biochimie 81, 835^840.
[14] Speck, C. and Messer, W. (2001) Mechanism of origin unwinding: [35] Kato, J. and Katayama, T. (2001) Hda, a novel DnaA-related pro-
sequential binding of DnaA initiator protein to double-stranded and tein, regulates the replication cycle in Escherichia coli. EMBO J. 20,
single-stranded DNA in the AT-rich region of the replication origin. 4253^4262.
EMBO J. 20, 1469^1476. [36] Fujita, M.Q., Yoshikawa, H. and Ogasawara, N. (1990) Structure of
[15] Bramhill, D. and Kornberg, A. (1988) Duplex opening by dnaA the dnaA region of Micrococcus luteus: Conservation and variations
protein at novel sequences in initiation of replication at the origin among eubacteria. Gene 93, 73^78.
of the E. coli chromosome. Cell 52, 743^755. [37] Skovgaard, O. and Hansen, F.G. (1987) Comparison of dnaA nucle-
[16] Gille, H. and Messer, W. (1991) Localized unwinding and structural otide sequences of Escherichia coli, Salmonella typhimurium, and Ser-
perturbations in the origin of replication, oriC, of Escherichia coli in ratia marcescens. J. Bacteriol. 169, 3976^3981.
vitro and in vivo. EMBO J. 10, 1579^1584. [38] Messer, W., Blaesing, F., Majka, J., Nardmann, J., Schaper, S.,
[17] Krause, M. and Messer, W. (1999) DnaA proteins of Escherichia coli Schmidt, A., Seitz, H., Speck, C., Tu«ngler, D., Wegrzyn, G., Weigel,
and Bacillus subtilis: coordinate actions with single-stranded DNA- C., Welzeck, M. and Zakrzewska-Czerwinska, J. (1999) Functional
binding protein and interspecies inhibition during open complex for- domains of DnaA proteins. Biochimie 81, 819^825.
mation at the replication origins. Gene 228, 123^132. [39] Sutton, M.D. and Kaguni, J.M. (1997) The Escherichia coli dnaA
[18] Hiasa, H. and Marians, K.J. (1994) Fis cannot support oriC DNA gene: Four functional domains. J. Mol. Biol. 274, 546^561.
replication in vitro. J. Biol. Chem. 269, 24999^25003. [40] Schaper, S. and Messer, W. (1997) Prediction of the structure of the
[19] Wold, S., Crooke, E. and Skarstad, K. (1996) The Escherichia coli Fis replication initiator protein DnaA. Proteins Struct. Funct. Genet. 28,
protein prevents initiation of DNA replication from oriC in vitro. 1^9.
Nucleic Acids Res. 24, 3527^3532. [41] Neuwald, A.F., Aravind, L., Spouge, J.L. and Koonin, E.V. (1999)
[20] Dixon, N.E. and Kornberg, A. (1984) Protein HU in the enzymatic AAA(+): A class of chaperone-like ATPases associated with the as-
replication of the chromosomal origin of Escherichia coli. Proc. Natl. sembly, operation, and disassembly of protein complexes. Genome
Acad. Sci. USA 81, 424^428. Res. 9, 27^43.
[21] Skarstad, K., Baker, T.A. and Kornberg, A. (1990) Strand separation [42] Rost, B. and Sander, C. (1994) Combining evolutionary information
required for initiation of replication at the chromosomal origin of and neural networks to predict protein secondary structure. Proteins
E. coli is facilitated by a RNA-DNA hybrid. EMBO J. 9, 2341^2348. 19, 55^72.
[22] Sekimizu, K., Bramhill, D. and Kornberg, A. (1988) Sequential early [43] Rossmann, M.G., Moras, D. and Olsen, K.W. (1974) Chemical and
stages in the in vitro initiation of replication at the origin of the biological evolution of nucleotide-binding protein. Nature 250, 194^
Escherichia coli chromosome. J. Biol. Chem. 263, 7124^7130. 199.
[23] Messer, W. (1972) Initiation of DNA replication in E. coli B/r. Chro- [44] Guenther, B., Onrust, R., Sali, A., O’Donnell, M. and Kuriyan, J.
nology of events and transcriptional control of initiation. J. Bacteriol. (1997) Crystal structure of the deltaP subunit of the clamp-loader
112, 7^12. complex of E. coli DNA polymerase III. Cell 91, 335^345.

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


370 W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374

[45] Roth, A. and Messer, W. (1995) The DNA binding domain of the defect by mutation in the initiation gene, dnaA, requires functional
initiator protein DnaA. EMBO J. 14, 2106^2111. oriC. Mol. Microbiol. 36, 913^925.
[46] Weigel, C., Schmidt, A., Seitz, H., Tuengler, D., Welzeck, M. and [64] Kubota, T., Ito, Y., Sekimizu, K., Tagaya, M. and Katayama, T.
Messer, W. (1999) The N-terminus promotes oligomerisation of the (2001) DnaA protein Lys-415 is close to the ATP-binding site: ATP-
Escherichia coli initiator protein DnaA. Mol. Microbiol. 34, 53^66. pyridoxal a⁄nity labeling. Biochem. Biophys. Res. Commun. 288,
[47] Jakimowicz, D., Majka, J., Konopa, G., Wegrzyn, G., Messer, W., 1141^1148.
Schrempf, H. and Zakrzewska-Czerwinska, J. (2000) Architecture of [65] Hansen, F.G., Koefoed, S. and Atlung, T. (1992) Cloning and nucle-
the Streptomyces lividans DnaA protein-replication origin complexes. otide sequence determination of twelve mutant dnaA genes of Esche-
J. Mol. Biol. 298, 351^364. richia coli. Mol. Gen. Genet. 234, 14^21.
[48] Majka, J., Zakrzewska-Czerwinska, J. and Messer, W. (2001) Se- [66] Hwang, D.S. and Kaguni, J.M. (1988) Interaction of dnaA46 protein
quence recognition, cooperative interaction and dimerization of the with a stimulatory protein in replication from the Escherichia coli
initiator protein DnaA of Streptomyces. J. Biol. Chem. 276, 6243^ chromosomal origin. J. Biol. Chem. 263, 10633^10640.
6252. [67] Katayama, T. (1994) The mutant DnaAcos protein which overiniti-
[49] Seitz, H., Weigel, C. and Messer, W. (2000) The interaction domains ates replication of the Escherichia coli chromosome is inert to neg-
of the DnaA and DnaB replication proteins of Escherichia coli. Mol. ative regulation for initiation. J. Biol. Chem. 269, 22075^22079.
Microbiol. 37, 1270^1279. [68] Carr, K.M. and Kaguni, J.M. (1996) The A184V missense mutation
[50] Sutton, M.D., Carr, K.M., Vicente, M. and Kaguni, J.M. (1998) of the dnaA5 and dnaA46 alleles confers a defect in ATP binding and
Escherichia coli DnaA protein. The N-terminal domain and loading thermolability in initiation of Escherichia coli DNA replication. Mol.
of DnaB helicase at the E. coli chromosomal origin. J. Biol. Chem. Microbiol. 20, 1307^1318.
273, 34255^34262. [69] Braun, R.E., O’Day, K. and Wright, A. (1987) Cloning and charac-
[51] Mima, S., Yamagachi, Y., Kondo, T., Tsuchiya, T. and Mizushima, terization of dnaA(Cs), a mutation which leads to overinitiation of
T. (1999) Role of the amino-terminal region of the DnaA protein in DNA replication in Escherichia coli K-12. J. Bacteriol. 169, 3898^
opening of the duplex DNA at the oriC region. FEMS Microbiol. 3903.
Lett. 176, 163^167. [70] Nyborg, M., Atlung, T., Skovgaard, O. and Hansen, F.G. (2000)
[52] Walker, J.E., Saraste, M., Runswick, M.J. and Gay, N.J. (1982) Two types of cold sensitivity associated with the A184V change in
Distantly related sequences in the alpha and beta subunits of ATP the DnaA protein. Mol. Microbiol. 35, 1202^1210.
synthase, myosin, kinases and other ATP requiring enzymes and a [71] Kellenberger-Gujer, G., Podhajska, A.J. and Caro, L. (1978) A cold
commom binding fold. EMBO J. 1, 945^951. sensitive dnaA mutant of E. coli which overinitiates chromosome
[53] Saraste, M., Sibbald, P.R. and Wittinghofer, A. (1990) The P-loop ^ replication at low temperature. Mol. Gen. Genet. 162, 9^16.
a common motif in ATP- and GTP-binding proteins. Trends Bio- [72] Katayama, T. and Kornberg, A. (1994) Hyperactive initiation of
chem. Sci. 15, 430^434. chromosomal replication in vivo and in vitro by a mutant initiator
[54] Koonin, E.V. (1993) A common set of conserved motifs in a vast protein, DnaAcos, of Escherichia coli. J. Biol. Chem. 269, 12698^
variety of putative nucleic acid-dependent ATPases including MCM 12703.
proteins involved in the initiation of eukaryotic DNA replication. [73] Katayama, T. and Crooke, E. (1995) DnaA protein is sensitive to a
Nucleic Acids Res. 21, 2541^2547. soluble factor and is speci¢cally inactivated for initiation of in vitro
[55] Mizushima, T., Nishida, S., Kurokawa, K., Katayama, T., Miki, T. replication of the Escherichia coli minichromosome. J. Biol. Chem.
and Sekimizu, K. (1997) Negative control of DNA replication by 270, 9265^9271.
hydrolysis of ATP bound to DnaA protein, the initiator of chromo- [74] Kurokawa, K., Nishida, S., Emoto, A., Sekimizu, K. and Katayama,
somal DNA replication in Escherichia coli. EMBO J. 16, 3724^3730. T. (1999) Replication cycle-coordinated change of the adenine nucle-
[56] Su’etsugu, M., Kawakami, H., Kurokawa, K., Kubota, T., Takata, otide-bound forms of DnaA protein in Escherichia coli. EMBO J. 18,
M. and Katayama, T. (2001) DNA replication-coupled inactivation 6642^6652.
of DnaA protein in vitro: a role for DnaA arginine-334 of the [75] Lee, D.G. and Bell, S.P. (2000) ATPase switches controlling DNA
AAA(+) Box VIII motif in ATP hydrolysis. Mol. Microbiol. 40, replication initiation. Curr. Opin. Cell Biol. 12, 280^285.
376^386. [76] Hwang, D.S. and Kornberg, A. (1992) Opening of the replication
[57] Marszalek, J. and Kaguni, J.M. (1994) DnaA protein directs the origin of Escherichia coli by DnaA protein with protein HU or
binding of DnaB protein in initiation of DNA replication in Esche- IHF. J. Biol. Chem. 267, 23083^23086.
richia coli. J. Biol. Chem. 269, 4883^4890. [77] Makise, M., Tsuchiya, T. and Mizushima, T. (2002) Sequence-inde-
[58] Marszalek, J., Zhang, W.G., Hupp, T.R., Margulies, C., Carr, K.M., pendent DNA binding activity of DnaA protein, the initiator of chro-
Cherry, S. and Kaguni, J.M. (1996) Domains of DnaA protein in- mosomal DNA replication in Escherichia coli. J. Biochem. 131, 419^
volved in interaction with DnaB protein, and in unwinding the Es- 425.
cherichia coli chromosomal origin. J. Biol. Chem. 271, 18535^18542. [78] Sekimizu, K., Yung, B.Y. and Kornberg, A. (1988) The dnaA protein
[59] Biswas, E.E. and Biswas, S.B. (1999) Mechanism of DnaB helicase of of Escherichia coli. Abundance, improved puri¢cation, and mem-
Escherichia coli : Structural domains involved in ATP hydrolysis, brane binding. J. Biol. Chem. 263, 7136^7140.
DNA binding, and oligomerization. Biochemistry 38, 10919^10928. [79] Newman, G. and Crooke, E. (2000) DnaA, the initiator of Escheri-
[60] Blaesing, F., Weigel, C. and Messer, W. (2000) Analysis of the DNA chia coli chromosomal replication, is located at the cell membrane.
binding domain of Escherichia coli DnaA protein. Mol. Microbiol. J. Bacteriol. 182, 2604^2610.
36, 557^569. [80] Xia, W. and Dowhan, W. (1995) In vivo evidence for the involvement
[61] Majka, J., Jakimowicz, D., Messer, W., Schrempf, H., Lisowski, M. of anionic phospholipids in initiation of DNA replication in Esche-
and Zakrzewska-Czerwinska, J. (1999) Interactions of the Streptomy- richia coli. Proc. Natl. Acad. Sci. USA 92, 783^787.
ces lividans initiator protein DnaA with its target. Eur. J. Biochem. [81] Zheng, W.D., Li, Z.Y., Skarstad, K. and Crooke, E. (2001) Muta-
260, 325^335. tions in DnaA protein suppress the growth arrest of acidic phospho-
[62] Sutton, M.D. and Kaguni, J.M. (1997) Threonine 435 of Escherichia lipid-de¢cient Escherichia coli cells. EMBO J. 20, 1164^1172.
coli DnaA protein confers sequence-speci¢c DNA binding activity. [82] Wegrzyn, A., Wrobel, B. and Wegrzyn, G. (1999) Altered biological
J. Biol. Chem. 272, 23017^23024. properties of cell membranes in Escherichia coli dnaA and seqA mu-
[63] Blinkova, A., Gines-Candelaria, E., Ross, J.D. and Walker, J.R. tants. Mol. Gen. Genet. 261, 762^769.
(2000) Suppression of a DnaX temperature-sensitive polymerization [83] Yung, B.Y.M. and Kornberg, A. (1988) Membrane attachment acti-

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374 371

vates dnaA protein, the initiation protein of chromosome replication pa, G., Lurz, R., Marszalek, J., Taylor, K., Messer, W. and Wegr-
in Escherichia coli. Proc. Natl. Acad. Sci. USA 85, 7202^7205. zyn, G. (1998) Interaction of the Escherichia coli DnaA protein with
[84] Sekimizu, K. and Kornberg, A. (1988) Cardiolipin activation of lambda phage DNA. Mol. Gen. Genet. 259, 679^688.
DnaA protein, the initiation protein in E. coli. J. Biol. Chem. 263, [102] Seitz, H. (2000) Interaktionen des E. coli Initiatorproteins DnaA mit
7131^7135. der replikativen Helikase DnaB. PhD Thesis, FU Berlin. http://
[85] Castuma, C.E., Crooke, E. and Kornberg, A. (1993) Fluid mem- www.diss.fu-berlin.de/diss/index.html.
branes with acidic domains activate DnaA, the initiator protein of [103] Seitz, H., Welzeck, M. and Messer, W. (2001) A hybrid bacterial
replication in Escherichia coli. J. Biol. Chem. 268, 24665^24668. replication origin. EMBO Rep. 2, 1003^1006.
[86] Mizushima, T., Ishikawa, Y., Obana, E., Hase, M., Kubota, T., [104] Zakrzewska-Czerwinska, J., Jakimowicz, D., Majka, J., Messer, W.
Katayama, T., Kunitake, T., Watanabe, E. and Sekimizu, K. and Schrempf, H. (2000) Initiation of the Streptomyces chromosome
(1996) In£uence of cluster formation of acidic phospholipids on replication. Antonie van Leeuwenhoek 78, 211^221.
decrease in the a⁄nity for ATP of DnaA protein. J. Biol. Chem. [105] Zakrzewska-Czerwinska, J., Majka, J. and Schrempf, H. (1995)
271, 3633^3638. Minimal requirements of the Streptomyces lividans 66 oriC region
[87] Crooke, E., Castuma, C.E. and Kornberg, A. (1992) The chromo- and its transcriptional and translational activities. J. Bacteriol. 177,
some origin of Escherichia coli stabilizes DnaA protein during reju- 4765^4771.
venation by phospholipids. J. Biol. Chem. 267, 16779^16782. [106] Jakimowicz, D., Majka, J., Messer, W., Speck, C., Fernandez, M.,
[88] Hase, M., Yoshimi, T., Ishikawa, Y., Ohba, A., Guo, L., Mima, S., Martin, M.C., Sanchez, J., Schauwecker, F., Keller, U., Schrempf,
Makise, M., Yamaguchi, K., Tsuchiya, T. and Mizushima, T. (1998) H. and Zakrzewska-Czerwinska, J. (1998) Structural elements of the
Site-directed mutational analysis for the membrane binding of Streptomyces oriC region and their interactions with the DnaA pro-
DnaA protein. Identi¢cation of amino acids involved in the func- tein. Microbiology 144, 1281^1290.
tionsl interaction between DnaA protein and acidic phospholipids. [107] Schaper, S., Nardmann, J., Lu«der, G., Lurz, R., Speck, C. and
J. Biol. Chem. 273, 28651^28656. Messer, W. (2000) Identi¢cation of the chromosomal replication
[89] Yamaguchi, Y., Hase, M., Makise, M., Mima, S., Yoshimi, T., origin from Thermus thermophilus and its interaction with the rep-
Ishikawa, Y., Tsuchiya, T. and Mizushima, T. (1999) Involvement lication initiator DnaA. J. Mol. Biol. 299, 655^665.
of Arg-328, Arg-334 and Arg-342 of DnaA protein in the functional [108] San Martin, M.C., Stamford, N.P.J., Dammerova, N., Dixon, N.E.
interaction with acidic phospholipids. Biochem. J. 340, 433^438. and Carazo, J.M. (1995) A structural model for the Escherichia coli
[90] Makise, M., Mima, S., Tsuchiya, T. and Mizushima, T. (2000) DnaB helicase based on electron microscopy data. J. Struct. Biol.
Identi¢cation of amino acids involved in the functional interaction 114, 167^176.
between DnaA protein and acidic phospholipids. J. Biol. Chem. 275, [109] Egelman, H.H., Yu, X., Wild, R., Hingorani, M.M. and Patel, S.S.
4513^4518. (1995) Bacteriophage T7 helicase/primase proteins form rings
[91] Makise, M., Mima, S., Tsuchiya, T. and Mizushima, T. (2001) Mo- around single-stranded DNA that suggest a general structure for
lecular mechanism for functional interaction between DnaA protein hexameric helicases. Proc. Natl. Acad. Sci. USA 92, 3869^3873.
and acidic phospholipids ^ Identi¢cation of important amino acids. [110] Learn, B.A., Um, S.-J., Huang, L. and McMacken, R. (1997) Cryp-
J. Biol. Chem. 276, 7450^7456. tic single-stranded-DNA binding activities of the phage V P and
[92] Garner, J. and Crooke, E. (1996) Membrane regulation of the chro- Escherichia coli DnaC replication initiation proteins facilitate the
mosomal replication activity of E. coli DnaA requires a discrete site transfer of E. coli DnaB helicase onto DNA. Proc. Natl. Acad.
on the protein. EMBO J. 15, 3477^3485. Sci. USA 94, 1154^1159.
[93] Speck, C., Weigel, C. and Messer, W. (1997) From footprint to [111] Wahle, E., Lasken, R.S. and Kornberg, A. (1989) The dnaB-dnaC
toeprint : a close-up of the DnaA box, the binding site for the ini- replication protein complex of Escherichia coli. II. role of the com-
tiator protein DnaA of Escherichia coli. Nucleic Acids Res. 25, plex in mobilizing dnaB functions. J. Biol. Chem. 264, 2469^2475.
3242^3247. [112] Moriya, S., Fukuoka, T., Ogasawara, N. and Yoshikawa, H. (1988)
[94] Fuller, R.S. and Kornberg, A. (1983) Puri¢ed dnaA protein in ini- Regulation of initiation of the chromosomal replication by DnaA-
tiation of replication at the Escherichia coli chromosomal origin of boxes in the origin of the Bacillus subtilis chromosome. EMBO J. 7,
replication. Proc. Natl. Acad. Sci. USA 80, 5817^5821. 2911^2917.
[95] Fuller, R.S., Funnell, B.E. and Kornberg, A. (1984) The dnaA pro- [113] Fujita, M.Q., Yoshikawa, H. and Ogasawara, N. (1989) Structure of
tein complex with the E. coli chromosomal origin (oriC) and other the dnaA region of Pseudomonas putida : Conservation among three
sites. Cell 38, 889^900. bacteria, Bacillus subtilis, Escherichia coli and P. putida. Mol. Gen.
[96] Matsui, M., Oka, A., Takanami, M., Yasuda, S. and Hirota, Y. Genet. 215, 381^387.
(1985) Sites of dnaA protein-binding in the replication origin of [114] Smith, D.W., Yee, T.W., Baird, C. and Krishnapillai, V. (1991)
the E. coli K-12 chromosome. J. Mol. Biol. 184, 529^533. Pseudomonad replication origins: A paradigm for bacterial origins.
[97] Holz, A., Schaefer, C., Gille, H., Jueterbock, W.-R. and Messer, W. Mol. Microbiol. 5, 2581^2587.
(1992) Mutations in the DnaA binding sites of the replication origin [115] Suhan, M., Chen, S.-Y., Thompson, H.A., Hoover, T.A., Hill, A.
of Escherichia coli. Mol. Gen. Genet. 233, 81^88. and Williams, J.C. (1994) Cloning and characterization of an auton-
[98] Schaefer, C. and Messer, W. (1991) DnaA protein/DNA interaction. omous replication sequence from Coxiella burnetii. J. Bacteriol. 176,
Modulation of the recognition sequence. Mol. Gen. Genet. 226, 34^ 5233^5243.
40. [116] Park, K., Mukhopadhyay, S. and Chattoraj, D. (1998) Require-
[99] Konopa, G., Szalewska-Palasz, A., Schmidt, A., Srutkowska, S., ments for and regulation of origin opening of plasmid P1. J. Biol.
Messer, W. and Wegrzyn, G. (1999) The presence of two DnaA- Chem. 273, 24906^24911.
binding sequences is required for e⁄cient interaction of the Esche- [117] Kawasaki, Y., Matsunaga, F., Kano, Y., Yura, T. and Wada, C.
richia coli DnaA protein with each particular weak DnaA box re- (1996) The localized melting of mini-F by the combined action of
gion. FEMS Microbiol. Lett. 174, 25^31. the mini-F initiator protein (RepE) and HU and DnaA of Escheri-
[100] Berenstein, D., Olesen, K., Speck, C. and Skovgaard, O. (2002) chia coli. Mol. Gen. Genet. 253, 42^49.
Genetic organization of the Vibrio harveyi dnaA gene region and [118] Masai, H., Nomura, N. and Arai, K. (1990) The ABC-primosome.
analysis of the function of the V. harveyi DnaA protein in Escheri- A novel priming system employing dnaA, dnaB, dnaC, and primase
chia coli. J. Bacteriol. 184, 2533^2538. on a hairpin containing a dnaA box sequence. J. Biol. Chem. 265,
[101] Szalewska-Palasz, A., Weigel, C., Speck, C., Srutkowska, S., Kono- 15134^15144.

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


372 W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374

[119] Ogasawara, N., Moriya, S., von Meyenburg, K., Hansen, F.G. and Inducible transcription of the dnaA gene from Streptomyces lividans
Yoshikawa, H. (1985) Conservation of genes and their organization 66. Mol. Gen. Genet. 242, 440^447.
in the chromosomal replication origin region of Bacillus subtilis and [137] Nardmann, J. and Messer, W. (2001) Identi¢cation and character-
Escherichia coli. EMBO J. 4, 3345^3350. ization of the dnaA upstream region of Thermus thermophilus. Gene
[120] Zakrzewska-Czerwinska, J. and Schrempf, H. (1992) Characteriza- 261, 299^303.
tion of an autonomously replicating region from the Streptomyces [138] Richter, S. and Messer, W. (1995) Genetic structure of the dnaA
lividans chromosome. J. Bacteriol. 174, 2688^2693. region of the cyanobacterium Synechocystis sp. PCC6803. J. Bacte-
[121] Yoshikawa, H. and Wake, R.G. (1993) Initiation and termination of riol. 177, 4245^4251.
chromosome replication. In: Bacillus subtilis and Other Gram-Pos- [139] Richter, S., Hagemann, M. and Messer, W. (1998) Transcriptional
itive Bacteria : Biochemistry, Physiology, and Molecular Genetics analysis and mutation of a dnaA-like gene in Synechocystis sp.
(Sonenshein, A.L., Hoch, J.A. and Losick, R., Eds.), pp. 507^528. PCC6803. J. Bacteriol. 180, 4946^4949.
ASM, Washington, DC. [140] Kogoma, T. (1997) Stable DNA replication : interplay between
[122] Krause, M., Lurz, R., Ru«ckert, B. and Messer, W. (1997) Com- DNA replication, homologous recombination, and transcription.
plexes at the replication origin of Bacillus subtilis with homologous Microbiol. Mol. Biol. Rev. 61, 212^238.
and heterologous DnaA protein. J. Mol. Biol. 274, 365^380. [141] Marczynski, G.T., Dingwall, A. and Shapiro, L. (1990) Plasmid and
[123] Fukuoka, T., Moriya, S., Yoshikawa, H. and Ogasawara, N. (1990) chromosomal DNA replication and partitioning during the Caulo-
Puri¢cation and characterization of an initiation protein for chro- bacter crescentus cell cycle. J. Mol. Biol. 212, 709^722.
mosomal replication, DnaA, in Bacillus subtilis. J. Biochem. (Tokyo) [142] Marczynski, G.T. and Shapiro, L. (1992) Cell-cycle control of a
107, 732^739. cloned chromosomal origin of replication from Caulobacter cres-
[124] Yoshikawa, H. and Ogasawara, N. (1991) Structure and function of centus. J. Mol. Biol. 226, 959^977.
DnaA and the DnaA-box in eubacteria: Evolutionary relationships [143] Siam, R. and Marczynski, G.T. (2000) Cell cycle regulator phos-
of bacterial replication origins. Mol. Microbiol. 5, 2589^2597. phorylation stimulates two distinct modes of binding at a chromo-
[125] Fujita, M.Q., Yoshikawa, H. and Ogasawara, N. (1992) Structure of some rplication origin. EMBO J. 19, 1138^1147.
the dnaA and DnaA-box region in the Mycoplasma capricolum chro- [144] Quon, K.C., Yang, B., Domian, I.J., Shapiro, L. and Marczynski,
mosome: Conservation and variations in the course of evolution. G.T. (1998) Negative control of bacterial DNA replication by a cell
Gene 110, 17^23. cycle regulatory protein that binds at the chromosome origin. Proc.
[126] Ye, F., Renaudin, J., Bove¤, J.-M. and Laigret, F. (1994) Cloning Natl. Acad. Sci. USA 95, 120^125.
and sequencing of the replication origin (oriC) of the Spiroplasma [145] Domain, I.J., Quon, K.C. and Shapiro, L. (1997) Cell-type speci¢c
citri chromosome and construction of autonomously replicating ar- phosphorylation and proteolysis of a transcriptional regulator con-
ti¢cial plasmids. Curr. Microbiol. 29, 23^29. trols the G1 -to-S transition in a bacterial cell cycle. Cell 90, 415^424.
[127] Salazar, L., Fsihi, H., de Rossi, E., Riccardi, G., Rios, C., Cole, S.T. [146] Gorbatyuk, B. and Marczynski, G.T. (2001) Physiological conse-
and Taki¡, H.E. (1996) Organization of the origins of replication of quences of blocked Caulobacter crescentus dnaA expression, an es-
the chromosomes of Mycobacterium smegmatis, Mycobacterium le- sential DNA replication gene. Mol. Microbiol. 40, 485^497.
prae and Mycobacterium tuberculosis and isolation of a functional [147] Marczynski, G.T. (1999) Chromosome methylation and measure-
origin from M. smegmatis. Mol. Microbiol. 20, 283^293. ment of faithful, once and only once per cell cycle chromosome
[128] Zawilak, A., Cebrat, S., Mackiewicz, P., Krol-Hulewicz, A., Jaki- replication in Caulobacter crescentus. J. Bacteriol. 181, 1984^1993.
mowicz, D., Messer, W., Gosciniak, G. and Zakrzewska-Czerwin- [148] Schaefer, C. and Messer, W. (1988) Termination of the E. coli asnC
ska, J. (2001) Identi¢cation of a putative chromosomal replication transcript. The DnaA protein/dnaA box complex blocks transcribing
origin from Helicobacter pylori and its interaction with the initiator RNA polymerase. Gene 73, 347^354.
protein DnaA. Nucleic Acids Res. 29, 2251^2259. [149] Braun, R.E., O’Day, K. and Wright, A. (1985) Autoregulation of
[129] Richter, S., Hess, W.R., Krause, M. and Messer, W. (1998) Unique the DNA replication gene dnaA in E. coli. Cell 40, 159^169.
organization of the dnaA region from Prochlorococcus marinus [150] Atlung, T., Clausen, E. and Hansen, F.G. (1985) Autoregulation of
CCMP1375, a marine cyanobacterium. Mol. Gen. Genet. 257, the dnaA gene of Escherichia coli. Mol. Gen. Genet. 200, 442^450.
534^541. [151] Ku«cherer, C., Lother, H., Ko«lling, R., Schauzu, M.A. and Messer,
[130] Mushegian, A.R. and Koonin, E.V. (1996) Gene order is not con- W. (1986) Regulation of transcription of the chromosomal dnaA
served in bacterial evolution. Trends Genet. Sci. 12, 289^290. gene of Escherichia coli. Mol. Gen. Genet. 205, 115^121.
[131] Moriya, S., Atlung, T., Hansen, F.G., Yoshikawa, H. and Ogasa- [152] Wang, Q. and Kaguni, J.M. (1987) Transcriptional repression of the
wara, N. (1992) Cloning of an autonomously replicating sequence dnaA gene of Escherichia coli by dnaA protein. Mol. Gen. Genet.
(ars) from the Bacillus subtilis chromosome. Mol. Microbiol. 6, 309^ 209, 518^525.
315. [153] Polaczek, P. and Wright, A. (1990) Regulation of expression of the
[132] Moriya, S., Firshein, W., Yoshikawa, H. and Ogasawara, N. (1994) dnaA gene in Escherichia coli: Role of the two promoters and the
Replication of a Bacillus subtilis oriC plasmid in vitro. Mol. Micro- DnaA box. New Biol. 2, 574^582.
biol. 12, 469^478. [154] Lee, Y.S. and Hwang, D.S. (1997) Occlusion of RNA polymerase by
[133] Ogura, Y., Imai, Y., Ogasawara, N. and Moriya, S. (2001) Auto- oligomerization of DnaA protein over the dnaA promoter of Esche-
regulation of the dnaA-dnaN operon and e¡ects of DnaA protein richia coli. J. Biol. Chem. 272, 83^88.
levels on replication initiation in Bacillus subtilis. J. Bacteriol. 183, [155] Smith, R.W.P., McAteer, S. and Masters, M. (1997) Autoregulation
3833^3841. of the Escherichia coli replication initiator protein, DnaA, is indi-
[134] Autret, S., Levine, A., Vannier, F., Fujita, Y. and Seror, S.J. (1999) rect. Mol. Microbiol. 23, 1303^1315.
The replication checkpoint control in Bacillus subtilis : identi¢cation [156] Del Solar, G.H., Giraldo, R., Ruiz-Echevarr|¤a, M.J., Espinosa, M.
of a novel RTP-binding sequence essential for the replication fork and D|¤az-Orejas, R. (1998) Replication and control of circular bac-
arrest after induction of the stringent response. Mol. Microbiol. 31, terial plasmids. Microbiol. Mol. Biol. Rev. 62, 434^464.
1665^1679. [157] Abeles, A.L. and Austin, S.J. (1988) P1 plasmid replication requires
[135] Jakimowicz, D., Majka, J., Lis, B., Konopa, G., Wegrzyn, G., Escherichia coli Dam-methylated DNA. Gene 74, 185^186.
Messer, W., Schrempf, H. and Zakrzewska-Czerwinska, J. (2000) [158] Abeles, A.L., Reaves, L.D. and Austin, S.J. (1990) A single dnaA
Structure and regulation of the dnaA promoter region in three box is su⁄cient for initiation from the plasmid P1 origin. J. Bacte-
Streptomyces species. Mol. Gen. Genet. 262, 1093^1102. riol. 172, 4386^4391.
[136] Zakrzewska-Czerwinska, J., Nardmann, J. and Schrempf, H. (1994) [159] Mukhopadhyay, G., Carr, K.M., Kaguni, J.M. and Chattoraj, D.K.

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374 373

(1993) Open-complex formation by the host initiator, DnaA, at the coliphage lambda DNA replication is regulated by the host DnaA
origin of P1 plasmid replication. EMBO J. 12, 4547^4554. initiator function. Gene 154, 47^50.
[160] Park, K. and Chattoraj, D.K. (2001) DnaA boxes in the P1 plasmid [180] Szalewska-Palasz, A., Wegrzyn, A., Blaszczak, A., Taylor, K. and
origin : The e¡ect of their position on the directionality of replica- Wegrzyn, G. (1998) DnaA-stimulated transcriptional activation of
tion and plasmid copy number. J. Mol. Biol. 310, 69^81. orilambda: Escherichia coli RNA polymerase L subunit as a tran-
[161] Doran, K.S., Helinski, D.B. and Konieczny, I. (1999) A critical scriptional activator contact site. Proc. Natl. Acad. Sci. USA 95,
DnaA box directs the cooperative binding of the Escherichia coli 4241^4246.
DnaA protein to the plasmid RK2 replication origin. J. Biol. [181] Baranska, S., Gabig, M., Wegrzyn, A., Konopa, G., Herman-Anto-
Chem. 274, 17918^17923. siewicz, A., Hernandez, P., Schvartzman, J.B., Helinski, D.R. and
[162] Konieczny, I., Doran, K.S., Helinski, D.R. and Blasina, A. (1997) Wegrzyn, G. (2001) Regulation of the switch from early to late
Role of TrfA and DnaA proteins in origin opening during initiation bacteriophage lambda DNA replication. Microbiology 147, 535^
of DNA replication of the broad host range plasmid RK2. J. Biol. 547.
Chem. 272, 20173^20178. [182] Glinkowska, M., Konopa, G., Wegrzyn, A., Herman-Antosiewicz,
[163] Konieczny, I. and Helinski, D.R. (1997) Helicase delivery and acti- A., Weigel, C., Seitz, H., Messer, W. and Wegrzyn, G. (2001) The
vation by DnaA and TrfA proteins during the initiation of replica- double mechanism of incompatibility between V plasmids and Es-
tion of the broad host range plasmid RK2. J. Biol. Chem. 272, cherichia coli dnaA(ts) host cells. Microbiology 147, 1923^1928.
33312^33318. [183] Boye, E., Lobner-Olesen, A. and Skarstad, K. (2000) Limiting DNA
[164] Pacek, M., Konopa, G. and Konieczny, I. (2001) DnaA box sequen- replication to once and only once. EMBO Rep. 1, 479^483.
ces as the site for helicase delivery during plasmid RK2 replication [184] Messer, W., Bellekes, U. and Lother, H. (1985) E¡ect of dam-meth-
initiation in Escherichia coli. J. Biol. Chem. 276, 23639^23644. ylation on the activity of the replication origin, oriC. EMBO J. 4,
[165] Filutowicz, M., Dellis, S., Levchenko, I., Urh, M., Wu, F. and 1327^1332.
York, D. (1994) Regulation of replication in an iteron-containing [185] Smith, D.W., Garland, A.M., Herman, G., Enns, R.E., Baker, T.A.
DNA molecule. Progr. Nucleic Acids Res. Mol. Biol. 68, 239^273. and Zyskind, J.W. (1985) Importance of state of methylation of oriC
[166] Miron, A., Mukherjee, S. and Bastia, D. (1992) Activation of dis- GATC sites in initiation of DNA replication in Escherichia coli.
tant replication origins in vivo by DNA looping as revealed by a EMBO J. 4, 1319^1326.
novel mutant form of an initiator protein defective in cooperativity [186] Russel, D.W. and Zinder, N.D. (1987) Hemimethylation prevents
at a distance. EMBO J. 11, 1205^1216. DNA replication in E. coli. Cell 50, 1071^1079.
[167] Lu, Y.B., Datta, H.J. and Bastia, D. (1998) Mechanistic studies of [187] Ogden, G.B., Pratt, M.J. and Schaechter, M. (1988) The replicative
initiator-initiator interaction and replication initiation. EMBO J. 17, origin of the E. coli chromosome binds to cell membranes only when
5192^5200. hemimethylated. Cell 54, 127^135.
[168] Bernander, R., Dasgupta, S. and Nordstro«m, K. (1991) The E. coli [188] Campbell, J.L. and Kleckner, N. (1990) E. coli oriC and the dnaA
cell cycle and the plasmid R1 replication cycle in the absence of the gene promoter are sequestered from dam methyltransferase follow-
DnaA protein. Cell 64, 1145^1153. ing the passage of the chromosomal replication fork. Cell 62, 967^
[169] Tang, X.B., Womble, D.D. and Rownd, R.H. (1989) DnaA protein 979.
is not essential for replication of IncFII plasmid NR1. J. Bacteriol. [189] Boye, E. and Lobner-Olesen, A. (1990) The role of dam methyl-
171, 5290^5295. transferase in the control of DNA replication in E. coli. Cell 62,
[170] Hansen, E.B. and Yarmolinsky, M.B. (1986) Host participation in 981^989.
plasmid maintenance: Dependence upon dnaA of replicons derived [190] von Freiesleben, U., Krekling, M.A., Hansen, F.G. and Lobner-
from P1 and F. Proc. Natl. Acad. Sci. USA 83, 4423^4427. Olesen, A. (2000) The eclipse period of Escherichia coli. EMBO J.
[171] Giraldo, R. and Diaz, R. (1992) Di¡erential binding of wild-type 19, 6240^6248.
and a mutant RepA protein to oriR sequence suggests a model for [191] Landoulsi, A., Hughes, P., Kern, R. and Kohiyama, M. (1989) dam
the initiation of plasmid R1 replication. J. Mol. Biol. 228, 787^802. methylation and the initiation of DNA replication on oriC plasmids.
[172] Ortega-Jime¤nez, S., Giraldo-Sua¤rez, R., Ferna¤ndez-Tresguerres, Mol. Gen. Genet. 216, 217^223.
M.E., Berzal-Herranz, A. and D|¤az-Orejas, R. (1992) DnaA depen- [192] Boye, E. (1991) The hemimethylated replication origin of Escheri-
dent replication of plasmid R1 occurs in the presence of point mu- chia coli can be initiated in vitro. J. Bacteriol. 173, 4537^4539.
tations that disrupt the dnaA box of oriR. Nucleic Acids Res. 20, [193] Lu, M., Campbell, J.L., Boye, E. and Kleckner, N. (1994) SeqA : a
2547^2551. negative modulator of replication initiation in E. coli. Cell 77, 413^
[173] Masai, H. and Arai, K.-I. (1987) RepA and DnaA proteins are 426.
required for initiation of R1 plasmid replication in vitro and interact [194] von Freiesleben, U., Rasmussen, K.V. and Schaechter, M. (1994)
with the oriR sequence. Proc. Natl. Acad. Sci. USA 84, 4781^4785. SeqA limits DnaA activity in replication from oriC in Escherichia
[174] Stenzel, T.T., MacAllister, T. and Bastia, D. (1991) Cooperativity at coli. Mol. Microbiol. 14, 763^772.
a distance promoted by the combined action of two replication [195] Shakibai, N., Ishidate, K., Reshetnyak, E., Gunji, S., Kohiyama, M.
initiator proteins and a DNA bending protein at the replication and Roth¢eld, L. (1998) High-a⁄nity binding of hemimethylated
origin of pSC101. Genes Dev. 5, 1453^1463. oriC by Escherichia coli membranes is mediated by a multiprotein
[175] Datta, H.J., Khatri, G.S. and Bastia, D. (1999) Mechanism of re- system that includes SeqA and a newly identi¢ed factor, SeqB. Proc.
cruitment of DnaB helicase to the replication origin of the plasmid Natl. Acad. Sci. USA 95, 11117^11121.
pSC101. Proc. Natl. Acad. Sci. USA 96, 73^78. [196] Slater, S., Wold, S., Lu, M., Boye, E., Skarstad, K. and Kleckner,
[176] Sharma, R., Kachroo, A. and Bastia, D. (2001) Mechanistic aspects N. (1995) E. coli SeqA protein binds oriC in two di¡erent methyl-
of DnaA-RepA interaction as revealed by yeast forward and reverse modulated reactions appropriate to its roles in DNA replication
two-hybrid analysis. EMBO J. 20, 4577^4587. initiation and origin sequestration. Cell 82, 927^936.
[177] Taylor, K. and Wegrzyn, G. (1995) Replication of coliphage lambda [197] Brendler, T. and Austin, S. (1999) Binding of SeqA protein to DNA
DNA. FEMS Microbiol. Rev. 17, 109^119. requires interaction between two or more complexes bound to sep-
[178] Kur, J., Gorska, I. and Taylor, K. (1987) Escherichia coli dnaA arate hemimethylated GATC sequences. EMBO J. 18, 2304^2310.
initiation function is required for replication of plasmids derived [198] Skarstad, K., Lueder, G., Lurz, R., Speck, C. and Messer, W. (2000)
from coliphage lambda. Mol. Gen. Genet. 198, 203^210. The Escherichia coli SeqA protein binds speci¢cally and coopera-
[179] Wegrzyn, G., Szalewska-Palasz, A., Wegrzyn, A., Obuchowski, M. tively to two sites in hemimethylated and fully methylated oriC.
and Taylor, K. (1995) Transcriptional activation of the origin of Mol. Microbiol. 36, 1319^1326.

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


374 W. Messer / FEMS Microbiology Reviews 26 (2002) 355^374

[199] Skarstad, K., Torheim, N., Wold, S., Lurz, R., Messer, W., Fossum, The initiation mass for DNA replication in Escherichia coli K-12 is
S. and Bach, T. (2001) The Escherichia coli SeqA protein binds dependent on growth rate. EMBO J. 13, 2097^2102.
speci¢cally to two sites in fully and hemimethylated oriC and has [217] Helmstetter, C.E., Cooper, S., Pierucci, D. and Revelas, E. (1968)
the capacity to inhibit DNA replication and a¡ect chromosome to- The bacterial life cycle. Cold Spring Harbor Symp. Quant. Biol. 33,
pology. Biochimie 83, 49^51. 809^822.
[200] Taghbalout, A., Landoulsi, A., Kern, R., Yamazoe, M., Hiraga, S., [218] Donachie, W.D. (2001) Co-ordinate regulation of the Escherichia
Holland, B., Kohiyama, M. and Malki, A. (2000) Competition be- coli cell cycle or The cloud of unknowing. Mol. Microbiol. 40,
tween the replication initiator DnaA the sequestration factor SeqA 779^785.
for binding to the hemimethylated chromosomal origin of E. coli in [219] Filutowicz, M., Ross, W., Wild, J. and Gourse, R.L. (1992) Involve-
vitro. Genes Cells 5, 873^884. ment of Fis protein in replication of the Escherichia coli chromo-
[201] Braun, R.E. and Wright, A. (1986) DNA methylation di¡erentially some. J. Bacteriol. 174, 398^407.
enhances the expression of one of the two E. coli dnaA promoters in [220] Filutowicz, M. and Roll, J. (1990) The requirement of IHF protein
vivo and in vitro. Mol. Gen. Genet. 202, 246^250. for extrachromosomal replication of the Escherichia coli oriC in a
[202] Theisen, P., Grimwade, J.E., Leonard, A.C., Bogan, J.A. and Helm- mutant de¢cient in DNA polymerase I activity. New Biol. 2, 818^
stetter, C.E. (1993) Correlation of gene transcription with the time 827.
of initiation of chromosome replication in Escherichia coli. Mol. [221] Polaczek, P., Kwan, K., Liberles, D.A. and Campbell, J.L. (1997)
Microbiol. 10, 575^584. Role of architectrual elements in combinatorial regulation of initia-
[203] Torheim, N.K. and Skarstad, K. (1999) Escherichia coli SeqA pro- tion of DNA replication in Escherichia coli. Mol. Microbiol. 26,
tein a¡ects DNA topology and inhibits open complex formation at 261^275.
oriC. EMBO J. 18, 4882^4888. [222] Polaczek, P. (1990) Bending of the origin of replication of E. coli by
[204] Weitao, T., Nordstro«m, K. and Dasgupta, S. (2000) Escherichia coli binding of IHF at a speci¢c site. New Biol. 2, 265^271.
cell cycle control genes a¡ect chromosome superhelicity. EMBO [223] Gille, H., Egan, J.B., Roth, A. and Messer, W. (1991) The FIS
Rep. 1, 494^499. protein binds and bends the origin of chromosomal DNA replica-
[205] Lobner-Olesen, A. (1999) Distribution of minichromosomes in indi- tion, oriC, of Escherichia coli. Nucleic Acids Res. 19, 4167^4172.
vidual Escherichia coli cells: implications for replication control. [224] Roth, A., Urmoneit, B. and Messer, W. (1994) Functions of histone-
EMBO J. 18, 1712^1721. like proteins in the initiation of DNA replication at oriC of Esche-
[206] Jensen, M.R., Lobner-Olesen, A. and Rasmussen, K.V. (1990) Es- richia coli. Biochimie 76, 917^923.
cherichia coli minichromosomes : Absence of copy number control, [225] Samitt, C.E., Hansen, F.G., Miller, J.F. and Schaechter, M. (1989)
and random segregation. J. Mol. Biol. 215, 257^265. In vivo studies of DnaA binding to the origin of replication of Es-
[207] Roth, A. and Messer, W. (1998) High-a⁄nity binding sites for the cherichia coli. EMBO J. 8, 989^993.
initiator protein DnaA on the chromosome of Escherichia coli. Mol. [226] Cassler, M.R., Grimwade, J.E. and Leonard, A.C. (1995) Cell cycle-
Microbiol. 28, 395^401. speci¢c changes in nucleoprotein complexes at a chromosomal rep-
[208] Kitagawa, R., Mitsuki, H., Okazaki, T. and Ogawa, T. (1996) A lication origin. EMBO J. 14, 5833^5841.
novel DnaA protein-binding site at 94.7 min on the Escherichia coli [227] Grimwade, J.E., Ryan, V.T. and Leonard, A.C. (2000) IHF redis-
chromosome. Mol. Microbiol. 19, 1137^1147. tributes bound initiator protein, DnaA, on supercoiled oriC of Es-
[209] Kitagawa, R., Ozaki, T., Moriya, S. and Ogawa, T. (1998) Negative cherichia coli. Mol. Microbiol. 35, 835^844.
control of replication initiation by a novel chromosomal locus ex- [228] Lobner-Olesen, A., Skarstad, K., Hansen, F.G., von Meyenburg, K.
hibiting exceptional a⁄nity for Escherichia coli DnaA protein. and Boye, E. (1989) The DnaA protein determines the initiation
Genes Dev. 12, 3032^3043. mass of Escherichia coli K-12. Cell 57, 881^889.
[210] Morigen, Boye, E., Skarstad, K. and Lobner-Olesen, A. (2001) Reg- [229] Hansen, F.G., Christensen, B.B. and Atlung, T. (1991) The initiator
ulation of chromosomal replication by DnaA protein availability in titration model : Computer simulation of chromosome and mini-
Escherichia coli : e¡ects of the datA region. Biochim. Biophys. Acta chromosome control. Res. Microbiol. 142, 161^167.
1521, 73^80. [230] Christensen, B.B., Atlung, T. and Hansen, F.G. (1999) DnaA boxes
[211] Skarstad, K., Boye, E. and Steen, H.B. (1986) Timing of initiation are important elements in setting the initiation mass of Escherichia
of chromosome replication in individual E. coli cells. EMBO J. 5, coli. J. Bacteriol. 181, 2683^2688.
1711^1717. [231] Lobner-Olesen, A., Hansen, F.G., Rasmussen, K.V., Martin, B. and
[212] Boye, E., Stokke, T., Kleckner, N. and Skarstad, K. (1996) Coor- Kuempel, P.L. (1994) The initiation cascade for chromosome repli-
dinating DNA replication initiation with cell growth: Di¡erential cation in wild-type and Dam methyltransferase de¢cient Escherichia
roles for DnaA and SeqA proteins. Proc. Natl. Acad. Sci. USA coli cells. EMBO J. 13, 1856^1862.
93, 12206^12211. [232] DePamphilis, M.L. (1996) Origins of DNA replication. In: DNA
[213] Helmstetter, C.E. and Leonard, A.C. (1987) Coordinate initiation of Replication in Eukaryotic Cells (DePamphilis, M.L., Ed.), pp. 45^
chromosome and minichromosome replication in Escherichia coli. 86. Cold Spring Harbor Laboratory Press, Cold Spring Harbor,
J. Bacteriol. 169, 3489^3494. NY.
[214] Skarstad, K., von Meyenburg, K., Hansen, F.G. and Boye, E. [233] Lee, D.G., Makhov, A.M., Klemm, R.D., Gri⁄th, J.D. and Bell,
(1988) Coordination of chromosome replication initiation in Esche- S.P. (2000) Regulation of origin recognition complex conformation
richia coli: e¡ects of di¡erent dnaA alleles. J. Bacteriol. 170, 852^ and ATPase activity: di¡erential e¡ects of single-stranded and dou-
858. ble-stranded DNA binding. EMBO J. 19, 4774^4782.
[215] Donachie, W.D. (1968) Relationship between cell size and time of [234] Weigel, W. and Seitz, H. (2002) Strand-speci¢c loading of DnaB
initiation of DNA replication. Nature 219, 1077^1079. helicase by DnaA to a substrate mimicking unwound oriC. Mol.
[216] Wold, S., Skarstad, K., Steen, H.B., Stokke, T. and Boye, E. (1994) Microbiol., in press.

FEMSRE 756 24-10-02 Cyaan Magenta Geel Zwart


Molecular Microbiology (2000) 37(3), 492±500

MicroReview

Plasmid copy number control: an ever-growing story

Gloria del Solar* and Manuel Espinosa although they are likely to constitute a slight metabolic
Centro de Investigaciones BioloÂgicas, CSIC, VelaÂzquez, burden to the host. To co-exist stably with their hosts and
144, E-28006 Madrid, Spain. to minimize the metabolic load, plasmids must control
their replication, so that the copy number (N) of a given
plasmid is usually fixed within a given host and under
Summary
defined cell growth conditions.
Bacterial plasmids maintain their number of copies It is convenient to distinguish two different stages of the
by negative regulatory systems that adjust the rate of plasmid life cycle. The first, termed establishment, occurs
replication per plasmid copy in response to fluctua- when a plasmid copy enters a new permissive host. A
tions in the copy number. Three general classes of successful establishment may depend upon the plasmid's
regulatory mechanisms have been studied in depth, ability to replicate rapidly before the host divides (High-
namely those that involve directly repeated lander and Novick, 1987). This may result in an overshoot
sequences (iterons), those that use only antisense in N before it reaches the characteristic value (Nav). In the
RNAs and those that use a mechanism involving an second stage, the replicon enters into the steady state in
antisense RNA in combination with a protein. The which Nav is maintained because, on average, there is
first class of control mechanism will not be discussed one replicative event per plasmid copy and cell cycle
here. Within the second class (the most `classical' (NordstroÈm and Wagner, 1994). To maintain this average
one), exciting insights have been obtained on the rate, plasmids use self-encoded negative control systems
molecular basis of the inhibition mechanism that that are able to `sense' and correct up and down
prevents the formation of a long-range RNA structure fluctuations from the Nav in individual cells (Summers,
(pseudoknot), which is an example of an elegant 1996). These control systems adjust the rate of replication
solution reached by some replicons to control their per plasmid copy, so that it becomes higher or lower than
copy number. Among the third class, it is possible to 1, depending on whether there has been a decrease or an
distinguish between (i) cases in which proteins play increase, respectively, in copy number with respect to the
an auxiliary role; and (ii) cases in which transcrip- Nav in a given cell. Thus, the N-values from individual cells
tional repressor proteins play a real regulatory role. of a population should follow a narrow Gaussian distribu-
This latter type of regulation is relatively new and tion when the regulatory circuits are functioning under
seems to be widespread among plasmids from Gram- optimal conditions. This would not be the case for a
positive bacteria, at least for the rolling circle- replicon lacking specific control functions, such as the
replicating plasmids of the pMV158 family and the oriC minichromosomes (the cloned origin of replication of
theta-replicating plasmids of the Inc18 streptococcal Escherichia coli). Narrow distribution of N in several
family. plasmids and broad deviations in oriC minichromosomes
have been measured by a combination of flow cytometry
and plasmid-driven expression of the green fluorescent
Introduction protein (Lùbner-Olesen, 1999).
There are three general types of plasmid copy number
Bacterial plasmids are extrachromosomal genomes that control systems, depending on the type of negative
replicate autonomously and in a controlled manner. Many control element used: (i) directly repeated sequences
plasmids are self-transmissible or mobilizable by other (iterons) that complex with cognate replication (Rep)
replicons, thus having the ability to colonize new bacterial initiator proteins; (ii) antisense RNAs that hybridize to a
species. Plasmids may provide the host with valuable complementary region of an essential RNA, therefore
functions, such as drug resistance(s) or metabolic path- termed countertranscribed (ct) RNAs; and (iii) ctRNA and
ways useful under certain environmental conditions, a protein. Within this last group, there are two categories.
Accepted 9 May, 2000. *For correspondence. E-mail gdelsolar@ In one of them, the ctRNA plays the main regulatory role,
cib.csic.es; Tel. (134) 91 561 1800; Fax (134) 91 562 7518. whereas the protein has been proposed as only an
Q 2000 Blackwell Science Ltd
Control of plasmid replication 493

I Fig. 1. Control of replication by pseudoknot


formation in ColIb-P9. Genes repY (for leader
peptide) and repZ (for initiator of replication)
are translationally coupled. On the mRNA, the
Shine±Dalgarno sequence (SD) of repY is
III exposed, whereas the SD and the initiation
start repZ end repZ codon of repZ can be occluded within
SD repZ
structure III. If there is no interaction with Inc
RNA (left), repY translation takes place.
rep mRNA Stalled ribosomes at the end of repY unfold
SD structure III, allowing the formation of the
repY repY
repZ pseudoknot between the region located on the
loop of structure I (double lines) and its
start repY end repY Replication complementary region (stippled rectangle).
This facilitates binding of the ribosomes
(ellipses) to the repZ SD sequences, followed
by translation of repZ. The antisense Inc RNA
(right) would hinder pseudoknot formation and
Inc RNA translation of repY, leading to inhibition of
repZ translation. Events and elements acting
repY translation positively in replication are indicated in blue,
whereas negative ones are depicted in red.

unfolding of structure III Inc RNA


INHIBITION OF repZ TRANSLATION
tion
la
trans

I
T
O
repZ

KN
O
D

III
EU
PS

I
O
T
O

N
KN
O
D
EU
PS

Inc RNA-mRNA duplex formation


Inhibition of repY translation

auxiliary element. Within the second category, both they will be found to be more widespread. Finally, a single
elements, acting on different targets, could correct instance of a new control mechanism, involving only the
fluctuations in the N-value at the steady state (del Solar Rep protein in plasmids lacking iterons, will be discussed
et al., 1995; 1998). Replication control by iterons will be briefly (Burian et al., 1999).
covered in the accompanying review by Chattoraj, this
issue pp. 467±476. Here, we will consider the other two
Control by ctRNAs
types of mechanisms of replication control, focusing on
cases in which significant advances have recently been Control systems using ctRNAs (Wagner and Brantl, 1998)
achieved. The first case, including plasmids that use only are widespread within plasmids replicating by different
ctRNA as a control element, involves the inhibition of the mechanisms, but sharing a similar genetic structure in the
formation of a long-range RNA structure (the pseudo- control region: two oppositely oriented promoters direct,
knot), which actively enhances translation of an essential respectively, the synthesis of an RNA essential for
replication initiator gene. The second focus will be on replication and of the inhibitor ctRNA. The ctRNAs are
those systems that include proteins playing an auxiliary complementary to a region (the target) near the 5 0 end of
regulatory role. Next, we will pay attention to those the essential RNA. An important feature of this kind of
replicons with two plasmid-encoded control elements. control system is that the rate of synthesis of the inhibitor
There are few reports on plasmids using this last control ctRNA is much higher than that of the essential RNA. In
mechanism but, considering their efficiency, it is likely that addition, the ctRNAs are synthesized from a constitutive
Q 2000 Blackwell Science Ltd, Molecular Microbiology, 37, 492±500
494 G. del Solar and M. Espinosa

promoter and have a short half-life, so that their region in the loop of structure I. Appropriate termination of
intracellular concentration stays nearly proportional to N. translation of the leader repY gene unfolds structure III,
Replication is inhibited by RNA±ctRNA pairing and thus inducing the formation of the pseudoknot by
abolition of the essential RNA activity. Differences intramolecular pairing of loop I with its complementary
between various replicons regulated by ctRNAs are sequence. Pseudoknot formation facilitates the binding of
found based on how the inhibition occurs and can be the ribosomes to the Shine±Dalgarno sequence of the
exemplified as follows: (i) inhibition of maturation of the essential repZ gene. Mutations causing disruption of
primer essential for replication, as in plasmid ColE1 structure III have allowed the mapping of the pseudoknot
(Tomizawa and Itoh, 1981); (ii) premature termination of in vitro (Asano and Mizobuchi, 1998b). Translation of repY
the synthesis of the essential rep-mRNA (pT181; Novick and pseudoknot formation are inhibited by the interaction
et al., 1989); and (iii) inhibition of rep translation. This last of the ctRNA (Inc RNA) with the complementary region of
method of ctRNA-dependent copy number control can be the rep mRNA. Generation of the duplex Inc RNA±target
achieved by three mechanisms, namely inhibition of mRNA occludes the repY ribosome binding site, leading
translation of a leader peptide (plasmid R1; Blomberg to an indirect inhibition of repZ translation. In addition,
et al., 1992), inhibition of both translation of a leader pairing between the single-stranded loops on the com-
peptide and formation of an activator pseudoknot (ColIb- plementary structures of rep mRNA (structure I) and Inc
P9; Asano et al., 1991; Asano and Mizobuchi, 1998a) and RNA is sufficient to block generation of the pseudoknot,
direct inhibition of translation of the essential rep gene. A so that Inc RNA can repress repZ translation at the level
direct inhibition mechanism has been proposed for of a transient interaction with its target before a stable
plasmid ColE2, in which the formation of a stable complex duplex is detected. The same region in the loop of
between the ctRNA and the complementary region in the structure I of the rep mRNA is involved in the initial
rep mRNA would be able to inhibit rep expression, even interactions that take place in pseudoknot formation and
though the ctRNA does not overlap the putative transla- in Inc RNA binding. These initial interactions are
tion initiation region of the essential gene (T. Itoh, stimulated by the presence of a hexanucleotide (con-
personal communication). served in various antisense systems; Asano and
For a number of plasmids, studies on the kinetics of Mizobuchi, 1998a), which presumably supports the
interaction between ctRNAs and their RNA targets have element termed U-turn and includes the structure I
shown that stable duplex formation is not required for sequence involved in the initial interactions. The U-turn
inhibition of rep expression, as this inhibition takes place loop structures are general binding rate enhancers that
faster than stable binding (reviewed by Wagner and facilitate rapid RNA±RNA interactions (reviewed by
Brantl, 1998). In contrast, a recent report on the ctRNA± Franch and Gerdes, 2000). As the early stages in
rep mRNA interactions of the staphylococcal plasmid pseudoknot formation and in the binding of the inhibitory
pT181 shows that the rate constant of stable complex RNA are similar, an explanation as to how these two
formation is similar to the inhibition rate constant (Brantl processes can compete with each other is easily deduced
and Wagner, 2000). (Asano and Mizobuchi, 1998a). Although Inc RNA
New findings on the molecular and biochemical bases represses the translation of repY and repZ genes,
for the control mechanism involving an intramolecular repression of the former is much less efficient than that
RNA±RNA interaction within the leader region of the rep of the latter. Repression of repZ and repY expression are
mRNA have been provided for the IncIa ColIb-P9 and the accomplished at different stages during the pairing
IncL/M pMU604 plasmids (Asano and Mizobuchi, 1998a; between Inc RNA and rep mRNA (Asano and Mizobuchi,
2000; Athanasopoulos et al., 1999). Replication depends 2000). This differential repression allows the Inc RNA to
on the expression of the essential rep gene, which keep the total level of repZ expression constant. A
requires coupled translation of a gene encoding a leader constant total rate of synthesis of the initiator was
peptide and the formation of a pseudoknot by intra- demonstrated early for IncFII plasmids (Nielsen and
molecular pairing of two complementary sequences of the Molin, 1984). As the level of repZ expression is rate-
rep mRNA (Asano et al., 1991; Wilson et al., 1993). limiting for replication, the Inc RNA-based regulation
The model for the control of replication of ColIb-P9 mechanism maintains a constant N-value.
(reviewed by Wagner and Simons, 1994) involves two A similar genetic structure and control mechanism were
stem±loop structures in the repZ mRNA that have been shown to exist in the IncB plasmid pMU720, closely
mapped in vitro (Asano and Mizobuchi, 1998b) and are related to the IncIa-ColIb-P9 (Siemering et al., 1994;
located upstream (structure I) and in the middle (structure Wilson et al., 1994). However, some differences are found
III) of the leader repY gene (Fig. 1). Structure III occludes in plasmid pMU604, belonging to the IncL/M group
both the translation initiation sequences of the essential (Athanasopoulos et al., 1999). In this case, replication
repZ gene and a short sequence complementary to a control also involves translational coupling of a leader

Q 2000 Blackwell Science Ltd, Molecular Microbiology, 37, 492±500


Control of plasmid replication 495
pseudoknot formation inhibited translation of the leader
CopB Tap RepA gene in pMU604 (Athanasopoulos et al., 1999), a feature
ori that has not been reported to exist in ColIb-P9. The
existence of such complex regulatory circuits in other
P1 P2
plasmids awaits further investigation.
CopA

Fig. 2. Auxiliary elements controlling plasmid copy number: the R1 Accessory proteins as inhibitory elements
paradigm. The initiator RepA protein acts on the origin of replication
(ori) located downstream of the repA gene. Expression of repA is Plasmids R1 and ColE1, which control their N-values by
translationally coupled to that of tap (encoding a small leader
ctRNAs, also encode a protein as a second inhibitory
peptide). CopB protein represses transcription from promoter P2,
so that this promoter is normally silent. Transcription of the copB± element that, however, plays only an auxiliary role. In the
tap±repA mRNA is mainly directed by the constitutive weak case of R1, expression of the essential gene repA
promoter P1. The antisense RNA, CopA, acts by blocking the
requires the translation of the leader gene tap, which is
translation of tap. Negatively acting elements are depicted in red;
those acting positively are shown in blue. translationally coupled to repA (Blomberg et al., 1992).
The main replication control element is the ctRNA, CopA,
gene and the formation of a pseudoknot, although, unlike which inhibits translation of tap and, indirectly, that of
ColIb-P9, the positioning of sequences involved in the repA. The second inhibitory element of R1 is the product
expression of the essential gene repA is different. of the copB gene, which is co-transcribed with tap and
The most important variation is the spacer between the repA from promoter P1 (Fig. 2). CopB protein is a
pseudoknot and the translation initiation region of the transcriptional repressor of a second promoter, P2,
essential repA gene, which seems to be suboptimal in located downstream of copB, which directs the synthesis
pMU604. Mutational analyses showed that the require- of a tap±repA mRNA. At steady state, CopB is present at
ment for pseudoknot formation in pMU604 could be saturating concentrations, blocking transcription from P2,
partially replaced by improving the initiation of translation so that repA is expressed almost exclusively from P1.
signals of the essential gene repA. Interestingly, Deletion of the entire copB gene (including promoter P1)
results in plasmids with an eightfold increase in N (Riise
RNA II pre-primer
et al., 1982). The CopB regulatory loop has been
suggested to serve as a rescue mechanism that prevents
RNAP Origin rom plasmid loss in newborn cells harbouring very few plasmids.
RNA I
Rom However, computer simulation of mini-R1 plasmid replica-
tion indicated that the CopB regulatory circuit contributes
RNA I little to the stability of these replicons (Rosenfeld and
Origin
Grover, 1993).
The second instance of plasmids with auxiliary proteins
is ColE1 (Fig. 3). In this case, replication is mediated by
RNase H the synthesis of a preprimer RNA (RNA II) by the host
RNA I
Rom RNA polymerase (RNAP) and the formation of a DNA±
RNA hybrid between the RNA II and the template DNA
Primer maturation No DNA-RNA hybrid strand at the origin region. This hybrid is cleaved by
RNase H, generating a 3 0 -OH end, which is used by DNA
polymerase I to initiate leading strand synthesis. The
No primer maturation availability of the primer 3 0 -OH end is rate-limiting for
Replication
initiation, and it is modulated by the ctRNA I control
element. Interaction between ctRNA I and its comple-
Replication inhibited mentary region in the preprimer alters the secondary
structure of the latter, leading to the inhibition of stable
Fig. 3. Copy number control in ColE1. Synthesis of the preprimer
RNA II by RNAP (stippled circle) is essential for replication. In the DNA±RNA hybrid formation. This, in turn, leads to
absence of interaction with the RNA I (left), the RNA II forms a inhibition of replication. The second element of this circuit
stable hybrid with the template DNA at the origin of replication. This is protein Rom (Rop), which enhances the rate of
hybrid is cleaved by RNase H to generate the 3 0 -OH end of the
RNA primer, from which replication starts. Interaction between the formation of a stable complex between the ctRNA I and
inhibitor RNA I and the complementary region in the RNA II the preprimer RNA. Rom does not seem to be an
preprimer (right) is aided by Rom protein (ellipse). RNA I±RNA II essential component of the ColE1 control system.
interaction inhibits the formation of the DNA±RNA II hybrid at the
origin region, preventing maturation of the RNA II into the Deletion of the rom gene leads to a two- to threefold
replication primer. Colours as in the legends to Figs 1 and 2. increase in N in slowly growing cells, but it has no
Q 2000 Blackwell Science Ltd, Molecular Microbiology, 37, 492±500
496 G. del Solar and M. Espinosa

pMV158 Fig. 4. Dual regulation of plasmid copy


number, as exemplified by pMV158 (top) and
by two members of the Inc18 plasmid family
(middle and bottom). In pMV158, the initiator
protein, RepB, acts on the leading strand
CopG RepB origin, dso. The CopG repressor protein
blocks transcription from promoter Pcr, which
directs synthesis of the copG±repB mRNA, so
that CopG autoregulates its own synthesis.
dso Pcr Pctll From promoter PctII, the ctRNA II is
RNA II synthesized, which, in turn, blocks translation
of repB. In the case of pIP501, the initiator
RepR protein acts on the origin (oriR) located
downstream of repR. The transcriptional
repressor CopR regulates transcription from
pII, which is the only promoter that directs the
expression of repR. CopR is synthesized from
promoter pI, which would be constitutive.
From promoter pIII, the long-lived inhibitor
pIP501
ctRNA III is synthesized. Interaction of ctRNA
CopR RepR III with its target in the leader region of the
+ oriR repR mRNA induces (%) generation of an
mRNA secondary structure, which acts as a
plII transcriptional attenuator, avoiding repR
pl plI
expression. CopR-dependent inhibition of pII
RNA III + increases transcription from pIII (%) by
alleviating the effect of convergent
transcription. The replication and control
regions of pSM19035 share similarities in the
genes, promoters and regulatory circuits to
those of pIP501. The segB region is shown as
pSM19035 composed of three genes, their expression
being controlled by a single promoter (Pv).
The product of gene v regulates this operon
and also the promoter of gene copS. The
CopS RepS main regulatory circuits would be the same as
+ oriS ω ε ζ in pIP501 (Brantl et al., 1990). Colours as in
the legends to previous figures.
Pcop1 Prep P
III Pω
RNA +

phenotypic consequences on the N-value in fast-growing experimentally. Secondly, Rom would act by making the
bacteria (Atlung et al., 1999). When cloned on a probability of plasmid replication very close to zero at high
compatible multicopy plasmid, the rom gene is able to ctRNA I concentration because, in the absence of Rom,
complement a rom2 derivative, although it has no further the intrinsic rate of ctRNA I±RNA II duplex formation
effect upon the replication of a co-resident ColE1. The would be too slow to ensure total inhibition of replication.
absence of incompatibility caused by extra copies of rom Thus, Rom would ensure an efficient copy number control
shows that Rom is not a primary inhibitor of ColE1 system. Thirdly, Rom could act as a back-up system when
replication, as it exerts its maximum effect at the wild-type N (and, subsequently, Rom concentration) is greatly
concentration (Summers, 1996). Mathematical models of reduced: under normal conditions, the replication fre-
the dynamic features of copy number control in ColE1 and quency would not depend on small deviations in Rom
the experimental observations about the small effect concentration but, if this concentration decreases greatly
caused by variations in the dosage of the rom gene have (as a result of a large reduction in N), inhibition of primer
left open the question of why there is a Rom protein formation would decrease, thus leading to an increase in
(Paulson et al., 1998). At least three theoretical proposals the replication frequency. However, and as far as we are
have been made to account for an important role of Rom aware, no experiments have been performed to clarify
in the dynamics of ColE1 copy number control (Paulson these hypotheses. On the other hand, experimental
et al., 1998). First, Rom concentration would be propor- evidence has shown that the presence of ColE1 deriva-
tional to the N-value, so that the response in replication tives lacking rom reduced bacterial growth in medium
frequency to variations in the N-value would be sharper impoverished in carbon sources, whereas rom1 deriva-
than if RNA I acted alone. This hypothesis requires that tives did not show an adverse effect on cell growth (Atlung
Rom is rapidly degraded, which has not been tested et al., 1999). This is thought to be related to the

Q 2000 Blackwell Science Ltd, Molecular Microbiology, 37, 492±500


Control of plasmid replication 497
amplification of ColE1-type plasmids in slowly growing suggest the importance of derepression of the CopG-
cells, which is higher in plasmids lacking rom. From these regulated Pcr promoter to overcome severe decreases in
observations, a key role for Rom protein has been N (del Solar et al., 1995). Owing to its autorepressor
envisaged, that is to prevent ColE1-type plasmids from capacity, CopG would keep the level of synthesis of the
representing a metabolic burden to their hosts in natural cop±rep mRNA within narrow limits. Variable concen-
habitats, in which cells grow slowly because of nutrient trations of RNA II (the concentration depending upon the
limitation. N-value because of the constitutive synthesis of RNA II
In conclusion, CopB and Rom are inhibitory elements and of its short half-life) would act on these practically
that limit plasmid replication, as their inactivation leads to constant levels of mRNA. A similar genetic structure
an increase in the N-value of R1 or ColE1 respectively. exists in all plasmids from the pMV158 family.
However, the genes encoding these proteins do not Three closely related streptococcal plasmids belonging
constitute an incompatibility determinant against the wild- to the Inc18 family (pIP501, pAMb1 and pSM19035) have
type plasmid, so that both proteins would be auxiliary been studied. In the case of pIP501, control of N involves
negative elements unable to correct up-fluctuations in N. the transcriptional repressor CopR and the antisense
However, the influence of these proteins on the dynamics RNA III (Fig. 4). Promoter pI directs the expression of
of plasmid copy number control remains to be solved. gene copR, whose product inhibits transcription from
promoter pII located downstream of pI (Brantl, 1994).
CopR-mediated repression does not completely inhibit
Dual regulation by ctRNA and inhibitory protein
transcription from pII, which is the only promoter that
A novel mode of regulation of replication, involving a directs the expression of gene repR, essential for plasmid
ctRNA and a transcriptional repressor protein, has been replication. A third promoter, pIII, directs the synthesis of
shown for two plasmid families represented by pMV158 RNA III, a ctRNA that inhibits the expression of repR by a
(del Solar and Espinosa, 1992) and pIP501 (Brantl, 1994). mechanism of transcriptional attenuation (Brantl et al.,
Unlike plasmids controlling their replication only by 1993), similar to that postulated for plasmids pAMb1 (Le
ctRNAs, the promoter directing the expression of the Chatelier et al., 1996) and pT181 (Novick et al., 1989), so
essential rep gene in these plasmids is not constitutive, that interaction between repR mRNA and RNA III leads to
but regulated by a Cop protein. Although the dual circuits premature termination of the mRNA synthesis. Interest-
of control of N in these two plasmids share similarities, ingly, RNA III has an unusually long half-life of about
significant differences exist with respect to the features 30 min, which would make it unable to correct down-
and inhibitory mechanisms of both regulatory elements. fluctuations in N rapidly. To explain how the control
In the case of pMV158, characterization of mutations system of pIP501 copes with a reduction in N, a dual
that result in increased N-values and definition of DNA function has been assigned to CopR (Brantl and Wagner,
regions (inc determinants) that affect plasmid replication 1997). A decrease in N would result in a decrease in
in trans showed a third general mechanism of replication CopR concentration, which would lead to derepression of
control involving the transcriptional repressor protein promoter pII and to the concomitant increase in the
CopG and the ctRNA (RNA II), both involved in regulation expression of repR. In addition, transcription from
of synthesis of the initiator RepB protein (del Solar et al., promoter pIII is inhibited by the increased convergent
1995). Gene copG is co-transcribed with repB from a transcription from pII, so that the slow reduction in the
single promoter (Pcr), and the copG product binds to a amounts of intracellular stable RNA III inhibitor could be
DNA region that includes promoter Pcr, thus inhibiting slightly accelerated.
transcription of the cop±rep operon (Fig. 4). Competition In the case of plasmid pAMb1, the situation may be
between purified CopG and RNAP proteins indicates that more complex. The repressor CopF (equivalent to CopR)
CopG impairs the binding of RNAP to promoter Pcr (our not only inhibits RepF (equivalent to RepR) synthesis, but
unpublished observations). CopG protein is very stable, at it may also decrease primer formation, as the activating
least when overproduced in Escherichia coli. Promoter transcription fork that passes through the origin of
PctII directs the synthesis of RNA II, which has a short replication originates from the repF promoter (L. JannieÁre
half-life (<2 min, G. del Solar, unpublished) and seems to in Espinosa et al., 2000). A further degree of complexity
inhibit repB expression by direct pairing with the transla- arises from new findings on plasmid pSM19035. In this
tion initiation signals of repB. Both CopG and RNA II are plasmid, part of the segB region (involved in better-than-
able to sense and correct up-fluctuations in the N-value at random segregation) contains genes v , 1 and z (Fig. 4).
the steady state, and together constitute an inc determi- These genes are organized in an operon controlled by the
nant much more powerful than each element alone. This product of gene v . Purified Omega protein not only binds
synergistic effect argues in favour of a primary role for to its promoter region, but also to a DNA fragment
both elements in pMV158 control of N and allowed us to containing the promoter region of gene copS (the
Q 2000 Blackwell Science Ltd, Molecular Microbiology, 37, 492±500
498 G. del Solar and M. Espinosa
homologue of pIP501-copR and pAMb1-copF). Conse- Whether this mechanism is widespread among small
quently, it would appear that plasmid partition and plasmid plasmids of enterobacteria remains to be determined.
copy number control are under a common pathway of
regulation (de la Hoz et al., 2000).
Perspectives
There is a fundamental difference between the control
mechanisms governed only by ctRNA and those The recently reported new mechanisms controlling
governed by dual regulators. In the former, the rate of plasmid copy number show that these processes are
synthesis per plasmid copy of the essential RNA needed more complex than previously envisaged. Pseudoknot
for replication is constant but rather low compared with formation, known for a number of years, has only recently
that of the ctRNA. In the latter, expression of the rep gene been characterized in some depth. Mechanisms control-
is directed by a strong and Cop-regulated promoter so ling the replication of newly described replicons, espe-
that, when the regulatory protein does not operate, there cially those isolated from extremophiles, are mostly
is a high rate of rep transcription. This seems to be at unknown, and a novel control mechanism (still to be
least the case for the pMV158-Pcr promoter, which is the explored in some depth), independent of iterons and
strongest promoter of the plasmid (our unpublished ctRNAs, seems to exist in small cryptic plasmids of E. coli.
results). A high potential to transcribe the essential rep New and exciting findings will be derived from a general
gene would represent an advantage for these plasmids mechanism for interactions between antisense and target
during the establishment stage, as they would replicate RNAs in prokaryotes. This mechanism involves rapid
rapidly to reach Nav, thus decreasing the frequency of interaction between both RNAs mediated by a motif (Pyr-
appearance of plasmid-free cells from newly colonized U-N-Pur, the so-called U-turn) that is conserved in RNA
bacteria. We have preliminary evidence suggesting that recognition loops in either the antisense or the target
the efficiency of establishment of pMV158 derivatives is RNAs in many antisense RNA-regulated gene systems
influenced by the activity of the promoter directing the (Franch et al., 1999; Franch and Gerdes, 2000). However,
expression of the repB gene. It is worth pointing out that and in spite of the knowledge accumulated during recent
plasmids from the Inc18 family and several replicons of years, there is a lack of structural studies on the elements
the pMV158 family are mobilizable, and that these involved in the regulation of replication, with a few
replicons have a very broad host range. Plasmids with exceptions. The structure of pMV158-CopG repressor
auxiliary proteins involved in their control may also share has been solved both alone and in complex with its target
the same advantage during the establishment stage DNA, and it has been shown that the CopG dimer has a
(Summers, 1996). ribbon±helix±helix structure, resembling that of the Arc
repressor of phage P22 (Gomis-RuÈth et al., 1998).
Modelling of the first 62 residues of the pIP501-CopR
Non-iteron non-ctRNA plasmid control?
repressor indicates that it belongs to the helix±turn±helix
It seems that a new mechanism for copy number control family of DNA-binding proteins (Steinmetzer et al., 2000),
independent of antisense RNA and iterons operates in and preliminary X-ray diffraction data are available from
small cryptic plasmids of E. coli, with the Rep protein the pSM19035-Omega repressor (Murayama et al.,
being the only plasmid-encoded factor involved in initia- 1999). In addition, a three-dimensional model of the
tion and repression of replication (Burian et al., 1999). The antisense RNA of plasmid R1, together with in vitro
copy number control (cop) region, which also constitutes structural determinations, have revealed that the anti-
an inc determinant, includes the rep promoter and two sense-mRNA binding product is not a full duplex, but a
Rep binding sites (BD1 and BD2). BD1 is close to the complicated extended complex that is stabilized by distal
replication initiation region and seems to bind preferen- basepairings (Kolb et al., 2000). Determination of the
tially monomers/dimers of the Rep protein, whereas BD2 crystal structure of this antisense RNA may throw new
is adjacent to the rep promoter region and would appear light on the nature of the contacts between the two RNA
to bind Rep oligomers preferentially. Initiation of replica- species. Finally, and in spite of the early characterization
tion would require binding of the Rep protein to both of auxiliary proteins such as CopB or Rom, there seems
binding sites. Increases in N, and subsequent elevation of not to be enough information on the precise role of these
the Rep concentration, would favour the formation of Rep proteins in replication control at the steady state and
oligomers, displacing Rep monomers/dimers from BD2, during the plasmid establishment stage.
thereby autorepressing rep transcription. Thus, control of
replication is proposed to be exerted by the monomer±
multimer Rep equilibrium and, unlike the iteron-based Acknowledgements
control systems, incompatibility in these replicons would Thanks are due to D. Chattoraj for his critical reading of the
easily be over-ridden by an excess of Rep protein. manuscript, and T. Itoh for communicating his unpublished

Q 2000 Blackwell Science Ltd, Molecular Microbiology, 37, 492±500


Control of plasmid replication 499
results. The authors' laboratories were financed by CICYT (ed.). Amsterdam: Harwood Academic Publishers, pp. 1±
(grants BIO97-0347 and 2FD97-0518, to M.E.) and by 47.
Comunidad AutoÂnoma de Madrid (grant 07B/0049/1999 to Franch, T., Petersen, M., Wagner, E.G.H., Jacobsen, J.P.,
G.d.S.). and Gerdes, K. (1999) Antisense RNA regulation in
prokaryotes: rapid RNA/RNA interaction facilitated by a
general U-turn loop structure. J Mol Biol 294: 1115±1125.
References Franch, T., and Gerdes, K. (2000) U-turns and regulatory
Asano, K., and Mizobuchi, K. (1998a) Copy number control of RNAs. Curr Opin Microbiol 3: 159±164.
IncIa plasmid ColIb-P9 by competition between pseudo- Gomis-RuÈth, F.X., SolaÂ, M., Acebo, P., PaÂrraga, A., Guasch,
knot formation and antisense RNA binding at a specific A., Eritja, R., et al. (1998) The structure of plasmid-
RNA site. EMBO J 17: 5201±5213. encoded transcriptional repressor CopG unliganded and
Asano, K., and Mizobuchi, K. (1998b) An RNA pseudoknot as bound to its operator. EMBO J 17: 7404±7415.
the molecular switch for translation of the repZ gene Highlander, S.K., and Novick, R.P. (1987) Plasmid repopulation
encoding the replication initiator of IncIa plasmid ColIb-P9. kinetics in Staphylococcus aureus. Plasmid 17: 210±221.
J Biol Chem 273: 11815±11825. de la Hoz, A.B., Ayora, S., Sitkiewicz, I., Pankiewicz, R.,
Asano, K., and Mizobuchi, K. (2000) Structural analysis of Alonso, J.C., and Ceglowski, P. (2000) Plasmid copy-
late intermediate complex formed between plasmid ColIb- number control and better-than-random segregation genes
P9 Inc RNA and its target RNA. How does a single of pSM19035 share a common regulator. Proc Natl Acad
antisense RNA repress translation of two genes at different Sci USA 97: 728±733.
rates? J Biol Chem 275: 1269±1274. Kolb, F.A., Malmgren, C., Westhof, E., Ehresmann, C.,
Asano, K., Moriwaki, H., and Mizobuchi, K. (1991) Positive Ehresmann, B., Wagner, E.G.H., et al. (2000) An unusual
and negative regulations of plasmid ColIb-P9 repZ gene structure formed by antisense-target RNA binding involves
expression at the translational level. J Biol Chem 266: an extended kissing complex with a four-way junction and a
3774±3781. side-by-side helical alignment. RNA 6: 311±324.
Athanasopoulos, V., Praszkier, J., and Pittard, A.J. (1999) Le Chatelier, E., Ehrlich, S.D., and JannieÁre, L. (1996)
Analysis of elements involved in pseudoknot-dependent Countertranscript-driven attenuation system of the pAMb1
expression and regulation of the repA gene of an IncL/M repE gene. Mol Microbiol 20: 1099±1112.
plasmid. J Bacteriol 181: 1811±1819. Lùbner-Olesen, A. (1999) Distribution of minichromosomes in
Atlung, T., Christensen, B.B., and Hansen, F.G. (1999) Role individual Escherichia coli cells: implications for replication
of the Rom protein in copy number control of plasmid control. EMBO J 18: 1712±1721.
pBR322 at different growth rates in Escherichia coli K-12. Murayama, K., de la Hoz, A.B., Alings, C., LoÂpez, G., Orth,
Plasmid 41: 110±119. P., Alonso, J.C., et al. (1999) Crystallization and pre-
Blomberg, P., Nordstrom, K., and Wagner, E.G.H. (1992) liminary X-ray diffraction studies of Streptococcus pyo-
Replication control of plasmid R1: RepA synthesis is genes plasmid pSM19035-encoded v transcriptional
regulated by CopA RNA through inhibition of leader peptide repressor. Acta Crystallog D-55: 2041±2042.
translation. EMBO J 11: 2675±2683. Nielsen, P.F., and Molin, S. (1984) How the R1 replication
Brantl, S. (1994) The copR gene product of plasmid pIP501 control system responds to copy number deviations.
acts as a transcriptional repressor at the essential repR Plasmid 11: 264±267.
promoter. Mol Microbiol 14: 473±483. NordstroÈm, K., and Wagner, E.G.H. (1994) Kinetic aspect of
Brantl, S., and Wagner, E.G.H. (1997) Dual function of the control of plasmid replication by antisense RNA. Trends
copR gene product of plasmid pIP501. J Bacteriol 179: Biol Sci 19: 294±300.
7016±7024. Novick, R.P., Iordanescu, S., Projan, S.J., Kornblum, J., and
Brantl, S., and Wagner, E.G.H. (2000) Antisense RNA- Edelman, I. (1989) pT181 plasmid replication is regulated
mediated transcriptional attenuation: an in vitro study of by a countertranscript-driven transcriptional attenuator.
plasmid pT181. Mol Microbiol 35: 1469±1482. Cell 59: 395±404.
Brantl, S., Behnke, D., and Alonso, J.C. (1990) Molecular Paulson, J., NordstroÈm, K., and Ehrenberg, M. (1998)
analysis of the replication region of the conjugative Requirements for rapid plasmid ColE1 copy number
Streptococcus agalactiae plasmid pIP501 in Bacillus adjustments: a mathematical model of inhibition modes
subtilis. Comparison with plasmids pAMb1 and and RNA turnover rates. Plasmid 39: 215±234.
pSM19035. Nucleic Acids Res 18: 4783±4790. Riise, E., Stougaard, P., Bindslev, B., NordstroÈm, K., and
Brantl, S., Birch-Hirschfeld, E., and Behnke, D. (1993) RepR Molin, S. (1982) Molecular cloning and functional char-
protein expression on plasmid pIP501 is controlled by an acterization of a copy number control gene (copB) of
antisense RNA-mediated transcription attenuation plasmid R1. J Bacteriol 151: 1136±1145.
mechanism. J Bacteriol 175: 4052±4061. Rosenfeld, R., and Grover, N.B. (1993) Control of mini-R1
Burian, J., StuchlõÂk, S., and Kay, W.W. (1999) Replication plasmid replication: a computer simulation. Plasmid 29:
control of a small cryptic plasmid of Escherichia coli. J Mol 94±116.
Biol 294: 49±65. Siemering, K.R., Praszkier, J., and Pittard, A.J. (1994)
Espinosa, M., Cohen, S.N., Couturier, M., del Solar, G., DõÂaz- Mechanisms of binding of the antisense and target RNAs
Orejas, R., Giraldo, R., et al. (2000) Plasmid replication and involved in the regulation of IncB plasmid replication. J
copy number control. In The Horizontal Gene Pool: Bacteriol 176: 2677±2688.
Bacterial Plasmids and Gene Spread. Thomas, C.M. del Solar, G., and Espinosa, M. (1992) The copy number of

Q 2000 Blackwell Science Ltd, Molecular Microbiology, 37, 492±500


500 G. del Solar and M. Espinosa
plasmid pLS1 is regulated by two trans-acting plasmid Tomizawa, J., and Itoh, T. (1981) Plasmid ColE1 incom-
products: the antisense RNA II and the repressor protein, patibility determined by interaction of RNA1 with primer
RepA. Mol Microbiol 6: 83±94. transcript. Proc Natl Acad Sci USA 78: 6096±6100.
del Solar, G., Acebo, P., and Espinosa, M. (1995) Replication Wagner, E.G.H., and Brantl, S. (1998) Kissing and RNA
control of plasmid pLS1: efficient regulation of plasmid copy stability in antisense control of plasmid replication. Trends
number is exerted by the combined action of two plasmid Biochem Sci 23: 451±454.
components, CopG and RNA II. Mol Microbiol 18: 913±924. Wagner, E.G.H., and Simons, R.W. (1994) Antisense RNA
del Solar, G., Giraldo, R., Ruiz-Echevarria, M.J., Espinosa, M., control in bacteria, phages, and plasmids. Annu Rev
and Diaz-Orejas, R. (1998) Replication and control of circular Microbiol 48: 713±742.
bacterial plasmids. Microbiol Mol Biol Rev 62: 434±464. Wilson, I.W., Praszkier, J., and Pittard, A.J. (1993) Mutations
Steinmetzer, K., Hillisch, A., Behlke, J., and Brantl, S. (2000) affecting pseudoknot control of the replication of B group
Transcriptional repressor CopR: structure model based plasmids. J Bacteriol 175: 6476±6483.
localization of the DNA binding motif. Proteins 38: 393±406. Wilson, I.W., Praszkier, J., and Pittard, A.J. (1994) Molecular
Summers, D.K. (1996). The Biology of Plasmids. Oxford: analysis of RNAI control repB translation in IncB plasmids.
Blackwell Science. J Bacteriol 176: 6497±6508.

Q 2000 Blackwell Science Ltd, Molecular Microbiology, 37, 492±500


CONJUGACION EN BACTERIAS GRAM NEGATIVAS
Fig. 4. Model for regulation of T-strand production and delivery to the translocation machinery of the MOBF T4SS (indicated by ‘uptake of DNA’ in abox). Steps in the
initiation reaction cascade (I–VII) are described in DNA processing of MOBF relaxosomes. Protein components are illustrated as follows:the bifunctional TraI
phosphodiesterase (blue) with active site tyrosine (Tyr) and TraI helicase domains (light blue), IHF (orange), TraY (purple), TraM (green) and the T4CP TraD
(yellow). See DNA processing of MOBF relaxosomes for details. r Journal of Bacteriology 19: 6888-6899, doi:10.1128/JB.00920-09, reproduced with permission from
the American Society for Microbiology.
fig07: Stages of T4 nucleoprotein transfer initiation mediated by plasmid R1-16.A and B. The T4 transfer apparatus is
constitutively expressed and assembled. Stage 1: The relaxosome, containing oriT bound by TraM (green), TraY
(violet), IHF (orange) and TraI (blue) is docked to the T4CP (yellow) via TraI translocation signals (TSA, TSB) and TraM
binding. Stage 2: Productive pilus contacts with adherent R17 phage (A) or another cell (B) produces signals (lightning
bolt) conveyed over the pilus to the T4CP-relaxosome receptor. Processing of those distinct signals into nucleoprotein
translocation requires at a minimum, a conserved relaxase activation domain. TraI N1-992 comprises this domain for R1
(inset). Stage 3: During phage infection (A) downstream activation of the T4CP ATPase activity is essential for uptake of
the R17 RNA–protein A complex. Alternatively, when cell contact permits plasmid self-transfer (B) initiation still requires
productive interactions between the T4CP and R1 TraM bound to oriT. Signal processing is performed by the relaxase
activation domain occupying the T4CP sites (inset). If TraI is bound to TraD, then the TraI N1-992 domain converts the
cell contact signal into localized oriT melting (inset) and helicase activation in a separate downstream step.

Mentions: In this model (Fig. 7) TraD is anchored to the base of the transfer channel while its cytosolic domain binds TraM, TraI and oriT
DNA (Stage 1). The relaxosome is catalytically active at nic in the absence of TraD, but cleavage is stimulated by its presence (Mihajlovic
et al., 2009). NTP hydrolysis by TraD appears to be silent. Progression from this stage requires signals communicated over the pilus from
the cell exterior (Stage 2). In the case of R17 phage adsorption the productive receptor for the incoming signal is TraD docked by the R1
relaxosome (Fig. 7A). The accessory factors bound at oriT are important, but the key component is the TraI N1-992 docking and
activation domain (inset). We propose that processing of the external signal through this mechanism depends on the physical link
between catalytic activity at the plasmid nick site and the T4CP to modulate TraD conformation and thereby activate the essential
ATPase. The activation initiates translocation of the RNA–protein A complex into the host cell (Stage 3). Activities related to TraI helicase
and its C-terminal domain are dispensable. The fact that this form of translocation activation is independent of the helicase domain and
that the helicase itself is not activated on the docked oriT under these conditions is logical, since phage penetration would otherwise
result in plasmid DNA being extruded pointlessly into the medium. This has never been observed.

BACTERIAS hfr (HIGH FREQUENCY RECOMBINATION)

Map eo de
genes mediante Hfrs
crossmark
MINIREVIEW

Enterococcal Sex Pheromones: Evolutionary Pathways to Complex,


Two-Signal Systems
Gary M. Dunny,a Ronnie Per-Arne Berntssonb
Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USAa; Department of Medical Biochemistry and Biophysics, Umeå
University, Umeå, Swedenb

Gram-positive bacteria carry out intercellular communication using secreted peptides. Important examples of this type of com-
munication are the enterococcal sex pheromone systems, in which the transfer of conjugative plasmids is controlled by intercel-
lular signaling among populations of donors and recipients. This review focuses on the pheromone response system of the con-
jugative plasmid pCF10. The peptide pheromones regulating pCF10 transfer act by modulating the ability of the PrgX
transcription factor to repress the transcription of an operon encoding conjugation functions. Many Gram-positive bacteria reg-
ulate important processes, including the production of virulence factors, biofilm formation, sporulation, and genetic exchange

Downloaded from http://jb.asm.org/ on March 7, 2017 by guest


using peptide-mediated signaling systems. The key master regulators of these systems comprise the RRNPP (RggRap/NprR/
PlcR/PrgX) family of intracellular peptide receptors; these regulators show conserved structures. While many RRNPP systems
include a core module of two linked genes encoding the regulatory protein and its cognate signaling peptide, the enterococcal sex
pheromone plasmids have evolved to a complex system that also recognizes a second host-encoded signaling peptide. Additional
regulatory genes not found in most RRNPP systems also modulate signal production and signal import in the enterococcal pher-
omone plasmids. This review summarizes several structural studies that cumulatively demonstrate that the ability of three
pCF10 regulatory proteins to recognize the same 7-amino-acid pheromone peptide arose by convergent evolution of unrelated
proteins from different families. We also focus on the selective pressures and structure/function constraints that have driven the
evolution of pCF10 from a simple, single-peptide system resembling current RRNPPs in other bacteria to the current complex
inducible plasmid transfer system.

BACKGROUND AND SIGNIFICANCE Suzuki et al. (9) research groups reported the identification of
several different molecules that mediated signaling for various
I n 1965, Tomasz (1) described “a new type of regulatory mecha-
nism in bacteria,” in which the control of competent cell genetic
transformation in pneumococci was expressed in a density-de-
plasmids; these signals were unmodified hydrophobic peptides 7
to 8 amino acid residues in length. These studies were the first
pendent fashion (1). He reported that the culture medium of cells demonstrations that the prevalent extracellular signaling mole-
grown to the optimal density for maximum competence con- cules of Gram-positive bacteria were oligopeptides, in contrast
tained a soluble factor capable of inducing competence expression to the acyl-homoserine-lactone signals that frequently mediate
when added to low-density noncompetent cultures. Conceptu- quorum sensing in Gram-negative microbes (10). Both the pep-
ally, the phenomenon of density-dependent pneumococcal com- tide-mediated signaling mechanisms and the peptide signals
petence expression mediated by intercellular signaling molecules themselves fall into two categories. Some signals are secreted as
is very similar to the autoinduction of light production in marine unmodified peptides processed from longer precursors, while
Vibrio species described a few years later by Nealson (2). These others are both processed and posttranslationally modified (11–
seminal studies initiated a paradigm shift in microbial research, 13). Likewise, sensing of peptide signals can involve either signal
changing the concept of normal bacterial behavior from single transduction across the membrane or signal import, followed by
cells acting independently to coordinated behaviors of microbial binding to a cytoplasmic receptor protein, which is often a tran-
populations via communication between individuals. Quorum scription factor (14).
sensing, in which a single cell type monitors its population density The enterococcal sex pheromone systems function by import
to coordinate activity (3), is perhaps the best studied mechanism of a signaling pheromone peptide encoded by the chromosome.
for the modulation of multicellular behaviors by intercellular sig- For simplicity, we use “C” as an abbreviation for all conjugation/
naling, which is more broadly termed sociomicrobiology (4). clumping-inducing peptide pheromones, where cCF10 is the pep-
Enterococcus faecalis is a major cause of opportunistic infec- tide that specifically induces cells carrying pCF10, cAD1 induces
tions of hospital patients, and E. faecalis clinical isolates are noto-
rious for their carriage of antibiotic resistance genes (5, 6). These
Accepted manuscript posted online 28 March 2016
are frequently disseminated by conjugation. In 1978, Dunny et al.
Citation Dunny GM, Berntsson RP-A. 2016. Enterococcal sex pheromones:
(7) reported that donor/recipient clumping and conjugative evolutionary pathways to complex, two-signal systems. J Bacteriol
transfer of plasmids in Enterococcus (formerly Streptococcus) 198:1556 –1562. doi:10.1128/JB.00128-16.
faecalis could be induced by low-molecular-weight signaling mol- Editor: W. Margolin, University of Texas Medical School at Houston
ecules excreted by recipient cells and sensed by plasmid-contain- Address correspondence to Gary M. Dunny, dunny001@umn.edu.
ing donor cells; it was suggested that these signals served as bacte- Copyright © 2016, American Society for Microbiology. All Rights Reserved.
rial sex pheromones. A few years later, the Clewell et al. (8) and

1556 jb.asm.org Journal of Bacteriology June 2016 Volume 198 Number 11


Minireview

FIG 1 Diagram of the signaling circuits in the E. faecalis pCF10 conjugation system (adapted from Annual Review of Genetics [19]). Recipient and donor have
similar chromosomes, but the donor also carries pCF10. The plasmid confers a response to the chromosomally encoded peptide C, which induces conjugation.

Downloaded from http://jb.asm.org/ on March 7, 2017 by guest


The plasmid encodes the antagonistic peptide I, which inhibits C competitively. Two constitutively expressed pCF10 gene products, PrgZ and PrgY, function in
pheromone import and in reduction of the amount of active C excreted by plasmid-carrying cells, respectively, as detailed in the text. Imported C interacts with
PrgX (not shown) in the cytoplasm to induce a conjugation response. Pheromone induction of donor cells results in the synthesis of conjugation-related gene
products, including surface adhesin proteins, type 4 secretion proteins (T4SS), and DNA transfer proteins (DTR).

those carrying pAD1, etc. Mature C is processed by host-encoded essay will focus on the tetracycline-resistant pheromone-respon-
proteins, and all known members of the sex pheromone family are sive plasmid pCF10 to illustrate the salient features of many sex
processed from the cleaved signal peptides of secreted lipoproteins pheromone plasmids (19) and to explore how the current com-
(15, 16). Binding of imported C by its cytoplasmic receptor initi- plex systems may have evolved from simpler progenitor systems
ates the pheromone response in the donor; the presence of C in the similar to the peptide-regulated RRNPP signaling systems that
growth medium of donor cells thus serves as a cue for the presence have now been implicated in the control of virulence, develop-
of recipients (Fig. 1). Peptide binding modulates the ability of the mental processes, and horizontal gene transfer in numerous
C receptor (PrgX in the case of pCF10) to regulate the transcrip- Gram-positive pathogens (20, 21).
tion of an operon containing conjugation genes (17). However,
the enterococcal sex pheromone systems have several additional OVERVIEW OF THE PEPTIDE-MEDIATED REGULATION OF
layers of complexity, including a second plasmid-encoded peptide pCF10 CONJUGATION
(inhibitor [I]) that competes directly with C for binding to the Figure 2 depicts a simplified map of the pheromone-inducible
same receptor (17, 18). In addition, several layers of posttranscrip- conjugation genes of pCF10 (22). The prgQ operon confers pro-
tional regulation greatly amplify the direct effects of the peptides duction of ⬎30 polypeptides and regulatory RNAs required for
on the expression of conjugation genes (17). The remainder of this regulated expression of conjugation. The pheromone receptor

FIG 2 Genetic organization of pheromone-inducible conjugation genes found on enterococcal plasmids (approximate size of the entire region indicated at the
top). This map depicts the prg genes of pCF10 with single-letter designations, but similar gene content and organization are found on other well-studied plasmids,
such as pAD1 and pPD1 (17). The left portion of the map shows conserved genes involved in pheromone sensing, and the relative locations of the genes of the
pheromone-inducible prgQ operon encoding the I peptide, surface adhesin gene module (ABUC), downstream type IV secretion system (T4SS) genes, and
conjugative DNA transfer genes (Dtr) are shown. The prgQ gene encodes the production of I, whereas an ⬃1-kb segment between prgQ and prgA encodes two
small open reading frames (ORFs) and small RNAs (sRNAs) that regulate the expression of downstream genes posttranscriptionally (65). The sizes of the
individual genes are not drawn to scale. I, the putative origin of the system as a surface protein module negatively regulated by quorum sensing through the X/Q
cassette; this gene pair resembles RRNPP systems recently identified in numerous Gram-positive pathogens (21, 31). II shows how the system became more
complex as it acquired the ability to enable its host cell to recognize C as an indicator of close proximity of plasmid-free recipients (mate sensing). At the
mechanistic level, the C peptide competes with I, which functions as a classic quorum-sensing signal of donor density (self-sensing) (64). Evolution of the ability
to differentially respond to these two antagonistic peptides was accompanied by the acquisition of genes encoding an oligopeptide binding protein, PrgZ, which
binds both C and I with high affinity and increases their import via the Opp ABC transporter (37, 38), and PrgY, a predicted membrane peptidase that reduces
the production of endogenous C by the host cell (36). III depicts the acquisition of T4SS and Dtr genes conferring conjugative transfer ability. There is high
conservation of the regions indicated by I and II among many pheromone plasmids, suggesting that they all arose from a common ancestor, but step III likely
occurred multiple times to link different conjugation gene cassettes to the pheromone-inducible aggregation module.

June 2016 Volume 198 Number 11 Journal of Bacteriology jb.asm.org 1557


Minireview

PrgX controls the initiation of transcription of this long operon the cleaved signal peptide of a predicted secreted lipoprotein
from the prgQ promoter; the interaction of I with PrgX reduces CcfA, whose function has not been demonstrated (15); likewise,
transcription, whereas the interaction of C with PrgX allows for all known pheromone-responsive plasmids analyzed to date en-
increased transcription. It is important to note that the direct ef- code a response to a specific peptide encoded by one of the ⬎50
fects of the peptides on control of the prgQ promoter by PrgX are potential lipoprotein genes in the organism (16). As indicated in
actually quite modest, but they are greatly amplified by several step II, the system acquired additional components that recognize
posttranscriptional mechanisms, which are described elsewhere C; PrgY prevents self-induction of donors by decreasing the
(23–27). Determination of the structures of Apo-PrgX and of amount of mature C released (36), and PrgZ binds both C and I
PrgX bound to I or C, along with extensive genetic and biochem- and facilitates their import into the cell via a chromosomally en-
ical analyses, indicates that Apo-PrgX and PrgX-I complexes re- coded peptide transporter (37, 38). PrgX also needed to evolve to
press transcription from the prgQ promoter, while PrgX-C com- recognize C and I. These 3 proteins are all from different families
plexes are impaired in repression (28, 29). It was originally and share only 9 to 13% sequence identity and no significant ho-
suggested that the replacement of I by C in PrgX-DNA complexes mology at the structural level. We have structural data on the
disrupts PrgX tetramers within repressing complexes, allowing interactions of PrgZ with C (37) and of PrgX with both C and I (28,
RNA polymerase to access the prgQ promoter (28–30). Very re- 29), but to date, there are no structural data available on PrgY.
cent data (Y. Chen, A. Bandyopadhyay, B. K. Kozlowicz, H. A. H. PrgZ belongs to the family of substrate-binding proteins found

Downloaded from http://jb.asm.org/ on March 7, 2017 by guest


Haemig, A. Tai, W.-S. Hu, and G. Dunny, unpublished data) sug- in ABC transporters, G protein-coupled receptors (GPCRs), and
gest that PrgX forms tetramers when complexed with either pep- DNA binding proteins (39, 40). It is likely that PrgZ evolved from
tide, but conformational differences of the DNA-bound tetramers a chromosomal oligopeptide-binding protein. Previous experi-
account for differential repression. In both models, the ultimate ments have shown that the oligopeptide-binding protein OppA of
induction state of a donor cell is dependent on the relative intra- E. faecalis can facilitate the import of C, even though a higher
cellular levels of I and C in donor cells. Interestingly, all of the concentration of C is required than is produced by recipients un-
peptide-controlled transcription factors of the RRNPP family ap- der normal physiological conditions (38). PrgZ can bind both C
pear to have a structure that is very similar to that of PrgX (20, 21, and I and has a typical Venus flytrap fold of a cluster C substrate-
31), and in most cases, the gene organization of the determinants binding protein (37), with C bound within an internal cavity (Fig.
for regulatory protein and the cognate regulatory peptide is simi- 3a). C is firmly bound to PrgZ via 10 direct hydrogen bonds. These
lar to that of prgX and prgQ. Below, we focus on the evolutionary bonds are mostly formed with the peptide backbone, with one
processes that likely shaped the emergence of the dual-peptide- exception, an H-bond that is formed with the side chain of Thr3,
controlled pCF10 system and how it may have evolved from a giving an explanation for results from genetic screens that Thr3 of
simple RRNPP-like system to its present complex state. C was important for PrgZ binding (41). Further H-bonds between
C and PrgZ are formed via bridging water molecules, and there is
HOW AND WHY DID THE pCF10 SYSTEM BECOME SO also a salt bridge that anchors the N terminus of C. Although no
COMPLEX? structure of PrgZ complexed with I is available, it is highly likely
The key functional components of the pCF10 system, which are that I binds in the same way as C (37). This is expected due to the
also found in other pheromone plasmids (17, 32), are illustrated in similarities of PrgZ to other oligopeptide-binding proteins (37,
Fig. 2. It is likely that current pheromone-inducible conjugation 40, 42, 43).
systems originated from a system with a single I-regulated Q-X- As noted previously, PrgX serves as the primary cytoplasmic
like module. This module likely controlled the expression of ad- receptor for both peptides and acts as the master transcription
jacent genes for surface adhesins, as similar surface adhesion gene regulator of the prgQ operon. PrgX has a conserved helix-turn-
content and organization are conserved with other pheromone- helix (HTH) domain responsible for its interaction with DNA (28,
controlled systems (17). Contemporary pheromone plasmids may 29). The binding site for C and I is located in the larger dimeriza-
have a common ancestor that includes contiguous genes corre- tion domain (Fig. 3b and c), where both peptides form ␤-sheet-
sponding to prgZ through prgQ and extending through the down- like interactions with PrgX and have a similar amount of H-bonds
stream cassette of LPXTG-anchored cell surface protein genes to PrgX. There is no conservation in the binding site between PrgX
(prgABC) and the small regulatory prgU gene (22, 33); this gene and PrgZ at either the sequence level or structurally. The two
cluster is indicated by the Roman numeral II in Fig. 2. Prior1 to the peptides favor different conformational states of PrgX, with the
acquisition of the ability to recognize the peptide signal C, the consequence that one alpha-helix is preferentially stabilized (I
I-autoregulated X-Q surface protein cluster may have functioned bound) or unwound (C bound) (28, 29). These different confor-
to increase the ability of the host bacterium to attach to other mations either favor transcriptional repression when inhibitor is
bacterial or metazoan host cells at low density while reducing bound, or they reduce the level of repression of PrgX when C is
these interactions at high bacterial density to enable escape from bound. While the basis for the differential effects of the two pep-
stagnant communities and recolonization of new niches. tides on the conformation of PrgX is not completely understood,
The next major event in the evolution of the system was prob- it probably relates to the N-terminal regions of the two peptides.
ably the ability to recognize the host-encoded C peptide as an The bulky leucine residue at the N terminus of C likely crowds the
indicator of the presence of plasmid-free enterococci in close surrounding PrgX residues in the binding pocket to a greater ex-
proximity. In strict evolutionary biology parlance (34, 35), C tent than the alanine at the N terminus of I (for a partial illustra-
would be classified as a cue rather than a signal, since the sensing tion, compare the PrgX residues surrounding the bound peptide
system seems to have hijacked this molecule produced from a gene N termini in Fig. 3b and c). These differences may indirectly affect
not linked to the sensing genes in physical proximity or in func- the conformation of the C terminus of PrgX.
tion. In the case of pCF10, the C peptide is produced by processing PrgY is required to prevent cells carrying pCF10 from being

1558 jb.asm.org Journal of Bacteriology June 2016 Volume 198 Number 11


Minireview

Downloaded from http://jb.asm.org/ on March 7, 2017 by guest


FIG 3 Comparison of the peptide binding of PrgZ (gray) complexed with C (a) and PrgX (dark red) complexed with C (b) or I (c). Each upper subfigure shows
the full protein structure in a cartoon representation, with the bound ligand in spheres, C in teal, and I in green. The lower enlarged representations at the bottom
show the ligand (as sticks) with interacting protein residues. As can be seen by comparing panels b and c, the C peptide has a very different structure when bound
to PrgZ compared with its structure when bound to PrgX.

self-induced by their own endogenous pheromone (36). Its amino stream genes predicted to encode the type 4 secretion systems
acid sequence, in conjunction with genetic and biochemical stud- (T4SSs) and DNA transfer (Dtr) machinery required for conjuga-
ies, suggest that a C-terminal subdomain anchors the protein in tion are located immediately downstream from the surface pro-
the membrane with the N-terminal region outside the cell; the tein cassettes in other pheromone plasmids, but these T4SS loci
external N-terminal subdomain confers the ability to specifically
bind the mature C peptide (36, 44) and may contribute to its
degradation. Initial studies of PrgY suggested that similar pro-
teins, none with known functions, were present in organisms from
all kingdoms, and that the protein phylogenies correlated with
those of the host organisms (36). Recently, an important new
study provided new insights into the structure/function relation-
ships of these proteins. Zhang et.al. (45) identified Tiki as a pro-
tease family playing a critical role in cell growth and development
via specific cleavage of the Wnt protein (45). PrgY is homologous
to the human Tiki metalloprotease, both having a pair of GX2H
motifs and a conserved glutamate residue, and it is predicted to
have structural similarity to the so-called EraA/ChaN-like family
of proteins (46). The structure of PrgY has not been determined,
but structural modeling using Phyre2 gives a model with a 96%
confidence level over most of the extracellular domain (Fig. 4).
This model does not contain any structural motif that resembles
the pheromone-binding site of either PrgZ or PrgX. From the
homology to the Tiki metalloproteases, we can deduce which res-
idues likely form the active site in PrgY, with some of those specific
residues, like His21, having previously been verified to be impor-
tant for function (36, 44). To date, only PrgY and Tiki are known
to have specific interactions with polypeptide substrates.
The cumulative analysis suggests that the pCF10 system did
not independently evolve these 3 different components from a FIG 4 Predicted structure of PrgY. The extracellular part of PrgY, here shown
single protein with a peptide-binding motif. More likely, an an- as a cartoon representation, was modeled using Phyre2 and colored from the N
cestral system, i.e., the inhibitor-regulated Q-X module, at some terminus (blue) toward the C-terminal end of the model (yellow). The C-ter-
minal domain, which could not be modeled, is predicted to contain 4 trans-
point acquired genes that coded for the early versions of PrgY and membrane helices, shown here as rectangles in a membrane. The predicted
PrgZ, and those proteins then evolved specific binding affinity to active site, based on the homology of PrgY to the Tiki metalloproteases (46), is
the cognate C and I peptides, as illustrated in Fig. 2. The down- shown within the dashed line.

June 2016 Volume 198 Number 11 Journal of Bacteriology jb.asm.org 1559


Minireview

show considerable divergence (22). This suggests that phero- donor densities, donors are poorly induced even by high concen-
mone-inducible aggregation cassettes became linked to the addi- trations of C (64). These cumulative effects of the inhibitor appar-
tional components required for conjugation on multiple occa- ently limit the extent of induction in mixed populations of donors
sions (Fig. 2, step III). Interestingly, the available data suggest that and recipients. This raises the question of whether the system may
the downstream conjugation functions for all known plasmids are have evolved to maintain mixed populations of donors and recip-
transcriptionally regulated by the peptide signals even though they ients in shared niches in the natural environment of the bacteria,
became linked to the upstream regions in multiple events (22). e.g., the intestinal tract. The maintenance of recipient populations
by limiting their conversion to donors should result in a steady
REMAINING QUESTIONS AND FUTURE DIRECTIONS supply of C within the niche, whose inducing capacity is limited by
While significant questions about the molecular mechanisms of the inhibitor. In this scenario, basal levels of expression of the
pheromone-mediated control of conjugation remain, the most inducible genes could be maintained within the mixed popula-
compelling areas for future study may be the analysis of structure/ tion, providing the previously described benefits (note that induc-
function relationships of the key regulatory components and in- tion of a few donors can coaggregate recipients and uninduced
vestigations of how these systems function in the natural environ- donors in close proximity) while minimizing costs of overexpres-
ment, including their impacts on maintenance and dissemination sion. The pheromone system has thus evolved under strong con-
of the plasmid itself, and on the fitness of the bacterial hosts. The flicting selective pressures for an extremely sensitive detection sys-

Downloaded from http://jb.asm.org/ on March 7, 2017 by guest


significance of such studies is heightened by the fact that consid- tem to induce expression while simultaneously limiting the extent
erable experimental and theoretical investigations of the evolu- and duration of induction. This may have driven convergent evo-
tionary aspects of the well-studied acyl-homoserine-lactone auto- lution of the three unrelated proteins with vital, but distinct, reg-
inducer systems in Gram-negative alphaproteobacteria have been ulatory functions to recognize the same peptide. Direct experi-
carried out (reviewed in reference 47). For example, a recent study mental testing of these ideas in the mammalian gastrointestinal
by Cornforth et al. (48) provided evidence that combining two (GI) tract, along with further mechanistic and structural studies of
separate quorum-sensing systems allowed for greater resolution regulatory components, is in progress and might yield insights
of the local environment of a bacterium, both in terms of sensing into more effective approaches to reduce the spread of antibiotic
social (microbial cell population density) and physical (diffusion, resistance and to impair the ability of resistant strains to overgrow
etc.) parameters. In contrast, much less attention has been di- and disrupt the gut microbiota of hospital patients.
rected toward the naturally occurring and more complex intercel-
lular communication system represented by pCF10. While pCF10 ACKNOWLEDGMENTS
was discovered because of its role in the transmission of antibiotic The research for this review was supported by U.S. PHS grant GM49530 to
resistance (49), enterococcal pheromone-responsive plasmids fre- G.M.D. and by the Kempe Foundations grant JCK-1524 to R.P.-A.B.
quently do not carry resistance genes (50, 51), suggesting that they
may encode other traits that increase the fitness of their host cells, FUNDING INFORMATION
relative to the costs of plasmid maintenance. It is interesting to This work, including the efforts of Gary M. Dunny, was funded by HHS |
consider how the expression of the pheromone-inducible conju- National Institutes of Health (NIH) (GM49530). This work, including the
gation genes may impact host fitness and how this relates to the efforts of Ronnie Per-Arne Berntsson, was funded by Kempestiftelserna
regulatory properties of the system. Published data from our (Kempe Foundations) (JCK-1524).
group (52–57) and others (58–61) suggest that the expression of
aggregation substance proteins, such as PrgB, can increase colo- REFERENCES
nization and virulence by promoting biofilm formation and at- 1. Tomasz A. 1965. Control of the competent state in Pneumococcus by a
hormone-like cell product: an example for a new type of regulatory
tachment to host tissues, and by increasing resistance to phago- mechanism in bacteria. Nature 208:155–159. http://dx.doi.org/10.1038
cytic killing. Notably, there are still no direct data on how PrgB or /208155a0.
other inducible proteins might impact fitness in the gut. On the 2. Nealson KH. 1999. Early observations defining quorum-dependent gene
other hand, the overexpression of these genes likely has very high expression, p 277–290. In Dunny GM, Winans SC (ed), Cell-cell signaling
in bacteria. ASM Press, Washington, DC.
costs for the induced cell, including the energy required for syn-
3. Fuqua WC, Winans SC, Greenberg EP. 1994. Quorum sensing in bac-
thesizing conjugation proteins, the likely inhibition of growth in teria: the LuxR-LuxI family of cell density-responsive transcriptional reg-
cells trapped in large aggregates, and cell death and lysis due to ulators. J Bacteriol 176:269 –275.
toxic effects of overexpressed gene products on highly induced 4. Parsek MR, Greenberg EP. 2005. Sociomicrobiology: the connections
cells (62). Interestingly, clusters of genes related to the plasmid- between quorum sensing and biofilms. Trends Microbiol 13:27–33. http:
//dx.doi.org/10.1016/j.tim.2004.11.007.
encoded pheromone-inducible adhesins/transfer determinants 5. Kristich CJ, Rice LB, Arias CA. 6 February 2014. Enterococcal infec-
have been identified within genomic islands in the chromosomes tion—treatment and antibiotic resistance. In Gilmore MS, Clewell DB, Ike
of some strains, but these chromosomal determinants are not ca- Y, Shankar N (ed), Enterococci: from commensals to leading causes of
pable of transfer unless a coresident pheromone plasmid inte- drug resistant infection. Massachusetts Eye & Ear Infirmary, Boston, MA.
http://www.ncbi.nlm.nih.gov/books/NBK190420/.
grates and mobilizes them via an Hfr-like mechanism (63).
6. Miller WR, Munita JM, Arias CA. 2014. Mechanisms of antibiotic resis-
Numerous studies have documented the extremely tight regu- tance in enterococci. Expert Rev Anti Infect Ther 12:1221–1236. http://dx
lation of the pheromone system (19). The system not only avoids .doi.org/10.1586/14787210.2014.956092.
spurious induction but also limits the duration of induction due 7. Dunny GM, Brown BL, Clewell DB. 1978. Induced cell aggregation and
to the fact that the induction process itself dramatically increases mating in Streptococcus faecalis: evidence for a bacterial sex pheromone.
Proc Natl Acad Sci USA 75:3479 –3483. http://dx.doi.org/10.1073/pnas.75
inhibitor production, leading to rapid shut off of the response .7.3479.
after a short period of induction (64). Furthermore, the inhibitor 8. Mori M, Sakagami Y, Narita M, Isogai A, Fujino M, Kitada C, Craig RA,
can function as a classic quorum sensor of donor density; at high Clewell DB, Suzuki A. 1984. Isolation and structure of the bacterial sex

1560 jb.asm.org Journal of Bacteriology June 2016 Volume 198 Number 11


Minireview

pheromone, cAD1, that induces plasmid transfer in Streptococcus faecalis. 2006. Molecular basis for control of conjugation by bacterial pheromone
FEBS Lett 178:97–100. http://dx.doi.org/10.1016/0014-5793(84)81248-X. and inhibitor peptides. Mol Microbiol 62:958 –969. http://dx.doi.org/10
9. Suzuki A, Mori M, Sakagami Y, Isogai A, Fujino M, Kitaga C, Craig RA, .1111/j.1365-2958.2006.05434.x.
Clewell DB. 1984. Isolation and structure of bacterial sex pheromone, 29. Shi K, Brown CK, Gu ZY, Kozlowicz BK, Dunny GM, Ohlendorf DH,
cPD1. Science 226:849 – 850. http://dx.doi.org/10.1126/science.6436978. Earhart CA. 2005. Structure of peptide sex pheromone receptor PrgX and
10. Fuqua C, Parsek MR, Greenberg EP. 2001. Regulation of gene expression PrgX/pheromone complexes and regulation of conjugation in Enterococ-
by cell-to-cell communication: acyl-homoserine lactone quorum sensing. cus faecalis. Proc Natl Acad Sci U S A 102:18596 –18601. http://dx.doi.org
Annu Rev Genet 35:439 – 468. http://dx.doi.org/10.1146/annurev.genet /10.1073/pnas.0506163102.
.35.102401.090913. 30. Kozlowicz BK. 2005. The molecular mechanism and peptide signaling
11. Chandler JR, Dunny GM. 2004. Enterococcal peptide sex pheromones: response of PrgX used to control pheromone-induced conjugative
synthesis and control of biological activity. Peptides 25:1377–1388. http: transfer of pCF10. Ph.D. dissertation. University of Minnesota, Min-
//dx.doi.org/10.1016/j.peptides.2003.10.020. neapolis, MN.
12. Dunny GM, Leonard BAB. 1997. Cell-cell communication in Gram- 31. Declerck N, Bouillaut L, Chaix D, Rugani N, Slamti L, Hoh F, Lereclus D,
positive bacteria. Annu Rev Microbiol 51:527–564. Arold ST. 2007. Structure of PlcR: insights into virulence regulation and
13. Dunny GM, Winans SC. 1999. Cell-cell signaling in bacteria. ASM Press, evolution of quorum sensing in Gram-positive bacteria. Proc Natl Acad Sci
Washington, DC. U S A 104:18490 –18495. http://dx.doi.org/10.1073/pnas.0704501104.
14. Waters CM, Bassler BL. 2005. Quorum sensing: cell-to-cell communica- 32. Clewell DB, Dunny GM. 2002. Conjugation and genetic exchange in
tion in bacteria. Annu Rev Cell Dev Biol 21:319 –346. http://dx.doi.org/10 enterococci, p 265–300. In Gilmore MS, Clewell DB, Courvalin P, Dunny
.1146/annurev.cellbio.21.012704.131001. GM, Murray BE, Rice LB (ed), The enterococci: pathogenesis, molecular
15. Antiporta MH, Dunny GM. 2002. ccfA, the genetic determinant for the biology and antibiotic resistance. ASM Press, Washington, DC.

Downloaded from http://jb.asm.org/ on March 7, 2017 by guest


cCF10 peptide pheromone in Enterococcus faecalis OG1RF. J Bacteriol 33. Kozlowicz BK, Dworkin M, Dunny GM. 2006. Pheromone-inducible
184:1155–1162. http://dx.doi.org/10.1128/jb.184.4.1155-1162.2002. conjugation in Enterococcus faecalis: a model for the evolution of biological
16. Clewell DB, An FY, Flannagan SE, Antiporta M, Dunny GM. 2000. complexity? Int J Med Microbiol 296:141–147. http://dx.doi.org/10.1016
Enterococcal sex pheromone precursors are part of signal sequences for /j.ijmm.2006.01.040.
surface lipoproteins. Mol Microbiol 35:246 –248. http://dx.doi.org/10 34. Platt TG, Fuqua C. 2010. What’s in a name? The semantics of quorum
.1046/j.1365-2958.2000.01687.x. sensing. Trends Microbiol 18:383–387.
17. Clewell DB, Weaver KE, Dunny GM, Coque TM, Francia MV, Hayes F. 35. Keller L, Surette MG. 2006. Communication in bacteria: an ecological
9 February 2014. Extrachromosomal and mobile elements in enterococci: and evolutionary perspective. Nat Rev Microbiol 4:249 –258. http://dx.doi
transmission, maintenance, and epidemiology. In Gilmore MS, Clewell .org/10.1038/nrmicro1383.
DB, Ike Y, Shankar N (ed), Enterococci: from commensals to leading 36. Chandler JR, Flynn AR, Bryan EM, Dunny GM. 2005. Specific control of
causes of drug resistant infection. Massachusetts Eye & Ear Infirmary, endogenous cCF10 pheromone by a conserved domain of the pCF10-
Boston, MA. http://www.ncbi.nlm.nih.gov/books/NBK190430/. encoded regulatory protein PrgY in Enterococcus faecalis. J Bacteriol 187:
18. Nakayama J, Ruhfel RE, Dunny GM, Isogai A, Suzuki A. 1994. The prgQ 4830 – 4843. http://dx.doi.org/10.1128/JB.187.14.4830-4843.2005.
gene of the Enterococcus faecalis tetracycline resistance plasmid pCF10 37. Berntsson RP, Schuurman-Wolters GK, Dunny G, Slotboom DJ, Pool-
encodes a peptide inhibitor, iCF10. J Bacteriol 176:7405–7408. man B. 2012. Structure and mode of peptide binding of pheromone re-
19. Dunny GM. 2013. Enterococcal sex pheromones: signaling, social behav- ceptor PrgZ. J Biol Chem 287:37165–37170. http://dx.doi.org/10.1074/jbc
ior, and evolution. Annu Rev Genet 47:457– 482. http://dx.doi.org/10 .M112.386334.
.1146/annurev-genet-111212-133449. 38. Leonard BA, Podbielski A, Hedberg PJ, Dunny GM. 1996. Enterococcus
20. Cook LC, Federle MJ. 2014. Peptide pheromone signaling in Streptococ- faecalis pheromone binding protein, PrgZ, recruits a chromosomal oligo-
cus and Enterococcus. FEMS Microbiol Rev 38:473– 492. http://dx.doi.org peptide permease system to import sex pheromone cCF10 for induction of
/10.1111/1574-6976.12046. conjugation. Proc Natl Acad Sci U S A 93:260 –264. http://dx.doi.org/10
21. Parashar V, Aggarwal C, Federle MJ, Neiditch MB. 2015. Rgg protein .1073/pnas.93.1.260.
structure-function and inhibition by cyclic peptide compounds. Proc 39. Berntsson RP, Doeven MK, Fusetti F, Duurkens RH, Sengupta D,
Natl Acad Sci U S A 112:5177–5182. http://dx.doi.org/10.1073/pnas Marrink SJ, Thunnissen AM, Poolman B, Slotbloom DJ. 2009. The
.1500357112. structural basis for peptide selection by the transport receptor OppA.
22. Hirt H, Manias DA, Bryan EM, Klein JR, Marklund JK, Staddon JH, EMBO J 28:1332–1340. http://dx.doi.org/10.1038/emboj.2009.65.
Paustian ML, Kapur V, Dunny GM. 2005. Characterization of the pher- 40. Berntsson RP, Smits SH, Schmitt L, Slotbloom DJ, Poolman B. 2010. A
omone response of the Enterococcus faecalis conjugative plasmid pCF10: structural classification of substrate-binding proteins. FEBS Lett 584:
complete sequence and comparative analysis of the transcriptional and 2606 –2617. http://dx.doi.org/10.1016/j.febslet.2010.04.043.
phenotypic responses of pCF10-containing cells to pheromone induction. 41. Fixen KR, Chandler JR, Le T, Kozlowicz BK, Manias DA, Dunny GM.
J Bacteriol 187:1044 –1054. http://dx.doi.org/10.1128/JB.187.3.1044-1054 2007. Analysis of the amino acid sequence specificity determinants of the
.2005. enterococcal cCF10 sex pheromone in interactions with the pheromone-
23. Bae T, Kozlowicz BK, Dunny GM. 2004. Characterization of cis-acting sensing machinery. J Bacteriol 189:1399 –1406. http://dx.doi.org/10.1128
prgQ mutants: evidence for two distinct repression mechanisms by Qa /JB.01226-06.
RNA and PrgX protein in pheromone-inducible enterococcal plasmid 42. Levdikov VM, Blagova EV, Brannigan JA, Wright L, Vagin AA, Wilkin-
pCF10. Mol Microbiol 51:271–281. son AJ. 2005. The structure of the oligopeptide-binding protein, AppA,
24. Chatterjee A, Johnson CM, Shu CC, Kaznessis YN, Ramkrishna D, from Bacillus subtilis in complex with a nonapeptide. J Mol Biol 345:879 –
Dunny GM, Hu WS. 2011. Convergent transcription confers a bistable 892. http://dx.doi.org/10.1016/j.jmb.2004.10.089.
switch in Enterococcus faecalis conjugation. Proc Natl Acad Sci U S A 43. Tame JR, Murshudov GN, Dodson EJ, Neil TK, Dodson GG, Higgins
108:9721–9726. http://dx.doi.org/10.1073/pnas.1101569108. CF, Wilkinson AJ. 1994. The structural basis of sequence-independent
25. Dunny GM, Johnson CM. 2011. Regulatory circuits controlling entero- peptide binding by OppA protein. Science 264:1578 –1581. http://dx.doi
coccal conjugation: lessons for functional genomics. Curr Opin Microbiol .org/10.1126/science.8202710.
14:174 –180. http://dx.doi.org/10.1016/j.mib.2011.01.008. 44. Chandler JR, Dunny GM. 2008. Characterization of the sequence speci-
26. Johnson CM, Haemig HH, Chatterjee A, Wei-Shou H, Weaver KE, ficity determinants required for processing and control of sex pheromone
Dunny GM. 2011. RNA-mediated reciprocal regulation between two bac- by the intramembrane protease Eep and the plasmid-encoded protein
terial operons is RNase III dependent. mBio 2(5):e00189-11. http://dx.doi PrgY. J Bacteriol 190:1172–1183. http://dx.doi.org/10.1128/JB.01327-07.
.org/10.1128/mBio.00189-11. 45. Zhang X, Abreu JG, Yokota C, MacDonald BT, Singh S, Coburn KL,
27. Johnson CM, Manias DA, Haemig HA, Shokeen S, Weaver KE, Henkin Cheong SM, Zhang MM, Ye QZ, Hang HC, Steen H, He X. 2012. Tiki1
TM, Dunny GM. 2010. Direct evidence for control of the pheromone- is required for head formation via Wnt cleavage-oxidation and inactiva-
inducible prgQ operon of Enterococcus faecalis plasmid pCF10 by a coun- tion. Cell 149:1565–1577. http://dx.doi.org/10.1016/j.cell.2012.04.039.
tertranscript-driven attenuation mechanism. J Bacteriol 192:1634 –1642. 46. Bazan JF, Macdonald BT, He X. 2013. The TIKI/TraB/PrgY family: a
http://dx.doi.org/10.1128/JB.01525-09. common protease fold for cell signaling from bacteria to metazoa? Dev
28. Kozlowicz BK, Shi K, Gu ZY, Ohlendorf DH, Earhart CA, Dunny GM. Cell 25:225–227. http://dx.doi.org/10.1016/j.devcel.2013.04.019.

June 2016 Volume 198 Number 11 Journal of Bacteriology jb.asm.org 1561


Minireview

47. Stevens AM, Schuster M, Rumbaugh KP. 2012. Working together for the stances enhance pathogenicity in rabbit models of Enterococcus faecalis
common good: cell-cell communication in bacteria. J Bacteriol 194:2131– endocarditis. Infect Immun 66:218 –223.
2141. http://dx.doi.org/10.1128/JB.00143-12. 58. Kreft B, Marre R, Schramm U, Wirth R. 1992. Aggregation substance of
48. Cornforth DM, Popat R, McNally L, Gurney J, Scott-Phillips TC, Ivens Enterococcus faecalis mediates adhesion to cultured renal tubular cells.
A, Diggle SP, Brown SP. 2014. Combinatorial quorum sensing allows Infect Immun 60:25–30.
bacteria to resolve their social and physical environment. Proc Natl Acad 59. Süssmuth SD, Muscholl-Silberhorn A, Wirth R, Susa M, Marre R,
Sci U S A 111:4280 – 4284. http://dx.doi.org/10.1073/pnas.1319175111. Rozdzinski E. 2000. Aggregation substance promotes adherence, phago-
49. Dunny G, Funk C, Adsit J. 1981. Direct stimulation of the transfer of cytosis, and intracellular survival of Enterococcus faecalis within human
antibiotic resistance by sex pheromones in Streptococcus faecalis. Plasmid macrophages and suppresses respiratory burst. Infect Immun 68:4900 –
6:270 –278. http://dx.doi.org/10.1016/0147-619X(81)90035-4. 4906. http://dx.doi.org/10.1128/IAI.68.9.4900-4906.2000.
50. Clewell DB. 1993. Bacterial sex pheromone-induced plasmid transfer. 60. Chow JW, Thal LA, Perri MB, Vazquez JA, Donabedian SM, Clewell
Cell 73:9 –12. http://dx.doi.org/10.1016/0092-8674(93)90153-H. DB, Zervos MJ. 1993. Plasmid-associated hemolysin and aggregation
51. Clewell DB. 1999. Sex pheromone systems in enterococci, p 47– 66. In substance production contribute to virulence in experimental enterococ-
Dunny GM, Winans SC (ed), Cell-cell signaling in bacteria. ASM Press, cal endocarditis. Antimicrob Agents Chemother 37:2474 –2477. http://dx
Washington, DC. .doi.org/10.1128/AAC.37.11.2474.
52. Chandler JR, Hirt H, Dunny GM. 2005. A paracrine peptide sex phero- 61. Rakita RM, Vanek NN, Jacques-Palaz K, Mee M, Mariscalco MM,
mone also acts as an autocrine signal to induce plasmid transfer and vir- Dunny GM, Snuggs M, van Winkle WB, Simon SI. 1999. Enterococcus
ulence factor expression in vivo. Proc Natl Acad Sci U S A 102:15617– faecalis bearing aggregation substance is resistant to killing by human neu-
15622. http://dx.doi.org/10.1073/pnas.0505545102. trophils despite phagocytosis and neutrophil activation. Infect Immun
53. Chuang ON, Schlievert PM, Wells CL, Manias DA, Tripp TJ, Dunny 67:6067– 6075.

Downloaded from http://jb.asm.org/ on March 7, 2017 by guest


GM. 2009. Multiple functional domains of Enterococcus faecalis aggrega- 62. Bhatty M, Cruz MR, Frank KL, Gomez JA, Andrade F, Garsin DA,
tion substance Asc10 contribute to endocarditis virulence. Infect Immun Dunny GM, Kaplan HB, Christie PJ. 2015. Enterococcus faecalis pCF10-
77:539 –548. http://dx.doi.org/10.1128/IAI.01034-08. encoded surface proteins PrgA, PrgB (aggregation substance) and PrgC
54. Chuang-Smith ON, Wells CL, Henry-Stanley MJ, Dunny GM. 2010. contribute to plasmid transfer, biofilm formation and virulence. Mol Mi-
Acceleration of Enterococcus faecalis biofilm formation by aggregation crobiol 95:660 – 677. http://dx.doi.org/10.1111/mmi.12893.
substance expression in an ex vivo model of cardiac valve colonization. 63. Manson JM, Hancock LE, Gilmore MS. 2010. Mechanism of chromo-
PLoS One 5:e15798. http://dx.doi.org/10.1371/journal.pone.0015798. somal transfer of Enterococcus faecalis pathogenicity island, capsule, anti-
55. Hirt H, Erlandsen SL, Dunny GM. 2000. Heterologous inducible expres- microbial resistance, and other traits. Proc Natl Acad Sci U S A 107:
sion of Enterococcus faecalis pCF10 aggregation substance Asc10 in Lacto- 12269 –12274. http://dx.doi.org/10.1073/pnas.1000139107.
coccus lactis and Streptococcus gordonii demonstrates contribution to cell 64. Chatterjee A, Cook LCC, Shu C-C, Chen Y, Manias DA, Ramkrishna D,
hydrophobicity and adhesion to fibrin. J Bacteriol 182:2299 –2306. http: Dunny GM, Hu W-S. 2013. Antagonistic self-sensing and mate-sensing
//dx.doi.org/10.1128/JB.182.8.2299-2306.2000. signaling controls antibiotic-resistance transfer. Proc Natl Acad Sci U S A
56. Olmsted SB, Dunny GM, Erlandsen SL, Wells CL. 1994. A plasmid- 110:7086 –7090. http://dx.doi.org/10.1073/pnas.1212256110.
encoded surface protein on Enterococcus faecalis augments its internaliza- 65. Bensing BA, Manias DA, Dunny GM. 1997. Pheromone cCF10 and
tion by cultured intestinal epithelial cells. J Infect Dis 170:1549 –1556. plasmid pCF10-encoded regulatory molecules act post-transcriptionally
57. Schlievert PM, Gahr PJ, Assimacopoulos AP, Dinges MM, Stoehr JA, to activate expression of downstream conjugation functions. Mol Micro-
Harmala JW, Hirt H, Dunny GM. 1998. Aggregation and binding sub- biol 24:285–294. http://dx.doi.org/10.1046/j.1365-2958.1997.3301710.x.

Gary M. Dunny received his B.S. and Ph.D. Ronnie Per-Arne Berntsson studied biotech-
from the University of Michigan and spent 11 nology at Chalmers University in Gothenburg,
years at Cornell University as a postdoctoral fel- Sweden. In 2010, he received his Ph.D. in bio-
low and as a faculty member before moving to chemistry from the University of Groningen,
the University of Minnesota in 1989, where he is the Netherlands, after working in the groups of
currently professor of microbiology. He has Bert Poolman and Dirk-Jan Slotboom on stud-
studied conjugation, cell signaling, and adapta- ies of ABC transporters and their domains. Af-
tion in enterococci using genetics, biochemis- ter his Ph.D., he moved to Stockholm Univer-
try, and microscopic imaging for his entire sity, Sweden, where he received an EMBO
career. fellowship to do postdoctoral research in the
group of Pål Stenmark on botulinum neurotox-
ins and their receptors. In 2015, he became an assistant professor at the
Department of Medical Biochemistry and Biophysics at Umeå University,
Sweden. His laboratory studies the function, structure, and regulation of
type 4 secretion systems in Gram-positive bacteria.

1562 jb.asm.org Journal of Bacteriology June 2016 Volume 198 Number 11


ANRV260-GE39-18 ARI 15 October 2005 13:5

Switches in Bacteriophage
Lambda Development∗
Amos B. Oppenheim,1,3,4 Oren Kobiler,1
Joel Stavans,2 Donald L. Court,3
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org
by University of California - San Diego on 03/29/11. For personal use only.

and Sankar Adhya4


1
Department of Molecular Genetics and Biotechnology, The Hebrew
University-Hadassah Medical School, Jerusalem 91120
2
Department of Physics of Complex Systems, Weizmann Institute of Science,
Rehovot, Israel 76100
3
Gene Regulation and Chromosome Biology Laboratory, National Cancer Institute at
Frederick, Frederick, Maryland 21702
4
Laboratory of Molecular Biology, National Cancer Institute, Bethesda, Maryland
20892-4264; email: oppenhea@mail.nih.gov

Annu. Rev. Genet. Key Words


2005. 39:409–29
genetic circuits, gene modules, developmental pathways,
First published online as a
Review in Advance on developmental decisions, threshold effects, host factors
July 26, 2005
Abstract
The Annual Review of
Genetics is online at The lysis-lysogeny decision of bacteriophage lambda (λ) is a
http://genet.annualreviews.org paradigm for developmental genetic networks. There are three key
doi: 10.1146/ features, which characterize the network. First, after infection of
annurev.genet.39.073003.113656 the host bacterium, a decision between lytic or lysogenic develop-
Copyright 
c 2005 by ment is made that is dependent upon environmental signals and the
Annual Reviews. All rights number of infecting phages per cell. Second, the lysogenic prophage
reserved
state is very stable. Third, the prophage enters lytic development in

The U.S. Government response to DNA-damaging agents. The CI and Cro regulators de-
has the right to retain a
nonexclusive, royalty-free fine the lysogenic and lytic states, respectively, as a bistable genetic
license in and to any switch. Whereas CI maintains a stable lysogenic state, recent studies
copyright covering this indicate that Cro sets the lytic course not by directly blocking CI
paper.
expression but indirectly by lowering levels of CII which activates cI
0066-4197/05/1215- transcription. We discuss how a relatively simple phage like λ employs
0409$20.00
a complex genetic network in decision-making processes, providing
a challenge for theoretical modeling.

409
ANRV260-GE39-18 ARI 15 October 2005 13:5

tory networks. A growing number of the-


Contents oretical modeling investigators are attempt-
ing to explain the underlying principles of
INTRODUCTION . . . . . . . . . . . . . . . . 410
complex regulatory networks involved in nor-
The λ System . . . . . . . . . . . . . . . . . . . . 410
mal mammalian development, including al-
GENE ORGANIZATION
terations that can result in a disease state
AND REGULATION . . . . . . . . . . . 411
such as cancer. Analysis of such complex sys-
The Lytic Transcription Cascade . 413
tems would be greatly facilitated by simi-
The Lysogenic Process . . . . . . . . . . . 413
lar studies using an ideal paradigm in which
THE PROPHAGE STATE
most if not all of the elements composing
AND ITS MAINTENANCE . . . . . 413
the system were known. Phage λ, the most
THE DEFAULT LYTIC COURSE:
comprehensively studied bacteriophage, is the
Cro AND N FUNCTIONS . . . . . . 415
prototype of a class of lambdoid phages with
The N Antiterminator . . . . . . . . . . . 415
whom it shares similar genome organization
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

SWITCHING THE DEFAULT


and functions (38, 82). Studies of λ that began
by University of California - San Diego on 03/29/11. For personal use only.

LYTIC MODE TO LYSOGENY:


in the 1950s continue to reveal key molecu-
ROLE OF CII . . . . . . . . . . . . . . . . . . . 417
lar processes in gene regulatory mechanisms
Regulation of CII Activity . . . . . . . . 417
and development. However, despite years of
Regulation of CIII . . . . . . . . . . . . . . . . 418
study, many genetic interactions still remain
THE DECISION PROCESS:
to be uncovered and those that we already
GENETIC FUNCTIONS AND
know require reexamination. For an accurate,
EXTERNAL INFLUENCES . . . . 419
complete, and quantitative analysis of the ge-
Multiplicity Effect . . . . . . . . . . . . . . . . 419
netic network, in particular its temporal pro-
Physiological Effects . . . . . . . . . . . . . . 419
gression, these remaining questions need to
COEXPRESSION OF BOTH
be addressed. In this review we summarize
LYTIC AND LYSOGENIC
a systems biology approach to the study of
GENES FROM THE SAME
genetic regulatory circuits of phage λ. We
OPERON: A PARADOX . . . . . . . . 421
define the individual components of the cir-
THEORETICAL STUDIES . . . . . . . . 421
cuits and switches, describe the kinetics of
OPEN QUESTIONS, SUMMARY,
their interactions, and explain how the in-
AND CONCLUSION . . . . . . . . . . . 422
teractions achieve robustness in the perfor-
Counting Infecting Phage. . . . . . . . . 422
mance of the circuits. We also stress some
The Distinction Between Phage
puzzles that still exist in lambda’s regulatory
Infection and Prophage
system.
Induction . . . . . . . . . . . . . . . . . . . . . 423
Our citation of literature is not exhaus-
Role of Cro in Lytic Growth . . . . . 423
tive but provides examples to illustrate spe-
Evolution of Temperate Phages . . . 423
cific points. We direct the reader to several
EPILOG . . . . . . . . . . . . . . . . . . . . . . . . . . . 423
comprehensive reviews on phage biology (13,
15, 24, 31, 34, 37, 41, 82, 107).

For the concert of life no-one gets a program.


[From a Dutch tile (81)] The λ System
Bacteria and their temperate phages, like Es-
cherichia coli and λ, exist in symbiotic rela-
INTRODUCTION tionships. These phages can be present in a
A central challenge in the post genomic era dormant, lysogenic (prophage) state replicat-
is to understand processes governing the ing passively with the host or they can de-
dynamics of highly complex genetic regula- velop lytically, producing progeny phages and

410 Oppenheim et al.


ANRV260-GE39-18 ARI 15 October 2005 13:5
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org
by University of California - San Diego on 03/29/11. For personal use only.

Figure 1
Decision-making steps by the temperate λ phage. A cell infected with phage λ can follow (denoted as
Decision I) the lytic response (left) or the lysogenic response (right). The resulting lysogenic cell carries a
repressed prophage shown in orange. The prophage is irreversibly induced only when a threshold
amount of DNA damage causes an SOS response, leading to lytic development (Decision II). The
prophage is normally extremely stable and rarely undergoes this type of induction by random DNA
damage to produce progeny phage in a very small fraction of the lysogenic cell (basal or spontaneous
induction). Some of the spontaneously induced cells enter the lytic cycle abortively, lose the prophage
(curing), and become nonlysogens (Decision III) (65).

killing their hosts. λ phage infecting an E. ter infection abortive lytic or lysogenic events
coli cell makes a decision to follow either a may also occur (62). The importance of
lytic or a lysogenic pathway (Figure 1). If the these abortive events has not been thoroughly
lytic pathway is followed, the phage replicates studied.
its DNA autonomously, expresses the mor-
phogenetic genes, assembles virions, and ly-
ses the host. If the lysogenic course ensues, GENE ORGANIZATION
a stable lysogen is established in which the AND REGULATION
prophage is integrated into the host chro- The λ genetic map and transcription profile
mosome with lytic gene expression turned involved in early developmental processes are
off. The prophage DNA replicates as part shown in Figure 2. The gene organization
of the bacterial genome during subsequent of lambdoid phages is based on a number of
cell divisions, and confers immunity to the recurring principles. Phage λ and its many
cell against infection by another λ. Treatment relatives have genomes that evolved as highly
with DNA-damaging agents, which leads to mosaic, modular structures. This property has
an SOS response, causes the lysogenic state long been recognized and led to the formula-
to irreversibly switch into lytic development, tion of the “modular genome hypothesis” (10,
mimicking the lytic infection. Otherwise, the 14, 38, 101). A short summary of the λ phage
prophage state is extremely stable, rarely un- modules and submodules is given in Table 1.
dergoing induction by DNA damage. Some Thus, for example, it is possible to replace
of these rarely induced cells enter an abortive the “immunity” module of λ by that of an-
lytic cycle by losing the prophage and be- other lambdoid phage. The organization of
coming nonlysogens (curing). Similarly, af- the gene modules allows a typical cascade of

www.annualreviews.org • Switches in Bacteriophage Lambda Development 411


ANRV260-GE39-18 ARI 15 October 2005 13:5

Figure 2
Genetic map and transcriptional units of the phage regulatory region. Key genes and signals discussed in
the text are shown in their map order between the parallel lines. The early transcripts, the extended
delayed early transcripts, and the late transcripts are shown in black arrows. The transcripts initiated
from the pI, pRE, and paQ that are required for lysogeny are shown in blue. The pRM promoter, which is
activated by the CI regulator, is required for maintenance of the lysogenic state, which is shown in green.
Critical transcription terminators are marked in orange, including the sib region containing the tI
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

terminator. Leftward promoters are indicated above and the rightward ones below the map. pL and pR
by University of California - San Diego on 03/29/11. For personal use only.

are the early promoters and pR is the late lytic promoter. The role of the pOOP promoter is not fully
understood. The operators oL and oR, cognate to pL and pR respectively, are also shown. The immunity
module of the λ chromosome encompasses pLoL, rex, cI, oRpR, and cro. ori is the origin of O- and
P-mediated phage DNA replication (Table 1). Int carries the site-specific recombination reaction, and
Int and Xis support the excision reaction.

phage gene expression in lytic growth delin- ing to the oL and oR operators that overlap
eating the early, delayed early, and late stages the pL and pR promoters, allowing the main-
of transcription. This modular and temporal tenance of the lysogenic state to be governed
expression facilitates the alternative λ devel- by CI alone; when CI is inactivated, e.g., by
opmental pathways. Because of the transcrip- the SOS response, the lytic development fol-
tional cascade, the repression of the early lows. By blocking the pL and pR promoters
phage promoters pR and pL prevents expres- of an incoming phage genome, the CI reg-
sion of all lytic genes (Figure 2, Figure 3). ulator confers immunity against further pro-
In a prophage, this repression is carried out ductive infection by another λ (superinfection
by the phage CI protein, which acts by bind- immunity).

Table 1 A list of phage λ modules


Module Genes and sites Function
CI regulator pRM-cI CI activates pRM
Immunity pLoL-rex-cI-pRM-oRpR-cro CI represses pL and pR
CII regulator cII-pRE CII activates pRE
Site-specific recombination xis-int-att-tI Int and Xis catalyze recombination
General recombination gam-exo-bet Control and catalyze recombination
DNA replication ori-O-P Control of initiation of DNA replication
control
Early antitermination NUTL-N-tL1 N-antitermination and translational
control
Late antitermination Q-pR  -tR  Q-antitermination for late gene
expression
DNA packaging cos-Nu1-A Cleavage of head-full genomes
Head genes Nu1 to FII Morphogenesis of head particles
Tail genes Z to J Morphogenesis of tails

Description of the functions of genes not discussed in this review can be found in (38).

412 Oppenheim et al.


ANRV260-GE39-18 ARI 15 October 2005 13:5

The Lytic Transcription Cascade The Lysogenic Process


The gene expression cascade that leads to the The lysogenic pathway is also initiated during
lytic mode of growth is the default mode of λ the transcription cascade of the delayed early
development. It is carried out in three stages. genes. Under conditions leading to lysogeny,
(i) Transcription is initiated with the synthe- the expression of the lytic regulators fails be-
sis of the early transcripts from the pL and cause Q is actively switched off by the accumu-
pR promoters (Figure 2). Early transcripts, lation of the critical lysogenic regulator CII,
which encode two regulators, N and Cro, are thereby blocking the default lytic pathway
attenuated at the tL1 and tR1 terminators, re- and switching to the lysogenic course. As dis-
spectively. These transcriptional terminators cussed in greater detail below, an accumula-
play an important role in controlling the cas- tion of the CII protein above a threshold level
cade of gene expression. By acting as a weak is critical for initiating the lysogenic switch.
repressor for both pL and pR promoters, Cro During this active switching from the default
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

facilitates the lytic mode (described below). lytic to the lysogenic mode, CII stimulates the
by University of California - San Diego on 03/29/11. For personal use only.

The N protein is an antitermination factor synthesis of Int, which catalyzes the insertion
that promotes the assembly of a transcription of the phage DNA into the host chromosome,
complex (9, 35). This assembly occurs on the and of the CI regulator, which binds to oL and
RNA at the nutL and nutR sites and is made up oR to repress the early promoters. CII also in-
of RNA polymerase and a number of host pro- hibits Q function (see 52). These three activ-
teins called Nus. The N- and Nus-modified ities are mediated by transcription activation
RNA polymerase can overcome the tL1 and of three promoters, pI, pRE, and paQ, respec-
tR1 transcription terminators, resulting in ex- tively (Figure 2; see Figure 6 below). Activa-
pression of the distal delayed early functions tion of all three promoters is critical for the
(30, 84). (ii) The delayed early functions in- establishment of a stable prophage state. Once
clude the lysogenic regulators CII and CIII, the prophage genome integrates into the bac-
as well as the lytic DNA replication functions terial chromosome and CI protein represses
O and P, and the late gene regulator Q. (iii) pL and pR, the lysogenic state is established.
After sufficient accumulation, the Q protein The prophage state is extremely stable and is
modifies RNA polymerase that has just initi- maintained through many generations of divi-
ated transcription from the pR late promoter sion (8). How CI repressor synthesis is main-
(66). This modification causes the RNA poly- tained in the absence of CII is discussed later.
merase to become resistant to transcription
terminators present downstream to pR , al-
lowing the expression of the late genes, which THE PROPHAGE STATE
encode proteins for phage morphogenesis and AND ITS MAINTENANCE
host cell lysis. There is a kinetic separation In the prophage state, the CI regulator con-
between the expression of delayed early and trols the expression of three promoters. It
late genes. This is caused by the location of represses transcription from the pL and pR
the Q gene at the very end of the delayed promoters and positively and negatively reg-
early cascade and the high threshold level of ulates its own synthesis from the pRM pro-
Q protein needed for its activity (52, 63, 109). moter (Figure 3; see Figure 6 below). In the
During the late stage of the cascade, the late prophage state, pRM is responsible for CI syn-
gene products assemble phage virions and lyse thesis; CI expression from pRE is prevented
the host. A similar temporal lytic cascade of owing to repression of CII in a lysogen.
gene expression follows prophage induction Figure 3 illustrates a set of cooperative in-
(38). teractions of CI binding to DNA, which lead

www.annualreviews.org • Switches in Bacteriophage Lambda Development 413


ANRV260-GE39-18 ARI 15 October 2005 13:5

to extremely efficient repression (25, 64, 82).


The CI repressor is made of two domains teth-
ered by a short linker, an N-terminal DNA
binding domain with a helix-turn-helix mo-
tif, and a C-terminal oligomerization domain.
The intrinsic relative affinities of CI to its op-
erators at oR and oL are as follows: oR1 >
oR2 > oR3 and oL1 > oL2 > oL3. Dimers of
CI cooperatively bind to oL1 and oL2 on the
left and oR1 and oR2 on the right by forming
tetramers repressing pL and pR, respectively.
Furthermore, another cooperative interaction
between these two sets of tetramers bound to
oL and oR, 2.3 Kb apart, leads to the formation
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

of a DNA loop held by a CI octamer, i.e., two


by University of California - San Diego on 03/29/11. For personal use only.

interacting tetramers, that enhances repres-


sion. In this DNA-multiprotein complex, the
CI dimer bound at oR2 also stimulates pRM
transcription, thus activating CI synthesis in
a repressed prophage by a positive autoregu-
latory loop (44, 75). As the CI concentration
increases because of pRM activation, two ad-
ditional CI dimers are recruited to bind at oL3
and oR3 to further stabilize the oL-oR loop. In
this context, the binding of CI to oR3 represses
pRM and prevents CI overexpression. Based
on direct measurements of CI (86), recent cal-
culation shows that a lysogenic cell contains
about 250 CI monomers when the cells are
growing in rich media (23). This translates
into about 30 dimers per prophage copy, as-
suming an average of four chromosomes per
cell. This number of CI molecules not only
achieves a strong repression of the phage pro-
Figure 3
moters but also sets the threshold level for
A scheme showing the repression of pL and pR by CI induced DNA SOS induction. Note that cell division does
looping [adapted from (24, 82)]. RNA polymerase is symbolized as a green
ellipse; CI regulator monomers (depicted as orange dumbbells) have two
not randomly reduce the CI concentration
domains: the N-terminal DNA binding domain and the C-terminal to a point low enough to cause prophage
oligomerization domain. The top line shows a linear arrangement of the induction (8).
DNA. In the absence of CI, transcription is initiated at both pL and pR (top Repression of transcription from the pR
line). The second line shows how repressor molecules bind cooperatively promoter inhibits not only expression of genes
to oL1/oL2 and to oR1/oR2 prior to DNA looping repressing pL and pR
and activating the pRM promoter. The third line shows the first stage of
in that operon but also phage DNA repli-
DNA looping. RNA polymerase bound to pRM either before (second line) cation by preventing “transcriptional activa-
or after (third line) looping occurs transcribes pRM. Finally, further tion” of λ ori, the site where phage DNA
stabilization of DNA looping occurs by binding of CI to oL3 and oR3 replication is initiated (26, 33). This inhibition
together with repression of pRM when CI levels become very high ( fourth occurs even if O and P functions are present.
line). The additional CI molecules bound to oL3 and oR3 interact to form a
tetramer. The SOS response reverses the repression process (see text).
(71, 106). This regulation appears to be crit-
ical for establishing a lysogen. If the λ origin

414 Oppenheim et al.


ANRV260-GE39-18 ARI 15 October 2005 13:5

replicates after the phage is integrated because pL and pR promoters and that its role in turn-
O and P are still available, the replication ing down pRM is not critical but supplemen-
event is lethal to the cell, resulting in abortive tary (52, 94). An argument that Cro binding
lytic infection (11). Thus, it is important to to oR3 sets the lytic course has also been
immediately block phage replication as the made to explain the role of Cro following
choice for lysogeny is made. phage infection (82). This interpretation of
the role of Cro is also unlikely because a phage
carrying the same oR3 to oR1 variant (see
THE DEFAULT LYTIC COURSE: above) that reduces Cro but allows CI bind-
Cro AND N FUNCTIONS ing still shows lytic growth (64). However, un-
The gene coding for Cro, which is the essen- der certain conditions Cro binding to oR3 may
tial lytic regulator, is the first one to be tran- contribute to lytic growth after prophage in-
scribed from the pR promoter following phage duction. The function of Cro in lytic devel-
infection or prophage induction. It is a weak opment is addressed below.
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

repressor of the pR promoter, allowing contin-


by University of California - San Diego on 03/29/11. For personal use only.

uous expression from pR during lytic infection


(29, 94, 95). Cro is a single-domain protein The N Antiterminator
that binds as a dimer to the oL and oR opera- The N protein, like Cro, is a critical lytic
tors (60, 82). The intrinsic affinity of Cro for regulator but acts by a completely different
the individual operators is oR3 > oR1 ≥ oR2, mechanism. It antiterminates transcription at
and, unlike CI, higher-ordered structures, i.e., termination signals, allowing expression of
tetramers or octamers of Cro have not been distal genes in the pL and pR operons. N pro-
detected (19). However, Ackers and cowork- tein, once made, binds to the specific RNA
ers found a small amount of cooperative bind- sites NUTL and NUTR. The NUTL site
ing of Cro dimers to adjacent sites in the is located between the N gene and the pL
oR complex (19). Whether such cooperative promoter, and NUTR is downstream of cro,
binding is due to dimer-dimer interaction or the first gene in the pR operon (Figure 1).
to changes in DNA needs to be investigated. It The Nus factors, a complex set of host pro-
was shown, however, that changes in DNA ac- teins, take part in cellular transcriptional and
company the cooperative binding of CI (21). translational processes and interact with the
The higher affinity of Cro binding to oR3 N system (18, 93). Purified in vitro studies
supports the hypothesis that during lytic de- have defined NusA, NusB, NusE, and NusG
velopment Cro binds first to oR3, repressing as the host components of N-antitermination
transcription of the cI gene from the pRM (22, 68). Each NUT site can be divided into
promoter (45). It was therefore proposed that two parts, BOXA and BOXB (32). BOXB is
Cro binding to oR3 represses pRM and com- a stem-loop structure in the RNA and is spe-
pletes the switch to the lytic pathway as well cific for N binding (16). As it binds BOXB,
as sustaining it after an initial SOS-mediated N associates with NusA, NusG, and RNAP.
inactivation of CI in a lysogen (28, 46, 74). BOXA RNA is specific for NusB and NusE
However, recent experiments suggest that the binding (77, 80). It is proposed that N, Nus
presence of Cro might be unimportant for the factors, and Nut interact and complex with
lysogenic to lytic switch during induction of RNA polymerase while tethered on the same
the prophage (94). Furthermore, Little and RNA (76, 102, 104, 105). Although N is the
coworkers showed that replacing oR3 by the essential factor for antitermination, NUT and
weaker oR1 Cro-binding site has a marginal the Nus factors confer stability and full activ-
effect on prophage induction (64). Thus, it ity to the antitermination complex (22, 85).
appears that the critical role of Cro in lytic N is a critical regulatory protein of phage
development may be only to turn down the λ, which is reflected in the number of

www.annualreviews.org • Switches in Bacteriophage Lambda Development 415


ANRV260-GE39-18 ARI 15 October 2005 13:5

N expression from the pL-antiterminated


transcripts is dependent upon the level of
RNase III in cells. If RNaseIII concentration
is high, processing of the RNase III site (RIII
site) between NUTL and the N message is
rapid, causing high rates of N translation. If
RNase III concentration is low or zero, pro-
cessing of the RIII site occurs slowly if at
all, allowing repression of N translation. Cel-
lular RNase III concentrations rise and fall
with growth rate (12), and correspondingly,
N translation rises and falls with growth rate
(104).
Evidence suggests that the N translation
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

repression complex is dependent on E. coli


by University of California - San Diego on 03/29/11. For personal use only.

RNAP and the N-transcription antitermina-


tion complex (105). Under conditions of lim-
iting RNase III, the antiterminating complex
prevents translation of N from its antitermi-
nated message. This means that N is expressed
from RNA transcripts that terminate at tL1,
i.e., do not form an antitermination com-
Figure 4
plex. Thus, under minimal growth conditions
Models for N-mediated translation repression. The complex of RNAP is
where RNase III activity is limiting, N con-
modified by NUT, N, and Nus factors to form an antitermination
complex. Translation repression of N takes place on the uncleaved RNA centrations would be stringently controlled by
in the absence of RNase III action (a). In the presence of RNase III negative autoregulation. On the other hand,
cleavage occurs and the Shine–Dalgarno (SD) sequence for N is no longer in rich media, RNase III processing would
held in close contact to the antitermination complex, translational ensure high N concentrations. Viral lytic de-
repression is prevented, and efficient expression of N can take place (b).
velopment predominates in rich media, and
lysogenic development is enhanced under
limiting growth or starvation. High N con-
ways its expression is controlled. At the centration enhances lytic growth of the phage,
transcription level, N is regulated from oLpL as evidenced by λ forming clear plaques on
by CI- and Cro-mediated repression. At cells expressing N from a plasmid (104). Clear
the translation level, N represses and au- plaques indicate that only few cells follow the
toregulates its own translation, and the en- lysogenic pathway. Low levels of N are made
doribonuclease RNase III modulates this following λ infection of cells grown in poor
repression (Figure 4). By acting at their carbon conditions; these same conditions have
respective sites located upstream of the N been shown to enhance lysogen formation fol-
gene, N and RNase III control N translation lowing infection at low multiplicity of infec-
(103, 104). High concentrations of RNase III tion (56). These latter conditions are likely to
as found in cells grown in rich media pre- reduce Q concentrations and provide a bet-
vent N-mediated translation repression alto- ter opportunity for lysogeny. Regulation of N
gether, whereas low concentrations of RNase also appears to be a way in which λ senses envi-
III found in cells grown in poor media all- ronmental conditions through RNase III and
ow translation repression. At the posttrans- responds by increasing or decreasing N con-
lational level, the Lon protease degrades N centrations and altering the lytic/lysogenic
causing a relatively short half-life. response. We note, however, that at high

416 Oppenheim et al.


ANRV260-GE39-18 ARI 15 October 2005 13:5

multiplicity of infection, there is no effect (88). This mutation affects the N-terminal se-
of growth media on lysogenization efficiency quence and is not in the helix-turn-helix do-
(27). main of CII. This mutation also modified the
pRE promoter so that CII can activate the
mutant pRE but not the wild-type pRE. This
SWITCHING THE DEFAULT suggests that this N-terminal sequence of the
LYTIC MODE TO LYSOGENY: protein may play a role in determining the
ROLE OF CII specificity of CII binding to DNA (88).
The inhibition, or absence, of lytic functions The pI, pRE, and paQ promoters are lo-
is not sufficient for the switching to the lyso- cated within the Xis, CII, and Q protein cod-
genic mode. Rather, as noted above, infec- ing sequences, respectively. The Xis protein
tion resulting in a lysogenic response proceeds is needed for excision of the prophage DNA
through a number of required events: inte- from the chromosome after induction. For
gration of the DNA, efficient repression of rapid synthesis of Int, which facilitates inte-
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

the early promoters, as well as timely inhibi- gration after infection, CII activates the pI
by University of California - San Diego on 03/29/11. For personal use only.

tion of the lytic genes expression. These re- promoter. The integration reaction also re-
quirements are met by CII turning on pI, pRE, quires the integration host factor, IHF. IHF
and paQ promoters to express Int and CI, and is critical for generating the multicomponent
to inhibit Q function, respectively. All three Intasome structure, which catalyzes the in-
promoters contain a direct repeat TTGC- tegration reaction (38). Since pI is located
N6-TTGC sequence that binds to CII. The within the xis gene, the pI transcript synthe-
N6 region corresponds to the –35 elements sizes Int but not Xis, helping to ensure that
of these promoters. Expression from all three integration will not be accompanied by the
CII-activated promoters is coordinated by the presence of Xis function. By the same crite-
CII protein during infection. However, the rion, CII activates the pRE promoter to direct
mechanism of activation of the promoters by rapid synthesis of the CI regulator following
CII is not well understood. Specific muta- infection. The amount of CI made from pRE
tions in the α or σ subunits of RNAP prevent during lysogenic response was found to be
CII-mediated activation from these promot- as much as 10- to 20-fold higher than the
ers in vitro (48, 67). Consistently, these RNA amount made from pRM in an established
polymerase mutants prevent the establish- lysogen (86). The initial high concentrations
ment of lysogeny in vivo (78). of CI may guarantee that all infecting and
The quaternary structure of the CII pro- replicating phage genomes become repressed.
tein has been solved recently (20a, 81a). The But the CII-dependent paQ promoter, which
structure shows that a CII tetramer is made of lies within the Q gene, was found to reduce
two nearly equivalent dimers. Each of the four Q function (52), providing a mechanism by
monomers contains a helix-turn-helix DNA- which CII reduces late gene expression to en-
binding motif but only two of the monomers hance lysogeny (17, 70). Mutations affecting
appear, by modeling, to be involved in actual the ability of the pRE or paQ promoter to re-
DNA binding at the direct repeat sequence. spond to CII prevent the lysogenic response
The function of the other two helix-turn-helix resulting in clear plaque formation (42, 52).
motifs in the tetramer is not known. The di-
rect repeat sequence of pRE is located within
the N-terminal coding sequence of CII. Inci- Regulation of CII Activity
dentally, in an elegant genetic study, Friedman CII, which plays a key role in the lysis-
and coworkers isolated and analyzed a CII lysogeny decision, is regulated at numerous
mutant defective in transcription activation levels (Figure 5) (39, 43, 50, 83, 91): (i) The
of the pE promoter in lambdoid phage P22 transcription of the cII gene is inhibited both

www.annualreviews.org • Switches in Bacteriophage Lambda Development 417


ANRV260-GE39-18 ARI 15 October 2005 13:5
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

Figure 5
by University of California - San Diego on 03/29/11. For personal use only.

Multilevel regulation of CII activity. CI and Cro negatively regulate the CII gene at transcription
initiation. tR1 aided by Rho factor reduce transcription elongation into the CII gene. The N
antitermination factor allows the extension of transcription beyond tR1. IHF stimulates CII translation
initiation. The antisense OOP RNA together with RNase III reduces CII mRNA stability, and FtsH
protease acting at the C terminus of CII is responsible for the rapid proteolysis of CII. The center bar
represents the DNA; the CII gene and the positive controls are shown in blue and negative controls in
red. The direction of CII and OOP transcriptions are shown in blue and red, respectively.

by Cro and CI binding to oR, and is stimu- lysogenization frequency. A C-terminal flex-
lated by the N antitermination factor acting ible tail of CII, which is not required for
at NUTR. High rates of CII synthesis take CII activity, acts as a target for initiating
place only for a limited period before being rapid CII proteolysis by FtsH (20, 51). (v)
repressed, by Cro or CI. (ii) Translation initi- The long leader CI RNA initiated from pRE
ation of CII is stimulated by IHF (43). An IHF is antisense to cro, which could prevent the
binding site is located immediately upstream translation of Cro (92). Indeed, an ftsH host
of CII, which has been proposed to stimu- mutant that results in higher concentrations
late CII translation in the presence of IHF of CII was found to be defective in Cro (79,
by an unknown mechanism (72a, 80a). (iii) 83, 96). Unfortunately, the concentration of
The stability of CII mRNA is affected by the Cro as a function of pRE activity has not
OOP RNA, a short antisense transcript com- been directly measured. (vi) CIII, which is
plementary to the 3 end of the cII mRNA (57, a 54-residue long peptide and required for
58). RNase III recognizes and cleaves the CII- lysogeny, controls the rate of CII degradation
OOP double-stranded RNA, thereby initiat- by acting as an inhibitor of the FtsH protease
ing rapid CII mRNA degradation (57). The (40, 51, 53).
DNA coding for the protease target is also the
target of CII mRNA degradation mediated by
the antisense OOP RNA. It was reported that Regulation of CIII
the stability of the OOP RNA is reduced by The cIII gene expression is also subject to mul-
polyadenylation but whether this process reg- tiple controls by λ CI, Cro, and N and by the
ulates CII concentration has not been clarified host RNase III (Figure 5). CI and Cro in-
(108). (iv) The ATP-dependent protease FtsH hibit CIII synthesis by binding to oL, and the
is responsible for the rapid degradation of CII N-antiterminator stimulates CIII expression
(39, 50, 91). Host mutations in ftsH lead to by acting at NUTL. Unlike its negative effect
stabilization of CII and thereby an increased on CII, RNase III stimulates CIII translation.

418 Oppenheim et al.


ANRV260-GE39-18 ARI 15 October 2005 13:5

It was shown that the mRNA that codes for Following infection, transcription from
the amino terminal residues of CIII is present early pL and pR promoters would start the lytic
in two conformations (3, 54). In one con- pathway by default with N antitermination of
formation, the translation initiation region is transcription and subsequent expression of Q.
occluded, preventing cIII translation. In the Q in turn antiterminates transcription, lead-
other, the mRNA is open allowing efficient ing to late lytic gene expression, cell lysis, and
translation. Point mutations that favor one or phage release. To set the course for lysogeny,
the other structures have been described. It CII reduces Q function in two ways. First,
appears that RNase III regulates cIII trans- CII activates antisense paQ RNA inhibiting
lation by acting as an RNA chaperone to af- Q. Second, CII activates pRE for synthesis of
fect CIII mRNA structure without processing CI, which represses pR and thus Q transcrip-
(2). tion. CII continues to repress Q expression
via paQ until cII transcription is repressed by
CI at pR. This kinetic coordination of Q shut-
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

THE DECISION PROCESS: off by CII and CI ensures the switch from the
by University of California - San Diego on 03/29/11. For personal use only.

GENETIC FUNCTIONS AND default lytic pathway to lysogeny. Q protein


EXTERNAL INFLUENCES must build up to a high threshold concen-
We now summarize the critical events that tration to become functional, thus providing
take place in the decision making process fol- a window for CII to exert its effect on Q
lowing infection by λ (Decision I in Figure 1, through paQ. In this way, CII prevents Q from
and shown schematically in Figure 6). Fol- reaching its threshold concentration. If CI ac-
lowing infection, λ begins a cascade of tran- tion is prevented, CII inhibition of Q via paQ
scription destined for the lytic mode. Under is not sustained because of repression of CII
appropriate conditions the lytic cascade can be by Cro acting at pR and by rapid CII degra-
switched off, allowing entry into the lysogenic dation. Concordantly, λ phage mutants defec-
pathway. The switch requires high thresh- tive in CI, oR, or CII follow exclusively lytic
old concentrations of CII, which facilitates growth, whereas mutants defective in Cro are
CI and Int synthesis and reduces the regu- unable to follow the lytic pathway.
lator function Q. CI, once made, maintains
the repressed state of the prophage. In es-
tablishing repression, CII and CI act tempo- Multiplicity Effect
rally to inhibit lytic functions. CII acts first The lysis-lysogeny decision is influenced by
by direct reduction of Q function, and as CII the number of phage particles infecting the
function ceases, CI takes over by repressing cell as well as by the cell physiology (55, 56).
the pR promoter. CI executes repression after High multiplicity of infection favors lysogeny.
the CII-mediated switch and appears not to Direct measurements of CII activity showed,
participate in initiating the switch. The activ- in wild-type infection, functional CII activ-
ity of CII is programmed to allow an initial ity in cells infected with two or more phages
overshoot of CI expression from the pRE pro- (52). This threshold level of CII is reached
moter and a preemptive inhibition of Q activ- when two or more phage infect a cell. In sin-
ity (52, 86). The overshoot may be required gle infection this critical level of CII is not
to ensure the repression of all lytic genes of attained, as mentioned above, allowing lytic
the infecting and replicating phage genomes development.
in the cell during the establishment of repres-
sion. Repression of pR by CI at this stage
also ensures immediate cessation of phage Physiological Effects
DNA replication by preventing transcription In cells grown in rich media the lytic course
through the ori site (97). predominates, whereas growing cells in poor

www.annualreviews.org • Switches in Bacteriophage Lambda Development 419


ANRV260-GE39-18 ARI 15 October 2005 13:5
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org
by University of California - San Diego on 03/29/11. For personal use only.

Figure 6
Description of the λ lytic and lysogenic genetic network. Arrows mark the positive effects between
elements that make up the genetic network, whereas bars denote inhibitory effects. Promoters and
operators are shown in green, the phage genes in light orange, and cis antisense RNA is shown in light
blue (Cis acting has been used to note that it acts on the RNA from the opposite strand and not on an
RNA originating from different sequences in the genome). Arrows emanating from the promoters
denote transcription of specific functions. The OOP antisense RNA and the yet unknown threshold
effect of Q activity are not shown. For simplicity, the developmental network is divided into early gene
expression (a), delayed gene expression (b), lytic gene expression (c), lysogenic establishment (d ), and
lysogenic maintenance (e). The dotted line in (b) leading to Int expression signifies reduced level of Int
expression due to retroregulation of int mRNA.

media increases the chances of the lyso- the protein N favoring lytic growth [see (104)
genic pathway after single infection (56). Cells and references therein]. In carbon-starved
growing in rich media have higher concen- cells, on the other hand, RNase III and con-
tration of the host global regulator RNase sequently N concentrations are low. Under
III, which leads to high rates of expression of these conditions N translation is repressed.

420 Oppenheim et al.


ANRV260-GE39-18 ARI 15 October 2005 13:5

This reduction of N concentration would re- regulator Q activates the synthesis of proteins
duce Q expression to a level that provides for phage morphogenesis and cell lysis. Thus
more opportunity for lysogenic response. functions necessary for lysogenic and lytic de-
Nevertheless, when cells are infected by two velopment are both expressed from the pL and
or more phages, CII function is epistatic to pR promoters. The location of both lysogenic
the effects of growth conditions preferring and lytic genes in the same operons creates an
lysogeny. apparent paradox in our mind about the lysis-
Unlike the decision that λ makes after the lysogeny decision. This paradox is resolved
infection process described above whereby in- by regulating the synthesis and degradation
fected cells follow either lytic or lysogenic of critical RNA and proteins. This regula-
development, prophage induction leads ex- tion provides the catalytic or stoichiometric
clusively to the lytic course (Decision II in amounts of functions as required to pursue
Figure 1). It was proposed that Cro is respon- either the lytic or lysogenic course. As exam-
sible for repressing CI expression following ples, during lytic response, the CII regulator is
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

induction to keep the developmental switch rapidly eliminated by proteolysis, whereas in


by University of California - San Diego on 03/29/11. For personal use only.

in the default lytic mode (82), although the the lysogenic response, CII accumulates and
action of Cro in keeping the lytic course on limits Q activity. Presumably, the high thresh-
track is simply to reduce the concentration of old requirement for Q function allows the in-
the unstable master lysogenic regulator CII. fected complex a time window for controlling
the concentration of CII.
In the lysogenic response, high concentra-
COEXPRESSION OF BOTH tions of Int are made from pI under the control
LYTIC AND LYSOGENIC GENES of CII, while the expression of Int from the pL
FROM THE SAME OPERON: A is greatly reduced by retro-regulation because
PARADOX of Int mRNA degradation by RNase III from
The pL operon encodes a large number of a site called sib, located on the other side of att
ORFs of which only N, CIII, Xis, and Int are (see Figure 2) (36). Prophage induction leads
essential for either the lytic or lysogenic re- to the expression of both Int and Xis from
sponse (see Figure 2). N is a key regulator the pL promoter because the sib regulator is
for lytic growth of the phage. On the other detached from the int gene in the prophage
hand, the lysogenic function CIII acts as an DNA.
inhibitor of a host protease (FtsH, also called
HflB) that destabilizes the critical lysogenic
regulator CII (40, 47, 51). Proteins Int and Xis THEORETICAL STUDIES
carry out site-specific recombination (59, 73). The lambda genetic network has been a fertile
During lysogenization, Int catalyzes the inte- ground for theoretical modeling of decision-
gration of the phage genome into the bacterial making processes during the regulation of
chromosome site, att, whereas Int together development, and for testing new modeling
with Xis catalyze the reverse reaction to excise methodologies of genetic networks in general.
the λ chromosome during prophage induc- Models have been constructed addressing the
tion. The pR promoter also transcribes both lysis-lysogeny decision in terms of the lambda
lytic and lysogenic genes, cro, cII, O, P, and Q. genetic switch describing the competition
Whereas Cro is another critical regulator for between CI and Cro, using statistical mechan-
lytic growth of the phage, the very next gene ics with the explicit goal of obtaining bista-
in the operon encodes the CII protein, which bility. The earlier models focused on the (i)
coordinates the lysogenic pathway. O and P probabilities of the occupancy of the pR and
are needed for phage DNA replication prior pRM promoters by different binding configu-
to morphogenesis, whereas the second lytic rations of CI and Cro, and of pR and pRM by

www.annualreviews.org • Switches in Bacteriophage Lambda Development 421


ANRV260-GE39-18 ARI 15 October 2005 13:5

RNAP, and (ii) Gibbs free energies of binding inconsistencies with the observed behavior,
characterizing the different configurations as future theoretical studies with predictive val-
free parameters (1, 87, 90). These models ues are expected to play a more important role.
assumed that the behavior of the reactants
studied under in vitro conditions reflects
in vivo situations, and did not incorporate the OPEN QUESTIONS, SUMMARY,
possible existence of other levels of regulation, AND CONCLUSION
e.g., regulation of translation of CI and Cro,
degradation of CI and Cro, and anticooper-
Counting Infecting Phage
ative binding of the two proteins to adjacent The decision made by a phage-infected cell
operators (87). is dependent on the multiplicity of infec-
More recent models, based on the same ap- tion. When one phage infects, most infec-
proach, also incorporated stochastic effects in tion shows lytic development. However, when
the concentrations of the regulatory proteins two phage infect, the lysogenic pathway pre-
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

and, as a result, in the selection between lysis vails. What is the molecular basis for such a
by University of California - San Diego on 03/29/11. For personal use only.

and lysogeny (5, 7, 49, 69, 89, 98, 100, 110). dramatic response to a small change, which
Although apparent agreement between the to some may be counterintuitive? Further-
theoretical calculations and the experimen- more, the network response also suggests
tally observed values was noted, these efforts tight communication between two coinfect-
did not lead to a theoretical description with ing phage genomes. This suggests that repli-
improved predictive values. However, the the- cating phage genomes after single phage
oretical analysis of the exceptional stability the infection have little effect on the decision pro-
prophage state of λ led to the conclusion, now cess. This issue requires further investigation.
confirmed by experimental evidence, that a However, there are conditions when a single
view of the switch focusing only on oR to ex- infecting phage enters the lysogenic pathway.
plain the stability is incomplete, and that oL When infecting an hflA or hflB mutant host,
participates as well (7, 23). efficient lysogenization takes place. As dis-
An advantage of computer simulations is cussed, these mutant hosts increase the level
their ability to take into account multiple ele- of CII function.
ments and variables within the decision mod- A possible parsimonious model to explain
ule of the λ network (5, 49, 69). Furthermore, the multiplicity response runs as follows: First,
they can predict, for example, the values of multiple infection results in the titration of a
reactant concentrations during the temporal critical regulatory host factor that is present
execution of the lytic and lysogenic pathways, at a very low concentration. One such can-
which can be readily compared with biological didate is FtsH, the product of the hflB gene
experiments. Nevertheless, at present there of which there are less than 100 molecules
are only limited experimental data on the ki- per cell (99). Second, the phage CIII protein
netic changes in the concentration and activ- acting as an inhibitor of FtsH plays a critical
ity of the regulatory elements in the network role in the decision. Indeed, mutants that re-
and the strength of their interactions, which sult in a elevated CIII translation (by about
are crucial to achieve real agreement between threefold) no longer respond to the multi-
theoretical results and experimental observa- plicity of infection and can efficiently lyso-
tions. Furthermore, the in vivo values of the genize upon single infection (4; A. Rattray,
parameters may differ quantitatively by orders unpublished). Thus, we expect that a small in-
of magnitude from in vitro ones owing to such crease in multiplicity from one to two would
factors as macromolecular crowding, varia- raise CIII to concentrations that inhibit FtsH
tion in local concentrations, and yet unknown increasing CII expression to allow lysogenic
functions, as alluded to above. By exposing development.

422 Oppenheim et al.


ANRV260-GE39-18 ARI 15 October 2005 13:5

The Distinction Between Phage Evolution of Temperate Phages


Infection and Prophage Induction Phage λ lifestyles have been a textbook case
The idea that two infecting phage are critical for complex genetic control circuits. Gene or-
for switching on the lysogenic course raises der, modular construct, recruitment of spe-
interesting questions. A lysogen contains a cific host factors, and the presence of a
number of host chromosomes at various complex genetic network allow the infect-
stages of replication, and accordingly more ing phage to make decisions and to proceed
than one prophage. Would induction result through alternative developmental pathways.
in high CII activity as is found in multiple The lysis-lysogeny bistable decision is clearly
infections? If a high concentration of CII ac- much more complex than originally por-
cumulates, we would expect inhibition of Q trayed. This complexity was most probably
function by CII, even after CI inactivation by reached through evolutionary forces tinker-
induction. It remains to be seen whether such ing with specific elements present in the indi-
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

high concentrations of CII if they occur would vidual functional modules that evolved to act
by University of California - San Diego on 03/29/11. For personal use only.

affect Q function and thus reduce lytic phage in concert. Of interest is a recent approach
yield. of tinkering with λ modules that revealed the
robustness of its genetic network (6, 72). Al-
though the λ lysogenic promoter pRM can tol-
Role of Cro in Lytic Growth erate a number of mutational changes, we note
An open question is the role of the high- that such tolerance may be limited to specific
affinity binding of Cro to the oR3 operator environmental conditions.
site in lytic growth after phage infection or
prophage induction. It is clear that Cro is
essential to allow lytic development. Is this EPILOG
high-affinity binding critical to regulate pRM In summary, a small set of regulatory proteins
following prophage induction or does it have in an organism as simple as a bacteriophage
a critical role in lytic decision? This issue function through a diverse set of macro-
needs to be addressed by the use of phage molecular interactions in a temporal fashion.
mutants defective in oR3 that uniquely affect Future investigations into the detailed molec-
Cro binding and do not affect CI binding or ular aspects of the functions of specific mod-
pRM activity. Would such mutants affect lytic ules coupled with kinetic and quantitative
growth after phage infection or prophage in- analysis of the phage genetic network will
duction? The intercalation of CI, Cro, and yield a realistic picture of this important
RNAP binding site at this locus may make the paradigm for more complex developmental
isolation of such phage mutants problematic. processes (61).

ACKNOWLEDGMENTS
We apologize to our colleagues whose work we have omitted. We thank John Little, Ian Dodd,
Ted Cox, Pradeep Parrack, and Grzes Wegrzyn for communication of manuscripts before
publication and David Friedman for critical reading of the manuscript. The research carried
out by O. K. and A. B. O. was supported in part by The Israel Science Foundation (grants #
489/01–1 and 340/04).

LITERATURE CITED
1. Ackers GK, Johnson AD, Shea MA. 1982. Quantitative model for gene regulation by
lambda phage repressor. Proc. Natl. Acad. Sci. USA 79:1129–33

www.annualreviews.org • Switches in Bacteriophage Lambda Development 423


ANRV260-GE39-18 ARI 15 October 2005 13:5

2. Altuvia S, Kornitzer D, Kobi S, Oppenheim AB. 1991. Functional and structural elements
of the mRNA of the cIII gene of bacteriophage lambda. J. Mol. Biol. 218:723–33
3. Altuvia S, Kornitzer D, Teff D, Oppenheim AB. 1989. Alternative mRNA structures of
the cIII gene of bacteriophage lambda determine the rate of its translation initiation. J.
Mol. Biol. 210:265–80
4. Altuvia S, Oppenheim AB. 1986. Translational regulatory signals within the coding region
of the bacteriophage lambda cIII gene. J. Bacteriol. 167:415–19
5. Arkin A, Ross J, McAdams HH. 1998. Stochastic kinetic analysis of developmental path-
way bifurcation in phage lambda-infected Escherichia coli cells. Genetics 149:1633–48
6. Atsumi S, Little JW. 2004. Regulatory circuit design and evolution using phage lambda.
Genes Dev. 18:2086–94
7. Aurell E, Brown S, Johanson J, Sneppen K. 2002. Stability puzzles in phage lambda. Phys.
Rev. E 65:051914-1-9
8. Baek K, Svenningsen S, Eisen H, Sneppen K, Brown S. 2003. Single-cell analysis of
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

lambda immunity regulation. J. Mol. Biol. 334:363–72


by University of California - San Diego on 03/29/11. For personal use only.

9. Barik S, Ghosh B, Whalen W, Lazinski D, Das A. 1987. An antitermination protein


engages the elongating transcription apparatus at a promoter-proximal recognition site.
Cell 50:885–99
10. Botstein D. 1980. A theory of modular evolution for bacteriophages. Ann. NY Acad. Sci.
354:484–90
11. Brachet P, Eisen H, Rambach A. 1970. Mutations of coliphage lambda affecting the
expression of replicative functions O and P. Mol. Gen. Genet. 108:266–76
12. Britton RA, Powell BS, Dasgupta S, Sun Q, Margolin W, et al. 1998. Cell cycle arrest
in Era GTPase mutants: a potential growth rate-regulated checkpoint in Escherichia coli.
Mol. Microbiol. 27:739–50
13. Calender RL, ed. 2005. The Bacteriophages. New York: Oxford Univ. Press. 2nd ed.
14. Campbell A. 1994. Comparative molecular biology of lambdoid phages. Annu. Rev. Mi-
crobiol. 48:193–222
15. Campbell A. 2003. The future of bacteriophage biology. Nat. Rev. Genet. 4:471–77
16. Chattopadhyay S, Garcia-Mena J, DeVito J, Wolska K, Das A. 1995. Bipartite function
of a small RNA hairpin in transcription antitermination in bacteriophage lambda. Proc.
Natl. Acad. Sci. USA 92:4061–65
17. Court D, Green L, Echols H. 1975. Positive and negative regulation by the cII and cIII
gene products of bacteriophage lambda. Virology 63:484–91
18. Court DL, Patterson TA, Baker T, Costantino N, Mao X, Friedman DI. 1995. Struc-
tural and functional analyses of the transcription-translation proteins NusB and NusE.
J. Bacteriol. 177:2589–91
19. Darling PJ, Holt JM, Ackers GK. 2000. Coupled energetics of lambda cro repressor self-
assembly and site-specific DNA operator binding I: analysis of cro dimerization from
nanomolar to micromolar concentrations. Biochemistry 39:11500–7
20. Datta AB, Roy S, Parrack P. 2005. Role of C-terminal residues in oligomerization and
stability of lambda CII: implications for lysis-lysogeny decision of the phage. J. Mol. Biol.
345:315–24
20a. Datta AB, Panjikar S, Weiss MS, Chakrabarti P, Parrack P. 2005. Structure of λCII:
Implications for recognition of direct repeat DNA by an unusual tetrameric organization.
Proc. Natl. Acad. Sci. USA. In press
21. Deb S, Bandyopadhyay S, Roy S. 2000. DNA sequence dependent and independent
conformational changes in multipartite operator recognition by lambda-repressor. Bio-
chemistry 39:3377–83

424 Oppenheim et al.


ANRV260-GE39-18 ARI 15 October 2005 13:5

22. DeVito J, Das A. 1994. Control of transcription processivity in phage lambda: Nus factors
strengthen the termination-resistant state of RNA polymerase induced by N antitermi-
nator. Proc. Natl. Acad. Sci USA 91:8660–64
23. Dodd IB, Perkins AJ, Tsemitsidis D, Egan JB. 2001. Octamerization of lambda CI re-
pressor is needed for effective repression of P(RM) and efficient switching from lysogeny.
Genes Dev. 15:3013–22
24. Dodd IB, Shearwin KE, Egan JB. 2005. Revisited gene regulation in bacteriophage λ.
Curr. Opin. Genet. Dev. 15
25. Dodd IB, Shearwin KE, Perkins AJ, Burr T, Hochschild A, Egan JB. 2004. Cooperativity
in long-range gene regulation by the lambda CI repressor. Genes Dev. 18:344–54
26. Dove WF, Inokuchi H, Stevens WF. 1971. In The Bacteriophage Lambda, ed. AD Hershey,
pp. 747–71. Cold Spring Harbor, NY: Cold Spring Harbor Lab. Press
27. Echols H, Green L, Kudrna R, Edlin G. 1975. Regulation of phage lambda development
with the growth rate of host cells: a homeostatic mechanism. Virology 66:344–46
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org
by University of California - San Diego on 03/29/11. For personal use only.

28. Eisen H, Brachet P, Pereira da Silva L, Jacob F. 1970. Regulation of repressor expression
in lambda. Proc. Natl. Acad. Sci. USA 66:855–62
29. Folkmanis A, Maltzman W, Mellon P, Skalka A, Echols H. 1977. The essential role of
the cro gene in lytic development by bacteriophage lambda. Virology 81:352–62
30. Friedman DI, Court DL. 1995. Transcription antitermination: the lambda paradigm
updated. Mol. Microbiol. 18:191–200
31. Friedman DI, Court DL. 2001. Bacteriophage lambda: alive and well and still doing its
thing. Curr. Opin. Microbiol. 4:201–7
32. Friedman DI, Gottesman M. 1983. Lytic mode of lambda development. See Ref. 38,
pp. 21–51
33. Furth ME, Dove WF, Meyer BJ. 1982. Specificity determinants for bacteriophage lambda
DNA replication. III. Activation of replication in lambda ric mutants by transcription
outside of ori. J. Mol. Biol. 154:65–83
34. Gottesman ME, Weisberg RA. 2004. Little lambda, who made thee? Microbiol. Mol. Biol.
Rev. 68:796–813
35. Greenblatt J, Mah TF, Legault P, Mogridge J, Li J, Kay LE. 1998. Structure and mech-
anism in transcriptional antitermination by the bacteriophage lambda N protein. Cold
Spring Harb. Symp. Quant. Biol. 63:327–36
36. Guarneros G. 1988. Retroregulation of bacteriophage lambda int gene expression. Curr.
Top. Microbiol. Immunol. 136:1–19
37. Hendrix RW, Lawrence JG, Hatfull GF, Casjens S. 2000. The origins and ongoing
evolution of viruses. Trends Microbiol. 8:504–8
38. Hendrix RW, Roberts JW, Stahl FW, Weisberg RA. 1983. Lambda II. Cold Spring Har-
bor, NY: Cold Spring Harbor Lab. Press
39. Herman C, Ogura T, Tomoyasu T, Hiraga S, Akiyama Y, et al. 1993. Cell growth
and lambda phage development controlled by the same essential Escherichia coli gene,
ftsH/hflB. Proc. Natl. Acad. Sci. USA 90:10861–65
40. Herman C, Thevenet D, D’Ari R, Bouloc P. 1997. The HflB protease of Escherichia coli
degrades its inhibitor lambda cIII. J. Bacteriol. 179:358–63
41. Herskowitz I, Hagen D. 1980. The lysis-lysogeny decision of phage lambda: explicit
programming and responsiveness. Annu. Rev. Genet. 14:399–445
42. Hoopes BC, McClure WR. 1985. A cII-dependent promoter is located within the Q gene
of bacteriophage lambda. Proc. Natl. Acad. Sci. USA 82:3134–38

www.annualreviews.org • Switches in Bacteriophage Lambda Development 425


ANRV260-GE39-18 ARI 15 October 2005 13:5

43. Hoyt MA, Knight DM, Das A, Miller HI, Echols H. 1982. Control of phage lambda
development by stability and synthesis of cII protein: role of the viral cIII and host hflA,
himA and himD genes. Cell 31:565–73
44. Jain D, Nickels BE, Sun L, Hochschild A, Darst SA. 2004. Structure of a ternary tran-
scription activation complex. Mol. Cell 13:45–53
45. Johnson A, Meyer BJ, Ptashne M. 1978. Mechanism of action of the cro protein of
bacteriophage lambda. Proc. Natl. Acad. Sci. USA 75:1783–87
46. Johnson AD, Poteete AR, Lauer G, Sauer RT, Ackers GK, Ptashne M. 1981. λ Repressor
and cro—components of an efficient molecular switch. Nature 294:217–23
47. Kaiser AD. 1957. Mutations in a temperate bacteriophage affecting its ability to lysogenize
Escherichia coli. Virology 3:42–61
48. Kedzierska B, Lee DJ, Wegrzyn G, Busby SJ, Thomas MS. 2004. Role of the RNA
polymerase alpha subunits in CII-dependent activation of the bacteriophage lambda pE
promoter: identification of important residues and positioning of the alpha C-terminal
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

domains. Nucleic Acids Res. 32:834–41


by University of California - San Diego on 03/29/11. For personal use only.

49. Kiehl TR, Mattheyses RM, Simmons MK. 2004. Hybrid simulation of cellular behavior.
Bioinformatics 20:316–22
50. Kihara A, Akiyama Y, Ito K. 1997. Host regulation of lysogenic decision in bacteriophage
lambda: transmembrane modulation of FtsH (HflB), the cII degrading protease, by HflKC
(HflA). Proc. Natl. Acad. Sci. USA 94:5544–49
51. Kobiler O, Koby S, Teff D, Court D, Oppenheim AB. 2002. The phage lambda CII
transcriptional activator carries a C-terminal domain signaling for rapid proteolysis. Proc.
Natl. Acad. Sci. USA 99:14964–69
52. Kobiler O, Rokney A, Friedman N, Court DL, Stavans J, Oppenheim AB. 2005. Quanti-
tative kinetic analysis of the bacteriophage lambda genetic network. Proc. Natl. Acad. Sci.
USA 102:4470–75
53. Kornitzer D, Altuvia S, Oppenheim AB. 1991. Genetic analysis of the cIII gene of bac-
teriophage HK022. J. Bacteriol. 173:810–15
54. Kornitzer D, Teff D, Altuvia S, Oppenheim AB. 1989. Genetic analysis of bacteriophage
lambda cIII gene: mRNA structural requirements for translation initiation. J. Bacteriol.
171:2563–72
55. Kourilsky P. 1973. Lysogenization by bacteriophage lambda. I. Multiple infection and
the lysogenic response. Mol. Gen. Genet. 122:183–95
56. Kourilsky P, Knapp A. 1974. Lysogenization by bacteriophage lambda. III. Multiplicity
dependent phenomena occurring upon infection by lambda. Biochimie 56:1517–23
57. Krinke L, Mahoney M, Wulff DL. 1991. The role of the OOP antisense RNA in coliphage
lambda development. Mol. Microbiol. 5:1265–72
58. Krinke L, Wulff DL. 1990. RNase III-dependent hydrolysis of lambda cII-O gene mRNA
mediated by lambda OOP antisense RNA. Genes Dev. 4:2223–33
59. Landy A. 1993. Mechanistic and structural complexity in the site-specific recombination
pathways of Int and FLP. Curr. Opin. Genet. Dev. 3:699–707
60. LeFevre KR, Cordes MH. 2003. Retroevolution of lambda Cro toward a stable monomer.
Proc. Natl. Acad. Sci. USA 100:2345–50
61. Levine M, Davidson EH. 2005. Gene regulatory networks for development. Proc. Natl.
Acad. Sci. USA 102:4936–42
62. Lieb M. 1953. The establishment of lysogenicity in Escherichia coli. J. Bacteriol. 65:642–51
63. Little JW. 2005. Threshold effects in gene regulation: When some is not enough. Proc.
Natl. Acad. Sci. USA 102:5310–11

426 Oppenheim et al.


ANRV260-GE39-18 ARI 15 October 2005 13:5

64. Little JW, Shepley DP, Wert DW. 1999. Robustness of a gene regulatory circuit. EMBO
J. 18:4299–307
65. Livny J, Friedman DI. 2004. Characterizing spontaneous induction of Stx encoding
phages using a selectable reporter system. Mol. Microbiol. 51:1691–704
66. Marr MT, Datwyler SA, Meares CF, Roberts JW. 2001. Restructuring of an RNA poly-
merase holoenzyme elongation complex by lambdoid phage Q proteins. Proc. Natl. Acad.
Sci. USA 98:8972–78
67. Marr MT, Roberts JW, Brown SE, Klee M, Gussin GN. 2004. Interactions among
CII protein, RNA polymerase and the lambda PRE promoter: contacts between RNA
polymerase and the -35 region of PRE are identical in the presence and absence of CII
protein. Nucleic Acids Res. 32:1083–90
68. Mason SW, Greenblatt J. 1991. Assembly of transcription elongation complexes con-
taining the N protein of phage lambda and the Escherichia coli elongation factors NusA,
NusB, NusG, and S10. Genes Dev. 5:1504–12
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

69. McAdams HH, Shapiro L. 1995. Circuit simulation of genetic networks. Science 269:650–
by University of California - San Diego on 03/29/11. For personal use only.

66
70. McMacken R, Mantei N, Butler B, Joyner A, Echols H. 1970. Effect of mutations in the
c2 and c3 genes of bacteriophage lambda on macromolecular synthesis in infected cells.
J. Mol. Biol. 49:639–55
71. Mensa-Wilmot K, Carroll K, McMacken R. 1989. Transcriptional activation of bacte-
riophage lambda DNA replication in vitro: regulatory role of histone-like protein HU of
Escherichia coli. EMBO J. 8:2393–402
72. Michalowski CB, Short MD, Little JW. 2004. Sequence tolerance of the phage lambda
PRM promoter: implications for evolution of gene regulatory circuitry. J. Bacteriol.
186:7988–99
72a. Mahajna J, Oppenheim AB, Rattray A, Gottesman M. 1986. Translation initiation of
bacteriophage lambda gene cII requires integration host factor. J. Bacteriol. 165:167–74
73. Nash HA. 1981. Integration and excision of bacteriophage lambda: the mechanism of
conservation site specific recombination. Annu. Rev. Genet. 15:143–67
74. Neubauer Z, Calef E. 1970. Immunity phase-shift in defective lysogens: non-mutational
hereditary change of early regulation of lambda prophage. J. Mol. Biol. 51:1–13
75. Nickels BE, Dove SL, Murakami KS, Darst SA, Hochschild A. 2002. Protein-protein
and protein-DNA interactions of sigma70 region 4 involved in transcription activation
by lambda cI. J. Mol. Biol. 324:17–34
76. Nodwell JR, Greenblatt J. 1991. The nut site of bacteriophage lambda is made of RNA
and is bound by transcription antitermination factors on the surface of RNA polymerase.
Genes Dev. 5:2141–51
77. Nodwell JR, Greenblatt J. 1993. Recognition of boxA antiterminator RNA by the E. coli
antitermination factors NusB and ribosomal protein S10. Cell 72:261–68
78. Obuchowski M, Giladi H, Koby S, Szalewska-Palasz A, Wegrzyn A, et al. 1997. Impaired
lysogenisation of the Escherichia coli rpoA341 mutant by bacteriophage lambda is due to
the inability of CII to act as a transcriptional activator. Mol. Gen. Genet. 254:304–11
79. Oppenheim A, Honigman A, Oppenheim AB. 1974. Interference with phage lambda cro
gene function by a colicin-tolerant Escherichia coli mutant. Virology 61:1–10
80. Patterson TA, Zhang Z, Baker T, Johnson LL, Friedman DI, Court DL. 1994. Bacte-
riophage lambda N-dependent transcription antitermination. Competition for an RNA
site may regulate antitermination. J. Mol. Biol. 236:217–28
80a. Peacock S, Weissbach H. 1985. IHF stimulation of lambda cII gene expression is inhibited
by the E coli NusA protein. Biochem. Biophys. Res. Commun. 127:1026–31

www.annualreviews.org • Switches in Bacteriophage Lambda Development 427


ANRV260-GE39-18 ARI 15 October 2005 13:5

81. Perutz M. 2003. I Wish I’d Made You Angry Earlier. Cold Spring Harbor, NY: Cold Spring
Harbor Lab. Press
81a. Jain D, Youngchang K, Maxwell KL, Beasley S, Rongguang Z, et al. 2005. Crystal struc-
ture of bacteriophage λ cII and its DNA complex. Mol. Cell 19:1–11
82. Ptashne M. 2004. Genetic Switch: Phage Lambda Revisited. Cold Spring Harbor, NY: Cold
Spring Harbor Lab. Press
83. Rattray A, Altuvia S, Mahajna G, Oppenheim AB, Gottesman M. 1984. Control of bacte-
riophage lambda CII activity by bacteriophage and host functions. J. Bacteriol. 159:238–
42
84. Reed MR, Shearwin KE, Pell LM, Egan JB. 1997. The dual role of Apl in prophage
induction of coliphage 186. Mol. Microbiol. 23:669–81
85. Rees WA, Weitzel SE, Yager TD, Das A, von Hippel PH. 1996. Bacteriophage lambda
N protein alone can induce transcription antitermination in vitro. Proc. Natl. Acad. Sci.
USA 93:342–46
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

86. Reichardt L, Kaiser AD. 1971. Control of lambda repressor synthesis. Proc. Natl. Acad.
by University of California - San Diego on 03/29/11. For personal use only.

Sci. USA 68:2185–89


87. Reinitz J, Vaisnys JR. 1990. Theoretical and experimental analysis of the phage lambda
genetic switch implies missing levels of co-operativity. J. Theor. Biol. 145:295–318
88. Retallack DM, Johnson LL, Ziegler SF, Strauch MA, Friedman DI. 1993. A single-base-
pair mutation changes the specificities of both a transcription activation protein and its
binding site. Proc. Natl. Acad. Sci. USA 90:9562–65
89. Santillan M, Mackey MC. 2004. Why the lysogenic state of phage lambda is so stable: a
mathematical modeling approach. Biophys. J. 86:75–84
90. Shea MA, Ackers GK. 1985. The OR control system of bacteriophage lambda. A physical-
chemical model for gene regulation. J. Mol. Biol. 181:211–30
91. Shotland Y, Koby S, Teff D, Mansur N, Oren DA, et al. 1997. Proteolysis of the phage
lambda CII regulatory protein by FtsH (HflB) of Escherichia coli. Mol. Microbiol. 24:1303–
10
92. Spiegelman WG, Reichardt LF, Yaniv M, Heinemann SF, Kaiser AD, Eisen H. 1972.
Bidirectional transcription and the regulation of phage lambda repressor synthesis. Proc.
Natl. Acad. Sci. USA 69:3156–60
93. Squires CL, Zaporojets D. 2000. Proteins shared by the transcription and translation
machines. Annu. Rev. Microbiol. 54:775–98
94. Svenningsen SL, Costantino N, Court DL, Adhya S. 2005. On the role of Cro in λ
prophage induction. Proc. Natl. Acad. Sci. USA. In press
95. Takeda Y, Folkmanis A, Echols H. 1977. Cro regulatory protein specified by bacterio-
phage lambda. Structure, DNA-binding, and repression of RNA synthesis. J. Biol. Chem.
252:6177–83
96. Teff D, Koby S, Shotland Y, Ogura T, Oppenheim AB. 2000. A colicin-tolerant Escherichia
coli mutant that confers hfl phenotype carries two mutations in the region coding for the
C-terminal domain of FtsH (HflB). FEMS Microbiol. Lett. 183:115–17
97. Thomas R. 1971. Control circuits. In The Bacteriophage Lambda, ed. AD Hershey,
pp. 211–20. Cold Spring Harbor, NY: Cold Spring Harbor Lab. Press
98. Tian T, Burrage K. 2004. Bistability and switching in the lysis/lysogeny genetic regulatory
network of bacteriophage lambda. J. Theor. Biol. 227:229–37
99. Tomoyasu T, Yamanaka K, Murata K, Suzaki T, Bouloc P, et al. 1993. Topology
and subcellular localization of FtsH protein in Escherichia coli. J. Bacteriol. 175:1352–
57

428 Oppenheim et al.


ANRV260-GE39-18 ARI 15 October 2005 13:5

100. Vohradsky J. 2001. Neural model of the genetic network. J. Biol. Chem. 276:36168–73
101. Weisberg RA, Gottesmann ME, Hendrix RW, Little JW. 1999. Family values in the age
of genomics: comparative analyses of temperate bacteriophage HK022. Annu. Rev. Genet.
33:565–602
102. Whalen WA, Das A. 1990. Action of an RNA site at a distance: role of the nut genetic signal
in transcription antitermination by phage-lambda N gene product. New Biol. 2:975–91
103. Wilson HR, Kameyama L, Zhou JG, Guarneros G, Court DL. 1997. Translational re-
pression by a transcriptional elongation factor. Genes Dev. 11:2204–13
104. Wilson HR, Yu D, Peters HK 3rd, Zhou JG, Court DL. 2002. The global regulator
RNase III modulates translation repression by the transcription elongation factor N.
EMBO J. 21:4154–61
105. Wilson HR, Zhou JG, Yu D, Court DL. 2004. Translation repression by an RNA poly-
merase elongation complex. Mol. Microbiol. 53:821–28
106. Wold MS, Mallory JB, Roberts JD, LeBowitz JH, McMacken R. 1982. Initiation of
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

bacteriophage lambda DNA replication in vitro with purified lambda replication proteins.
by University of California - San Diego on 03/29/11. For personal use only.

Proc. Natl. Acad. Sci. USA 79:6176–80


107. Wommack KE, Colwell RR. 2000. Virioplankton: Viruses in aquatic ecosystems. Micro-
biol. Mol. Biol. Rev. 64:69–114
108. Wrobel B, Herman-Antosiewicz A, Szalewska-Palasz S, Wegrzyn G. 1998. Polyadenyla-
tion of oop RNA in the regulation of bacteriophage lambda development. Gene 212:57–65
109. Yang XJ, Hart CM, Grayhack EJ, Roberts JW. 1987. Transcription antitermination by
phage-lambda gene-Q protein requires a DNA segment spanning the RNA start site.
Genes Dev. 1:217–26
110. Zhu XM, Yin L, Hood L, Ao P. 2004. Calculating biological behaviors of epigenetic
states in the phage lambda life cycle. Funct. Integr. Genomics 4:188–95

www.annualreviews.org • Switches in Bacteriophage Lambda Development 429


Contents ARI 20 October 2005 19:18

Annual Review of

Contents Genetics

Volume 39, 2005

John Maynard Smith


Richard E. Michod p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 1
The Genetics of Hearing and Balance in Zebrafish
Teresa Nicolson p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 9
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org
by University of California - San Diego on 03/29/11. For personal use only.

Immunoglobulin Gene Diversification


Nancy Maizels p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p23
Complexity in Regulation of Tryptophan Biosynthesis in
Bacillus subtilis
Paul Gollnick, Paul Babitzke, Alfred Antson, and Charles Yanofsky p p p p p p p p p p p p p p p p p p p p p p p47
Cell-Cycle Control of Gene Expression in Budding and Fission Yeast
Jürg Bähler p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p69
Comparative Developmental Genetics and the Evolution of Arthropod
Body Plans
David R. Angelini and Thomas C. Kaufman p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p95
Concerted and Birth-and-Death Evolution of Multigene Families
Masatoshi Nei and Alejandro P. Rooney p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 121
Drosophila as a Model for Human Neurodegenerative Disease
Julide Bilen and Nancy M. Bonini p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 153
Molecular Mechanisms of Germline Stem Cell Regulation
Marco D. Wong, Zhigang Jin, and Ting Xie p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 173
Molecular Signatures of Natural Selection
Rasmus Nielsen p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 197
T-Box Genes in Vertebrate Development
L.A. Naiche, Zachary Harrelson, Robert G. Kelly, and Virginia E. Papaioannou p p p p p p 219
Connecting Mammalian Genome with Phenome by ENU Mouse
Mutagenesis: Gene Combinations Specifying the Immune System
Peter Papathanasiou and Christopher C. Goodnow p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 241
Evolutionary Genetics of Reproductive Behavior in Drosophila:
Connecting the Dots
Patrick M. O’Grady and Therese Anne Markow p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 263
v
Contents ARI 20 October 2005 19:18

Sex Determination in the Teleost Medaka, Oryzias latipes


Masura Matsuda p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 293
Orthologs, Paralogs, and Evolutionary Genomics
Eugene V. Koonin p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 309
The Moss Physcomitrella patens
David Cove p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 339
A Mitochondrial Paradigm of Metabolic and Degenerative Diseases,
Aging, and Cancer: A Dawn for Evolutionary Medicine
Douglas C. Wallace p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 359
Switches in Bacteriophage Lambda Development
Amos B. Oppenheim, Oren Kobiler, Joel Stavans, Donald L. Court,
Annu. Rev. Genet. 2005.39:409-429. Downloaded from www.annualreviews.org

and Sankar Adhya p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 409


by University of California - San Diego on 03/29/11. For personal use only.

Nonhomologous End Joining in Yeast


James M. Daley, Phillip L. Palmbos, Dongliang Wu, and Thomas E. Wilson p p p p p p p p p p 431
Plasmid Segregation Mechanisms
Gitte Ebersbach and Kenn Gerdes p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 453
Use of the Zebrafish System to Study Primitive and Definitive
Hematopoiesis
Jill L.O. de Jong and Leonard I. Zon p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 481
Mitochondrial Morphology and Dynamics in Yeast and Multicellular
Eukaryotes
Koji Okamoto and Janet M. Shaw p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 503
RNA-Guided DNA Deletion in Tetrahymena: An RNAi-Based
Mechanism for Programmed Genome Rearrangements
Meng-Chao Yao and Ju-Lan Chao p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 537
Molecular Genetics of Axis Formation in Zebrafish
Alexander F. Schier and William S. Talbot p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 561
Chromatin Remodeling in Dosage Compensation
John C. Lucchesi, William G. Kelly, and Barbara Panning p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 615

INDEXES

Subject Index p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 653

ERRATA

An online log of corrections to Annual Review of Genetics


chapters may be found at http://genet.annualreviews.org/errata.shtml

vi Contents
Modern Microbial Genetics, Second Edition. Edited by Uldis N. Streips, Ronald E. Yasbin
Copyright # 2002 Wiley-Liss, Inc.
ISBNs: 0-471-38665-0 (Hardback); 0-471-22197-X (Electronic)

21
Transduction in Gram-Negative Bacteria
GEORGE M. WEINSTOCK
Department of Biochemistry and Molecular Biology, University of Texas Medical School,
Houston, Texas 77225

I. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 561
II. Generalized Transduction . . . . . . . . . . . . . . . . . . . . . . . . 562
A. Events in the Donor: Bacteriophage P22 . . . . . . . . 563
1. P22 DNA Metabolism . . . . . . . . . . . . . . . . . . . . . 564
2. Formation of P22 Transducing Particles . . . . . . 565
B. Formation of Generalized Transducing Particles
by Other Phages. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 566
1. Phage P1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 566
2. Phage T4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 567
3. Phage l. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 567
4. Phage Mu. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 567
C. Events in the Recipient . . . . . . . . . . . . . . . . . . . . . . . 568
1. Homologous Recombination With the
Recipient's Chromosome . . . . . . . . . . . . . . . . . . . 568
D. Measuring Transduction. . . . . . . . . . . . . . . . . . . . . . 568
1. Cotransduction of Markers . . . . . . . . . . . . . . . . . 570
2. Marker Effects. . . . . . . . . . . . . . . . . . . . . . . . . . . . 571
3. Abortive Transduction . . . . . . . . . . . . . . . . . . . . . 572
4. Transduction of Plasmids . . . . . . . . . . . . . . . . . . 572
E. Uses for Generalized Transduction . . . . . . . . . . . . . 573
III. Specialized Transduction. . . . . . . . . . . . . . . . . . . . . . . . . 573
A. Bacteriophage l DNA Metabolism. . . . . . . . . . . . . 573
B. Events in the Recipient . . . . . . . . . . . . . . . . . . . . . . . 575
C. Uses for Specialized Transduction. . . . . . . . . . . . . . 576
APPENDIX: Bacteriophage l . . . . . . . . . . . . . . . . . . . . . . . . 577

I. INTRODUCTION carry parts of bacterial or plasmid chromo-


somes from one bacterial host to another. For
Transduction is the mode of genetic exchange
some phages this DNA is joined to the viral
between cells that is mediated by viruses. The
chromosome in the particle (as in specialized
term transduction refers to the transfer of cel-
transductionbyphagel),whileforothertrans-
lular DNA or RNA from one cell to another by
ducing phages only DNA of host origin is pre-
infection with a viral vector. Virus particles
sent in the particle (as in generalized
that carry and introduce cellular nucleic acids
on infection are called transducing particles.
Bacteriophages are transducing viruses if they
562 WEINSTOCK

transduction by phages P1 or P22). The forma- covery was made possible because fortuitously
tion of transducing particles is a part of the one of the Salmonella strains that Zinder and
normal DNA metabolism of viral infection. Lederberg had used in the mixing experiment
In addition, when foreign DNA is introduced was a lysogen for P22. During their experi-
into a virus chromosome by recombinant ments the P22 prophage had spontaneously
DNA techniques, the resulting phages that induced, forming particles that infected the
carry the cloned foreign DNA are transducing second Salmonella strain. The infection of the
particles. Finally, RNA viruses, such as retro- second strain then produced particles that
viruses, that carry host RNA and introduce it carried genes of the second host. When these
into a recipient cell are transducing viruses. particlesinfectedtheoriginalauxotrophicbac-
The process of transduction is divided into terium, they introduced the wild-type gene,
two types. In generalized transduction, the which could recombine and replace the defect-
virus can introduce any region from the donor ivesegmentofthechromosome,formingapro-
chromosome into a recipient. In specialized totrophic transductant bacterium.
transduction, the virus always carries the Subsequently this process of bacterio-
same segment into the recipient. A phage that phage-mediated genetic transduction was
randomly packages host DNA into particles is shown to occur in many other Gram-negative
a generalized transducing phage. A phage that bacteria, including Escherichia coli, Myxo-
has had a particular gene stably introduced bacteria, Rhizobium, Caulobacter, and Pseu-
into its chromosome is a specialized transdu- domonas. In E. coli, for example, transduction
cing phage. For detailed reviews of these pro- was shown to occur with bacteriophage P1. In
cesses, see Margolin (1987) and Weisberg addition it was possible to transfer any host
(1987); for a summary of the early literature, gene from one bacterium to another with
see Low and Porter (1978) (also see Hendrix, these phages, and hence this process came to
this volume). be known as generalized transduction. That
this was due to physical transfer of DNA
from the donor bacterium to the recipient
II. GENERALIZED
was demonstrated for both P22 (Ebel-Tsipis
TRANSDUCTION et al., 1972) and P1 (Ikeda and Tomizawa,
In 1952 Zinder and Lederberg first reported 1965a) transduction. To show this (Fig. 1),
generalized transduction in Salmonella tym- the donor bacteria were grown in a medium
phimurium. They were able to show that containing heavy isotopes of nitrogen and
genetic exchange occurred in Salmonella hydrogen (15 N and 2 H) to make the donor
by mixing different auxotrophic mutants to- DNA more dense. Then the bacteria were
gether and isolating prototrophic recombin- shifted to a normal medium (14 N and 1 H)
ants. To differentiate this mechanism of and infected with the phage. During growth,
genetic exchange from conjugation, they the newly synthesized phage DNA was light,
showed that transfer of the genetic material while the preexisting bacterial DNA was
did not require physical contact between heavy. It was then shown that the phage pre-
the donor and recipient bacteria. The agent pared in this way contained heavy DNA; that
responsible for transfer was able to pass is, they had packaged the donor bacterial
through the pores of a filter, pores that were DNA into particles, and when they were
too small to allow bacteria through. Moreover used to infect a recipient bacterium that had
the genetic exchange was not due to transform- light DNA, some of the heavy DNA became
ation (see Streips, this volume), since the pro- incorporated into the recipient chromosome.
cess was resistant to DNase treatment. It was These experiments illustrate the overall pro-
eventuallyshownthattheagentthatcarriedthe cess of generalized transduction. They show
genesfromthedonortotherecipientbacterium that there are two parts to the mechanism:
wasthetemperatebacteriophageP22.Thisdis- the packaging of donor DNA into a phage
GENETIC TRANSDUCTION 563

Fig. 1. Physical transfer of DNA in generalized transduction. An example of generalized transduction by


phage P22 is shown. The donor bacteria are grown in a medium containing heavy isotopes of nitrogen and
hydrogen so that their DNA has a heavier density than normal (the heavy DNA is represented by the thick
circular line in the cell at the upper left). Following P22 infection of this cell, some heavy chromosomal DNA
is packaged into phage particles. These can then be used to infect a cell grown in medium with light isotopes
of nitrogen and hydrogen, and thus containing DNA with a lighter density than the donor's. When heavy
donor DNA is introduced, it can recombine and replace some light DNA in the recipient's chromosome.
When the chromosome is broken into small pieces, the heavy DNA can be detected by equilibrium gradient
centrifugation because it bands at a different density than the bulk of the light DNA.

particle and the stable introduction of this A. Events in the Donor: Bacteriophage
packaged DNA into the recipient cell, usually P22
through genetic recombination with the re- A well-studied example of generalized trans-
cipient chromosome (Fig. 2). The ability of a duction is that of the Salmonella phage P22.
phage to perform generalized transduction This is a useful paradigm for DNA
thus depends on the mechanism of packaging metabolism leading to generalized transduc-
DNA into phage particles. If this mechanism tion, since it is not unique and is representa-
allows host as well as phage DNA to be en- tive of a number of other phages (see
capsidated, generalized transduction invari- Guttman and Kutter, this volume, for phage
ably occurs. As we will see, there are T4). The DNA metabolism of phage P22 has
numerous types of DNA metabolism that been reviewed by Susskind and Botstein
lead to generalized transduction. In each (1978).
case the capability for generalized transduc- The phage DNA in a P22 particle is ter-
tion is a result of the mode of packaging minally redundant and circularly permuted
phage DNA. (Fig. 3). Each phage chromosome has the
564 WEINSTOCK

circularization occurs by homologous re-


combination between the repetitious ends of
the chromosome. This is the significance
of the terminal redundancy of the chromo-
someÐit provides the substrate for the cir-
cularization reaction. The circular molecule
replicates and ultimately produces a conca-
temer, a long DNA molecule containing
multiple copies of the genome. This probably
arises from rolling circle replication, an effi-
cient process where a single replication fork
continually circles a circular chromosome
synthesizing a long concatemeric tail (see
Fig. 3). Packaging of this DNA initiates at

Fig. 2. Overview of generalized transduction. The


scheme shown applies to phages like P22 and P1.
During normal productive infection of bacteria two
kinds of particles are produced: those with a viral
chromosome and those containing a fragment of the
host chromosome. When the phage with a viral
chromosome infects a cell, another round of pro-
ductive infection occurs. However, when the particle
with the host DNA injects this DNA into a cell,
the fragment can recombine with the recipient's
chromosome, resulting in a transductant.

leftmost sequence of its chromosome


repeated at the right end (terminally redun- Fig. 3. Scheme of phage P22 DNA metabolism. At
dant). However, this repetitious segment the top is shown the linear DNA molecule carried by
(about 2% of the chromosome) is not unique the phage particle. This chromosome is terminally
and differs for each chromosome (circularly redundant, containing a copy of the ``a'' region of the
permuted). This chromosome structure is a genome at each end. The two copies of the ``a'' region
can recombine to circularize the chromosome. Then
result of the mechanism of packaging DNA a concatemer is formed by rolling circle replication.
employed by P22, the headful packaging This concatemeric DNA is packaged into phage par-
mechanism (Fig. 3). ticles by sequential cuts starting at a pac site. Each
packaging event measures out a ``headful'' of DNA,
1. P22 DNA metabolism which is slightly longer than the size of the genome.
As a result a series of chromosomes is produced.
When P22 infects a cell, its DNA must circu- Each chromosome is terminally redundant, but the
larize for the infection to be productive and redundant sequence is different for each one. Thus
the chromosome to replicate. This obligatory the chromosomes are said to be circularly permuted.
GENETIC TRANSDUCTION 565

a specific region in the genome (pac) and


proceeds processively along the concatemer
from this site. The first packaging cut, by
a phage-encoded enzyme, is site specific, at
pac, and a defined length of DNA is encap-
sidated. When this length (a ``head-ful'') is
packaged, the second cut is made and a
phage head containing DNA is formed.
This second cut also defines the start of
packaging for the next phage head to be
formed. The same headful length of DNA
is encapsulated starting at the second cut,
and when this length is reached a third cut
is made. The third cut plays the same role as
the second. It terminates packaging of the
second headful and starts the packaging Fig. 4. Packaging of host DNA by the phage P22
into the next head. This process continues. headful packaging mechanism. A: A wild-type bacter-
Thus, although DNA packaging starts at a ial chromosome contains a site, pac, that is recog-
defined sequence, the subsequent cuts are nized by the phage DNA packaging apparatus. The
not sequence specific. Rather, it is the same bacterial chromosome is packaged into phage par-
ticles starting at pac and proceeding sequentially in
length of DNA that is packaged each time. one direction. Each packaging event cuts a headful of
For phage P22, the size of the genome is DNA and each of the resulting transducing frag-
slightly less than the length of the headful. ments carries a characteristic set of genes. Genes
Thus, each time a head is formed, the en- on the same fragment are cotransducible because
capsidated DNA contains a short terminal they can be introduced into a cell together. B: The
packaging of a bacterial chromosome that is deleted
repeat. Since each packaging event starts for gene E is shown. The pac site is still recognized
at a different sequence, the repeats are dif- and the overall process of forming transducing par-
ferent, producing permuted chromosomes. ticles is the same as in A. However, because of the
Headful packaging thus provides the mech- deletion, the cotransduction of genes is altered. This
anism for generating the essential terminal is because each packaging event measures the same
amount of DNA and the deletion has changed the
repeats. distance of distal genes from the pac site.
2. Formation of P22 transducing particles
During P22 infection the host DNA is not
degraded (in contrast to infections with viru- initiates and proceeds processively, generat-
lent phages like T4; see Guttman and Kutter, ing a series of phage particles that carry
this volume), and thus is potentially also a different parts of the host chromosome.
substrate for DNA packaging. As just de- These transducing particles carry DNA frag-
scribed, the characteristics of DNA pack- ments that are all the same size but obviously
aging are that a concatemeric molecule is do not have the terminal repetition that the
encapsidated in processive headfuls starting phage chromosomes have, since the genome
from a unique site. Exactly the same process size of the host is much greater than the
operating on the host DNA generates trans- headful size that is packaged. For P22, al-
ducing particles (Fig. 4). When a sequence though there are roughly equal amounts of
similar to the pac sequence occurs in the host phage and host DNA in the cell during
chromosome, the phage packaging appar- infection, only a few percent of the host
atus uses this analogous pac site and the chromosome is packaged and only a few
host chromosome can be packaged. When a percent of the particles are transducing par-
pac site occurs in the host DNA, packaging ticles. What limits formation of transducing
566 WEINSTOCK

particles is the number of pac sites in the host packaging initiates at a different, more
DNA, as well as the fact that these chromo- common site (Casjens et al., 1987). With
somal sites are probably not identical in se- respect to packaging of the phage chromo-
quence to the phage pac site and are thus some, it is observed that in HT mutants there
inefficiently used. It has been estimated is a different distribution of cut sites. With
(Chelala and Margolin, 1974) that there are respect to generalized transduction, in the
10 to 15 of these pac-like sites in the Salmon- HT mutants other, more numerous sites in
ella chromosome. Other studies (reviewed in the bacterial chromosome can be used to
Margolin, 1987) indicate that at least 5±10% initiate packaging and hence the sites are no
of the bacterial chromosome is packaged in longer as limiting a factor in the initiation of
the sequential headfuls from each pac site. packaging. This means that the host DNA
However, the efficiency of packaging de- competes more effectively with phage DNA
clines the more distant a gene is from a pac for the packaging apparatus, and there is a
site, and this contributes to the wide range in higher proportion (up to 50%) of phage par-
transduction frequencies seen for different ticles that carry host DNA, resulting in a
genes transduced with P22. This variation higher frequency of transduction. In add-
in frequency can be two or three orders of ition there is less variation in the transduc-
magnitude between different genes. tion frequency of different genes. Because of
One experimental approach (Chelala and these properties the HT mutants are ex-
Margolin, 1974) that provided support for tremely useful tools when transduction is
this view of transducing particle formation needed for genetic manipulations.
comes from the study of the effects of dele-
tions in the bacterial chromosome on trans- B. Formation of Generalized
duction (Fig. 4). One effect of deletions is on Transducing Particles by Other Phages
the cotransduction of genes, the frequency at Phage P22 is a useful model, both to illus-
which two nearby genes are transduced to- trate a common mode of DNA metabolism
gether on the same fragment. Some deletions as well as its consequences for transduction.
were found to alter this frequency, presum- However, among the phages that perform
ably by altering the register of subsequent generalized transduction, there are both
packaging events. Effects on transduction minor variations on this theme as well as
have also been observed for insertions; these completely different modes of transduction.
are due to an analogous mechanism. The Some examples are given below.
studies imply that host DNA packaging ini-
tiates from a limited number of sites and 1. Phage P1
occurs processively with a characteristic The E. coli phage P1 is in many ways as well
register for each region. studied as P22 (see Sternberg and Hoess,
Another important approach to under- 1983, for a review). Phage P1 is noteworthy
standing this mechanism of generalized because it is the major generalized transdu-
transduction was the isolation and analysis cing phage used in E. coli and can infect a
of HT mutants of P22: mutants that perform broad range of hosts, making it an important
generalized transduction at a higher fre- tool for genetic manipulation. In general, P1
quency than a wild-type phage (Schmieger, is similar to P22 in using a processive headful
1972). HT mutants, which can increase the packaging process initiating at a specific site
frequency of transduction of some genes by to produce terminally redundant, circularly
as much as 10,000-fold, have an alteration in permuted phage DNA. The P1 pac region
the phage protein involved in recognizing the has been analyzed and is complex, compris-
pac site (Raj et al., 1974). As a consequence ing a number of short sequence elements in a
of this mutation, the HT mutants show an 161 bp region (Sternberg and Coulby, 1987).
altered specificity in making the first cut: The size of the P1 headful is about twice that
GENETIC TRANSDUCTION 567

of P22, thus larger segments of the bacterial some for packaging to occur. Cutting at cos
chromosome can be transduced. There is less produces a double-stranded break that is not
variation in transduction frequency between blunt ended; a 12 nucleotide single-strand
different genes with P1 than seen with P22. end is produced that is necessary for the
This could result from the larger headful as chromosome to circularize in the next infec-
well as there being more packaging sites tion. This DNA processing system, although
in the bacterial chromosome. These results suited for processing of the phage's DNA, is
have also been interpreted to mean that ini- apparently only loosely related to the mech-
tiation of packaging of the bacterial chromo- anism of formation of generalized transdu-
some by phage P1 does not use pac sites cing particles. A study of the formation
like those found in the phage chromosome of transducing particles suggests that l
(Sternberg, 1986). packages host DNA without recognition
of specific cos sites (Sternberg, 1986). More-
2. Phage T4 over formation of transducing particles
As detailed by Guttman and Kutter (this is not seen until 60 to 90 minutes after in-
volume), phage T4 is not usually thought of fection and is inhibited by the phage
as a generalized transducing phage. T4 is a exonuclease synthesized during infection.
virulent phage that degrades the host DNA Because of these properties, l transducing
during infection and normally packages only particles are difficult to detect in a wild-
phage DNA by a headful packaging mech- type infection, where cell lysis occurs after
anism. This degradation is a result of an- about 60 minutes. Lysis defective mutants
other feature of T4 DNA metabolism. T4 (also mutant for the exonuclease) must be
DNA is modified and contains glycosylated used for maximal transducing particle for-
hydroxymethylcytosine, a modification that mation.
results from phage-encoded enzymes. The
degradation is due to a phage-encoded 4. Phage Mu
nuclease that cuts DNA at unmodified cyto- Mu has quite a different DNA metabolism
sines, generating substrates for other DNases from the phages discussed so far (see Whittle
in the cell. A multiple mutant phage was and Salyers, this volume). Mu is a transpos-
constructed (Wilson et al., 1979) with muta- able element that replicates by copying its
tions to prevent degradation of host DNA. genome and integrating this copy at random
This phage can perform generalized trans- sites in bacterial DNA (replicative transpos-
duction, presumably packaging host DNA ition; Fig. 5). During infection the cell thus
as well as phage DNA because the mechan- accumulates a number of different insertions
ism to distinguish these two has been re- of the Mu chromosome. Packaging of these
moved. Mu chromosomes is by a headful mechan-
ism. In this case the pac site is at the c end
3. Phage l and one cut is made in bacterial DNA about
Although l is not usually thought of as a 100 nucleotides outside of this end while the
generalized transducing phage, it can pack- other cut is a fixed length in the direction of
age host DNA (Sternberg, 1986). This phage the S end, from 500 to 3200 nucleotides out-
does not use a headful packaging mechan- side of this end for the wild-type phage.
ism. During infection, replication of l DNA Hence the cuts are made in the bacterial
generates concatemers from a rolling circle, DNA that flanks the integrated Mu genome.
like the phages described above. However This genome structure is important for the
the packaging mechanism is site specific. A initial transposition event in subsequent
particular sequence, cos, is recognized and infections. Genetic studies (Howe, 1973)
the DNA is cut (discussed below; Fig. 9). showed that Mu is capable of generalized
Two cos sites must be present in the chromo- transduction. Presumably pac sites in the
568 WEINSTOCK

into a recipient cell, it can become stably


integrated into the recipient cell's chromo-
some by homologous recombination, or it
can remain unintegrated in the cytoplasm.
In this latter unintegrated case the DNA
will eventually be lost if it cannot replicate
in the cytoplasm (abortive transduction),
whereas it will be maintained as an extra-
chromosomal element if it can replicate as a
plasmid.

1. Homologous recombination with the


recipient's chromosome
Most of what is known about integration of
transducing fragments by recombination
comes from studies of phages P22 and P1.
In the case of P1 for example, linear double-
stranded DNA molecules are injected into
the recipient where they may assume a circu-
lar conformation due to the presence of a
bound protein. Studies with isotopically
labeled DNA showed that there is a physical
Fig. 5. Phage Mu DNA metabolism. Shown at the replacement of the recipient's DNA by the
top is the linear phage Mu chromosome as it is carried transduced fragment, as described earlier. In
in the phage particle. The central (wavy) portion the case of transduction by both P1 and P22,
represents phage genes while the terminal (straight)
portions flanking the phage c and s regions are bac-
it was determined that this is a double-strand
terial DNA segments. Upon infection, the Mu seg- replacement. That is, a double-stranded seg-
ment of the chromosome transposes into the circular ment of the recipient's chromosome is re-
bacterial chromosome. This integrated genome then placed by the homologous double-stranded
replicates and transposes to additional sites in the segment from the transducing fragment.
chromosome. Finally, the integrated genomes are
packaged by a process that involves cutting in the
Such an event requires two crossovers, one
bacterial DNA that flanks the integrated Mu at each end of the replaced segment (Fig. 6).
genomes. This produces chromosomes that contain It has been estimated that only about 5% of
different regions of the bacterial genome. the DNA introduced as transducing frag-
ments ends up being incorporated into the
bacterial chromosome. This is in part be-
host chromosome are also recognized, lead- cause some fragments do not recombine at
ing to the formation of transducing particles. all and remain in the cytoplasm. In addition,
for those fragments that do recombine, the
C. Events in the Recipient
entire transducing fragment that is intro-
As stated earlier, generalized transduction duced into the recipient need not integrate
can be thought of as a two-stage process. into the recipient chromosome. The portion
The varied ways in which the first stage, that is integrated is determined by where the
packaging of host DNA into phage particles, two crossovers occur.
can occur has been described. In this section
the second stage of transduction is de- D. Measuring Transduction
scribedÐthe fate of the host DNA after it In discussing the various fates of the DNA
has been introduced into a recipient cell. in the recipient, it is also useful to consider
When the packaged host DNA is injected the practical problems in assaying for
GENETIC TRANSDUCTION 569

mixed with the recipient cells and, after the


phages have adsorbed to the bacteria, the
mixture is plated on a medium that selects
for metE ‡ cells. Most of the cells fail to grow
on this medium because they remain metE ,
but a few cells can grow and these form
colonies. These are the cells that have re-
ceived a transducing fragment carrying the
metE ‡ gene and this wild-type gene has
become integrated into the recipient chromo-
some by recombination, replacing the metE
gene and making the cell metE ‡ (Fig. 6). The
problem is to quantitate the frequency of
transducing the metE ‡ gene.
One factor that complicates this quantita-
tion is the multiplicity of infection (MOI), the
ratio of phage particles to bacterial cells
(Fig. 7). At low multiplicities (MOI  1)
Fig. 6. Recombination in the recipient: possible most bacteria receive no phage particles and
outcomes of a three-factor transductional cross. those that are infected receive only a single
Shown at the top is a recipient chromosome (thin particle. Very few cells receive more than one
lines) carrying mutations in the ilv, cya, and metE particle and thus the number of metE ‡ trans-
genes and a donor transducing DNA fragment (thick
ductants observed is simply related to the
lines) that is wild-type at these three loci. In order to
integrate donor DNA into the recipient chromo- number of particles carrying metE ‡ DNA.
some, two crossovers must occur, and these can The number of transductants will be less
be in any of the four regions defined by the three than the number of particles carrying metE ‡
genes. At the bottom is shown the genotypes gene because, as mentioned above, not all of
resulting from crossovers in different regions. The
donor DNA (thick line) substituted for that of the
recipient (thin line) is shown for each case.

transduction. Imagine an experiment to


measure the transduction of a specific gene,
metE, that encodes the enzyme homocysteine
methylase, required for methionine biosyn-
thesis. The donor is metE ‡ and the recipient
is metE . After growth of a phage like P1 or
P22 on the donor strain, the resulting phage
lysate contains two kinds of particles (Fig.
2). The majority of particles carry phage
Fig. 7. Effect of multiplicity of infection (MOI) on
DNA and will cause phage production and transduction frequency. This curve illustrates the fact
cell death upon subsequent infection. The that at high MOI, where all cells are infected by a
remaining particles carry host DNA from particle containing phage DNA, there will be killing
all regions of the bacterial chromosome, so of cells receiving transducing DNA, leading to a
only a small fraction of these carry the decrease in frequency. Note that this effect will not
be as great if the phage can form lysogens. In that
metE ‡ gene. For P22, only a few percent of case there will be some lysogen formation at high
the particles carry host DNA, and of these, MOI and correspondingly less killing. However, mu-
at most only a few percent carry metE. In the tations are often introduced into transducing phages
transduction experiment, the phage lysate is to prevent lysogenization.
570 WEINSTOCK

these fragments recombine with the bacterial DNA-containing particles. The alternative
chromosome and those that do will some- method is to measure the frequency of trans-
times not integrate the metE region of the duction of several different markers by
fragment. At high multiplicities (MOI  1) always using the same volume of lysate (i.e.,
all cells become infected and most cells re- the same MOI) in the transductions and ig-
ceive more than one particle. In this case, noring the titer of phage DNA-containing
when a cell is infected by a particle that particles. In this case the ratio of transduc-
carries a metE ‡ gene, it is simultaneously tants for each marker can be normalized to
infected by other particles, and these will that for a common marker, allowing them to
carry phage DNA since this is the major be compared.
class of particles present. The particles intro-
ducing phage DNA will usually cause the cell 1. Cotransduction of markers
to produce more phage and lyse, thus One of the most important uses for general-
making it impossible for the cell to become ized transduction is in measuring how far
a transductant. At high MOI the only cells apart markers are or in determining the
that can become transductants are those order of three or more markers in the
that receive a transducing fragment and chromosome. An important concept for this
either do not become infected by particles analysis is that of contransduction, the abil-
carrying phage DNA, or if they do, the ity of two markers to be simultaneously inte-
phage infection leads to lysogeny rather grated into the recipient's chromosome on
than lytic growth. As a result of this, fewer the same transducting fragment (Figs. 4, 6).
transductants may be observed when large Since phages like P1 and P22 package a
amounts of lysate are used for transduction discrete length of DNA in their particles,
(high MOI) than when lower amounts of for two markers to be contransduced at all
lysate are used (low MOI). The optimum requires that they not be farther apart in
amount of lysate is somewhere near an the bacterial chromosome than the headful
MOI of 1, where the largest amount of length of DNA that the transducing phage
lysate (and transducing particles) has been will package. Furthermore the closer to-
added that does not cause significant loss of gether two markers are located, the higher
transductants by killing due to multiple in- is the probability that they can be integrated
fection with particles containing phage into the recipient chromosome together. This
DNA. probability is manifested operationally as the
In measuring transduction frequencies, contransduction frequency. In the example
two approaches can be used. In one method, of Figure 6, the donor strain is ilv‡ cya‡
after the growth of the phage on the donor metE ‡ , while the recipient is ilv cya
strain, the resulting lysate is titered for par- metE . When metE ‡ transductants are
ticles containing phage DNA by measuring selected, more of these will also be cya‡
the number of plaque-forming units (PFUs) (cotransduction of cya and metE) than will
on a suitable indicator bacterium. Next the be ilv‡ (cotransduction of ilv and metE) be-
phage lysate is titered for transducting par- cause cya is closer to metE than ilv is. More-
ticles by measuring the number of trans- over the metE ‡ transductants that are also
ductants in the volume of lysate added to ilv‡ will almost always be cya‡ because this
recipient cells. The ratio of transductants to marker is in the middle (only rare double
PFUs is then calculated, using the titer of recombinants will be ilv‡ cya metE ‡ ). In
transductants determined in the low MOI contrast, of the metE ‡ transductants that
portion of the titration. This method allows are also cya‡ , some will be ilv‡ and some
the frequency of transduction of two differ- will be ilv . These results would suggest the
ent markers to be compared, by normalizing gene order ilv-cya-metE. This mapping tech-
them both to the total number of phage nique is very similar to the three-factor cross
GENETIC TRANSDUCTION 571

procedure using bacterial transformation (see source of variability is due to differences


Streips, this volume). in the efficiency of recombination of trans-
There are actually two factors that contrib- ducing fragments from different regions of
ute to the frequency of cotransduction of two the chromosome. This appears to be the pri-
markers. One is the probability that they will mary cause of variation in P1 transduction.
reside on the same transducting fragment Treatments that stimulate recombination,
formed in the donor. The closer two markers such as UV irradiation (see chapters by Yas-
are to each other, the greater the chance that bin and Levene and Huffman, this volume),
they will be packaged together on a single reduce the variability of P1 transduction fre-
fragment. In addition, since the two cross- quencies.
overs resulting in integration in the recipient As a result of these factors, quantitating
can occur anywhere along the transducing transduction frequencies can be complex. An
fragment, the closer together the markers example of this is the asymmetry of cotrans-
are, the greater the chance that they will be duction frequencies. In an experiment where
integrated together. A formula relating dis- an ilv‡ metE ‡ donor and an ilv metE re-
tance to cotransduction frequency has been cipient are used, when ilv‡ is selected, about
derived, and the relation is nonlinear, reflect- 30% of these transductants are also found
ing these complexities. A theoretical treat- to be metE ‡ . In contrast, when metE ‡ is
ment of this mapping is presented in selected, about 70% of these are found to
Mandecki et al. (1986). be ilv‡ . Thus, although the distance is the
same between these markers in the two
2. Marker effects crosses, different cotransduction frequencies
Although generalized transduction is ``gen- are obtained depending on the selection.
eral'' because any gene in the donor chromo- This could reflect either asymmetries in
some can be introduced into the recipient packaging or recombination. In the case
chromosome, there are quantitative differ- of a packaging asymmetry (Fig. 8), it is pos-
ences in the frequency with which different sible that there are two types of transducing
genes can be transduced. This variation is fragments carrying the ilv‡ gene: a major
as much as 1000-fold for P22 transduction
and 10-fold for P1. Based on the previous
discussion of transduction, there are two
principal sources for variation. The first is
variation due to differences in the packaging
efficiency of different regions of the chromo-
some. This results from the requirement
for specific pac sequences, which would not
be expected to be placed periodically around
the chromosome. This appears to be the
major source of variation in P22 transduc-
Fig. 8. Hypothetical classes of transducing frag-
tion. It is estimated that there are only 10 to
ments in the ilv-metE region and their relative
15 pac sites in the Salmonella chromosome, abundance. When an ilv =metE recipient is trans-
and thus the transduction frequency of a duced to ilv ‡ , about 30% of these transductants
gene is strongly dependent on how close it are also metE‡ . In contrast, when metE‡ is
is to a pac site. In P22 HT mutants, which selected, about 70% of the transductants are ilv ‡ .
To account for the asymmetry by differential pack-
can package from other sites, the variation
aging in the donor, it is proposed that there are
in transduction frequencies is only about three types of transducing fragments packaged
10-fold, and markers that are poorly trans- from this region: a major one containing only ilv,
duced by wild-type P22 often show the a minor one carrying both ilv and metE, and a very
greatest increases with HT. The second minor one with only metE.
572 WEINSTOCK

one that does not contain the metE gene and daughter cells, and thus the daughter cell
a minor one that carries metE. Hence the that does not receive the fragment may
majority of the recombinants formed when remain phenotypically wild-type until the
ilv‡ is selected will retain the recipient's functional product is degraded or diluted
metE marker. On the other hand, there through subsequent cell divisions. Typically,
may be more metE ‡ transducing fragments in a transduction of an auxotrophic recipient
that carry the ilv‡ marker than those that to prototrophy, a large number of microco-
do not carry ilv, resulting in the higher lonies will be observed on minimal medium.
frequency of ilv‡ metE ‡ transductants. It These are abortive transductants that grow
is also possible to account for this difference by virtue of the small amount of nutrient
in cotransduction based on hot spots in made by the diploid. In contrast, the stable
recombination. From these considerations transductants (where the fragment has re-
it is clear that care must be taken in inter- combined into the bacterial chromosome)
preting data based solely on transduction form normal-sized colonies. Another classic
frequencies. At the minimum, reciprocal and beautiful example of this phenomenon is
crosses must be performed to determine if the behavor of nonmotile mutants (Leder-
there are marker effects. Ideally multiple- berg, 1956; Stocker, 1956). In this case the
factor crosses should be used (like the ilv diploid formed by abortive transduction is
cya metE example above) to determine gene motile. On a plate with a low agar concen-
order. tration the diploid can move, but at each cell
division a nonmotile daughter is produced
3. Abortive transduction that will grow into a colony. Eventually a
Not all of the DNA fragments introduced trail of these nonmotile daughters is made,
into a cell by a generalized transducing called a flare, which marks the path of the
phage recombine and integrate into the original abortive transductant.
bacterial chromosome; the fraction that do The unintegrated DNA introduced by
recombine are in fact the minority. The ma- transduction is quite stable in the cell. In
jority of transducing fragments remain in the the case of P1 transduction, when this
cytoplasm of the recipient cell, although the DNA is isolated from a recipient it is found
efficiency of recombination depends on the to be circular, even though the DNA was
region (Schmieger, 1982). These fragments linear in the phage particle (Sandri and Ber-
do not replicate, since they do not contain ger, 1980). The circles can be disrupted by
an origin of replication and thus are in- treatments with detergent or protease, imply-
herited by only one cell at each division. ing that the circle is held together by a pro-
Eventually these fragments are lost from tein linker. Evidence for protein attached to
the culture because very few cells carry an the DNA in P1 transducing particles has in
unintegrated, nonreplicating fragment and fact been reported (Ikeda and Tomizawa,
because they can be degraded. This phenom- 1965b). Normally, a linear double-stranded
enon of creating unstable transductants with DNA molecule is rapidly degraded in vivo
unintegrated fragments is called abortive by the action of the RecBCD nuclease. Thus
transduction. it appears that the protein at the DNA end
The genes in an unintegrated fragment can may be a special mechanism to protect trans-
be expressed and thus the cell is functionally ducing DNA from this degradation.
diploid for this region of its chromosome. If
the cell is mutant for a gene in the diploid 4. Transduction of plasmids
region, while the fragment is wild-type, com- Just as the bacterial chromosome is a sub-
plementation will be observed. At each cell strate for packaging by generalized transdu-
division the functional product generated in cing phages, so is any other DNA that is
the diploid can be distributed into the two inside an infected cell, including plasmid
GENETIC TRANSDUCTION 573

chromosomes. When the plasmid is larger combine with the donor's chromosome and
than a phage headful, only a part of the plas- become integrated. This integrated plasmid
mid can be packaged. This is the case with R may then be packaged and transduced by the
factors, which are much larger than a headful usual mechanism (Trun and Silhavy, 1987).
of a phage like P22. When this occurs, the Once in the recipient, the plasmid can recom-
fragment of the plasmid that is introduced bine out of the chromosome by the reverse
into the recipient cannot circularize efficiently of the integration process, leading to a trans-
because there is no homology between the ductant containing the free plasmid. This
fragment ends or with cellular sequences to is a useful mechanism for selecting for re-
allow recombination. Thus the majority of combination between a plasmid and the host
transduced plasmid fragments will be lost chromosome.
from the recipient. Nevertheless, at a low fre-
E. Uses for Generalized Transduction
quency it is possible to recover deleted ver-
sions of the plasmid following transduction. In the preceding sections several uses for
These represent fragments that retain the plas- generalized transduction have been described.
mid origin of replication. It is possible that These include genetic mapping, complemen-
these are plasmids that acquired a deletion in tation analysis, transduction of plasmids,
the donor that made them small enough to fit selection for deletions in plasmids, and selec-
completely into a phage head. Alternatively, tion for recombination between cloned genes
these may be fragments that were introduced and the bacterial chromosome. Other
into the recipient where they circularized by common uses include strain constructions,
some inefficient ``illegitimate'' recombination delivery of transposons, and isolation of
event. This process is called transductional chromosome rearrangements such as dupli-
shortening (Shipley and Olsen, 1975; also see cations (see Margolin, 1987, for a review).
the review by Low and Porter, 1978).
A different situation is found when the III. SPECIALIZED
plasmid is much smaller than the phage TRANSDUCTION
headful. In this case, the plasmid is packaged Specialized transduction is the second major
inefficiently because the chromosome is too class of virus-mediated genetic exchange. It
small to form a stable head. However, multi- differs from generalized transduction in two
meric forms of the plasmid can be packaged respects. First, in this mode of transduction
efficiently, as shown for transduction of the transduced genes are covalently joined to
plasmids by P22HT mutants (for a review the viral chromosome, allowing them to be
see Margolin, 1987) and T4 transducing replicated, packaged, and introduced into a
phages (Takahashi and Saito, 1982). The recipient with the rest of the viral chromo-
multimeric plasmid chromosomes may be some. Second, a specialized transducing par-
preexisting in the donor or may be caused ticle carries a specific chromosome segment,
by the phage infection, either by phage- and consistently only introduces this set of
promoted recombination between plasmids genes into the recipient. Hence the name,
or rolling circle replication of the plasmid. specialized transduction (see Hendrix, this
In these cases the multimers that are intro- volume, for a more complete discussion).
duced into the recipient circularize by recom-
bination between the repeated plasmid units. A. Bacteriophage l DNA Metabolism
A final case of plasmid transduction in- The temperate phage l (Fig. 9 and Appen-
volves plasmids that are smaller than a dix) is the classic example of a specialized
phage headful but contain sequences that transducing phage. The l phage particle
are homologous to the bacterial chromo- contains a linear double-stranded DNA
some. Examples of this are small plasmid molecule with complementary 12 nucleo-
vectors carrying cloned genes. These can re- tide single-stranded ends (see Furth and
574 WEINSTOCK

life cycles. In the lytic response, the chromo-


some replicates to form concatemers that are
packaged into particles by cutting at the cos
sites, reforming the single-stranded ends.
This packaging differs from the headful
mechanism in that the size of the DNA that
is encapsidated is determined by the distance
between the cos sites and can vary consider-
ably. This is the basis for cosmid cloning
vectors (see Geoghegan, this volume). How-
ever, there are limits on the size of the pack-
aged chromosome. DNA molecules that are
larger than about 50,000 bp, or smaller than
about 35,000 bp, produce unstable phage
particles. The alternative life cycle is the ly-
sogenic response. In this case the phage
chromosome integrates into the bacteria
chromosome and a repressor is produced
that prevents expression of the lytic gene
products. The phage chromosome is repli-
cated and passively carried by the bacterium
as a latent prophage. The integration reac-
Fig. 9. Phage l DNA metabolism. When l infects a
cell (1) the phage DNA circularizes by virtue of the
tion is performed by a phage-encoded site-
single-stranded cohesive ends (cos) and the cell can specific recombination system that breaks
have two fates. If the phage grows lytically (2), the the phage chromosome at a specific se-
chromosome replicates to form concatemers, which quence, attP, and joins it to the host at an-
are then packaged to form new phage (3). Alterna- other specific sequence, attB (see Weisberg
tively (4), the phage chromosome can integrate into
the host chromosome during the lysogenic response.
and Landy, 1983, for a review). When the
This is accomplished by site-specific recombination prophage becomes induced, the phage re-
between the attP site of the phage chromosome and pressor is inactivated and the reverse reac-
the attB site of the host (5). When a lysogen induces, tion occurs. The site-specific recombination
the phage genome excises by a reverse of the integra- system acts at the attP/attB joints to reform
tion process (6), leading to a wild-type chromosome
that can replicate to form concatemers (8) and
a circular chromosome that undergoes the
normal phage. However, on rare occasions the exci- lytic cycle.
sion process is aberrant (7) and the recombination Specialized transducing particles are
does not occur between the att sites. As a result a formed in vivo when an integrated l proph-
chromosome is formed that has lost some phage age excises from the bacterial chromosome
material and acquired some host sequences. As long
as none of the missing phage genes were essential, this
imprecisely (Fig. 9). In this case the breaking
hybrid chromosome is capable of replicating normally and joining events do not occur at the attP/
(8) and producing new phage. These phages are all attB joints, but are the result of ``illegitim-
specialized transducing phages since they carry host ate'' recombination events occurring at other
genes in their chromosomes. sites in the vicinity of the prophage. This
process occurs at low frequency and has little
sequence specificity. Markers either to the
Wickner, 1983, for a review of l DNA me- left or right of the prophage can be trans-
tabolism). When l infects a cell, its chromo- duced. The result is a circular chromosome
some circularizes by hybridization of these that has lost some of the phage DNA se-
single-strand extensions (cos sites), and the quences from one end of the prophage and
phage then chooses between two alternative in their place carries bacterial DNA that was
GENETIC TRANSDUCTION 575

adjacent to the other prophage end. This system at all, but rather utilizes transposons
hybrid chromosome can replicate and be (see Whittle and Salyers, this volume). If a
packaged, provided that the required lytic transposon insertion can be isolated in the
functions and cos site are present, and results bacterial chromosome near a gene of inter-
in the formation of transducing particles that est, then a phage chromosome that also
carry and can introduce the incorporated carries an insertion of the transposon can
bacterial genes. Some specialized transdu- integrate at this site by homologous recom-
cing phages carry the complete array of bination. This is similar to generating F plas-
genes and sites necessary for lytic growth. mid integration sites using transposon Tn10
These are ``plaque-forming'' phages, and insertions (see Porter, this volume). Finally,
this property is usually designated by includ- derivatives of l have been developed that
ing a ``p'' in their name, such as l pgal, a contain the transposition system from phage
plaque-forming gal transducing phage. There Mu (see Whittle and Salyers, this volume)
can also be defective transducing phages. instead of the l site-specific recombination
These are phages that have lost essential system (Bremer et al., 1984). These phages
genes from their chromosome in the process behave like l except that integration during
of excision. The minimum requirement for a lysogenization uses the Mu system and there
phage to be packaged and maintained is that is no sequence specificity. As a result pro-
it contain the cos site and the origin of repli- phages can be isolated anywhere around the
cation. All other functions can be provided bacterial chromosome. This is the most gen-
by helper phages. To designate that a trans- eral in vivo method, and it is as general as the
ducing phage is defective and requires a in vitro method for forming specialized
helper for growth, a ``d'' is included in its transducing phages: molecular cloning using
name, such as lpgal, a defective gal transdu- recombinant DNA methodology.
cing phage.
From this mechanism it is clear that only a B. Events in the Recipient
restricted set of bacterial genes can be trans- When a specialized transducing phage like l
duced. This is a consequence of the fact that stably transduces a recipient cell, it forms a
only genes near the attB site can be incorpor- lysogen that is, it integrates into the chromo-
ated, as well as the limits on the size of the some and expression of lytic functions is
chromosome that can give a stable phage repressed. There are several different ways
particle. However, a number of methods this can occur. If the phage has an intact
have been used to allow a broader range of attP site and int gene (the gene needed for
genes to be transduced by l. Rearrange- site-specific integration at attB), and the re-
ments of the bacterial chromosome, such as pression system of the phage is intact, lyso-
deletions, can bring different genes close geny can follow the same steps as for a wild-
enough to the attB site to be transduced. A type l phage. Often, however, the transdu-
second method relies on the observation that cing phage does not have attP or int, these
when the attB site itself is deleted, l now having been lost during formation of the
integrates at a large number of secondary transducing phage genome. In this case inte-
attachment sites. These sites are not used as gration can occur through homologous
efficiently as attB, presumably because their recombination between the bacterial chro-
sequences deviate from attB. However, lyso- mosomal sequences carried by the phage
gens with prophages located at these second- and its counterpart in the host chromosome.
ary sites can be used to isolate specialized In some cases, if the recipient is lysogenized
transducing phages carrying the neighboring by another wild-type helper l phage, this
genes. In a third method, the site of integra- prophage can also provide a homologous
tion of the phage chromosome does not region for recombination with the transdu-
depend on the site-specific recombination cing phage. Finally, under some conditions,
576 WEINSTOCK

the transducing phage can be maintained as ation of protein and DNA in the fragments. J Mol
Biol 14:110±119.
an un-integrated plasmid in the cell (see
Weisberg, 1987). Lederberg J (1956): Linear inheritance in transductional
clones. Genetics 41:845±871.
C. Uses for Specialized Transduction Low KB, Porter DD (1978):Modes of gene transfer and
recombination in bacteria. Annu Rev Genet 12:249±
Specialized transducing phages provide one 287.
of the most convenient ways of manipulating Mandecki W, Krajewska-Grynkiewicz K, Klopotowski
genes. They represent the original method T (1986): A quantitative model for nonrandom gener-
alized transduction, applied to the phage P22-Salmon-
for gene cloning, using in vivo genetic tech- ella typhimurium system. Genetics 114:633±657.
niques. Because these phages carry a small
Margolin P (1987): Generalized transduction. In Ingra-
region of the chromosome, genes carried on ham JL, Low KB, Magasanik B, Neidhardt FC,
phages can be mapped with great precision, Schaechter M, Umbarger HE (eds): ``Escherichia coli
as can mutations within these genes. More- and Salmonella typhimurlum: Cellular and Molecular
Biology.'' Washington, DC: American Society for
over mutagenesis can be targeted to specific Microbiology, pp 1154±1168.
genes. These phages also provide a con-
Ptashne M (1985): ``A Genetic Switch.'' Boston: Black-
venient means for constructing strains that well Scientific Publications.
are diploid for a very limited region of the
Raj AS, Raj AY, Schmieger H (1974): Phage genes
chromosome, useful for complementation involved in the formation of generalized transducing
analysis. particles in Salmonella-phage P22. Mol Gen Genet
135:175±184.
REFERENCES Sandri RM, Berger H (1980): Bacteriophage P1-
Bremer E, Silhavy TJ, Weisemann JM, Weinstock GM mediated generalized transduction in Escherichia
(1984): l placMu: A transposable derivative of bac- coli: Structure of abortively transduced DNA. Vir-
teriophage lambda for creating lacZ protein fusions in ology 106:30±40.
a single step. J Bacteriol 158:1084±1093. Schmieger H (1972): Phage P22 mutants with increased
Casjens S, Huang WM, Hayden M, Parr R (1987): or decreased transduction abilities. Mol Gen Genet
Initiation of bacteriophage P22 packaging series. An- 119:75±88.
alysis of a mutant that alters the DNA target specifi- Schmieger H (1982): Packaging signals for phage P22 on
city of the packaging apparatus. J Mol Biol 194:411± the chromosome of Salmonella typhimurium. Mol Gen
422. Genet 187:516±518.
Chelala CA, Margolin P (1974): Effects of deletions on Shipley PL, Olsen RH (1975): Isolation of a nontrans-
cotransduction linkage in Salmonella typhimurium: missible antibiotic resistance plasmid by transduc-
Evidence that bacterial chromosome deletions affect tional shortening of R factor RP1. J Bacteriol
the formation of transducing DNA fragments. Mol 123:20±27.
Gen Genet 131:97±112.
Sternberg N (1986): The production of generalized
Ebel-Tsipis J, Botstein D, Fox MS (1972): Generalized transducing phage by bacteriophage lambda. Gene
transduction by phage P22 in Salmonella typhimur- 50:69±85.
ium. I. Molecular origin of transducing DNA. J Mol
Biol 71:433±448. Sternberg N, Coulby J (1987): Recognition and cleavage
of the bacteriophage P1 packaging site (pac). II. Func-
Furth M, Wickner S (1983): Lambda DNA replication. tional limits of pac and location of pac cleavage ter-
In Hendrix R, Roberts J, Stahl F, Weisberg R (eds): mini. J Mol Biol 194:469±479.
``Lambda II.'' Cold Spring Harbor, NY: Cold Spring
Harbor Laboratory, pp 145±174. Sternberg N, Hoess R (1983): The molecular genetics of
bacteriophage P1. Annu Rev Genet 17:123±154.
Hendrix R, Roberts J, Stahl F, Weisberg R (eds) (1983):
``Lambda II.'' Cold Spring Harbor, NY: Cold Spring Stocker BAD (1956): Abortive transduction of motility
Harbor Laboratory. in Salmonella: A nonreplicated gene transmitted
through many generations to a single descendant. J
Howe M (1973): Transduction by bacteriophage Mu-1. Gen Microbiol 15:575±598.
Virology 55:103±117.
Susskind MM, Botstein D (1978): Molecular genetics of
Ikeda H, Tomizawa J (1965a): Transducing fragments in bacteriophage P22. Microbiol Rev 42:385±413.
generalized transduction by phage P1. I. Molecular
origin of the fragments. J Mol Biol 14:85±109. Takahashi H, Saito H (1982): High-frequency transduc-
tion of pBR322 by cytosine-substituted T4 bacterio-
Ikeda H, Tomizawa J (1965b): Transducing fragments phage: Evidence for encapsulation and transfer of
in generalized transduction by phage P1. II. Associ- head-to-tail plasmid concatemers. Plasmid 8:29±35.
GENETIC TRANSDUCTION 577

Trun NJ, Silhavy TJ (1987): Characterization and in vivo ologous (exo and bet) recombination systems
cloning of prlC, a suppressor of signal sequence mu-
tations in Escherichia coli K12. Genetics 116:513±521. of phage l as well as a number of genes
Weisberg RA (1987): Specialized transduction. In Ingra-
whose products are involved in interactions
ham JL, Low KB, Magasanik B, Neidhardt FC, with the host. Next is a short stretch contain-
Schaechter M, Umbarger HE (eds): ``Escherichia coli ing the important control genes N, cl, cro,
and Salmonella tymphimurium: Cellular and Molecu-
lar Biology.'' Washington, DC: American Society for
and cll, followed by the DNA replication
Microbiology, pp 1169±1176. region (genes O and P and the origin of
Weisberg R, Landy A (1983): Site-specific recombin- replication), the controller of late gene ex-
ation. In Hendrix R, Roberts J, Stahl F, Weisberg R pression, Q, and the genes for cell lysis, S,
(eds): ``Lambda II.'' Cold Spring Harbor, NY: Cold R, and Rz. This overall functional arrange-
Spring Harbor Laboratory, pp 211±250.
ment along the chromosome is followed by a
Wilson GG, Young KKY, Edlin GJ, Konigsberg W
(1979): High frequency generalized transduction by
number of other phages of E. coli (e.g.,
bacteriophage T4. Nature 280:80±82. phages 434 and 21) as well as phage P22 of
Zinder ND, Lederberg J (1952): Genetic exchange in Salmonella. Because of this relatedness, these
Salmonella. J Bacteriol 64:679±699. phages are collectively referred to as the lam-
boid phages, and it has been possible to
APPENDIX: create hybrids in which functions in one
lambdoid phage are replaced with the analo-
BACTERIOPHAGE l
gous genes from another phage in this race.
The bacteriophage l is one of the most thor-
oughly studied systems in biology. The DNA Transcriptional Units
sequence of the entire phage l chromosome There are four major transcriptional units in
has been determined (over 48,500 bp), and the phage (Fig. A1). The promoter PL is used
this organism has served as a paradigm for for the early leftward operon and the pro-
many phenomena in addition to genetic moter PR is used for the early rightward
transduction. Much of the information operon. The genes in these two operons are
about phage l is summarized in the mono- made early in infection. The PR0 promoter
graph Lambda II (Hendrix et al., 1983). An- (sometimes called the late promoter) drives a
other good reference is the book A Genetic large operon consisting of genes that are
Switch (Ptashne, 1985). The metabolism of l expressed late in infection, such as those for
DNA during infection has been described head and tail production and cell lysis. Last
above. Here we present an overview of the is the transcription unit that contains the cl
genetic organization of the phage and some gene, encoding the l repressor. There are
of the important aspects of the control of two promoters for this purpose: PRE , which
phage l gene expression to complement the is used early in infection, and PRM , which is
chapter by Hendrix, this volume. used in a repressed lysogenic cell. In addition
to these is the P1 promoter, which produces a
Genetic Organization small RNA to augment the expression of the
The l chromosome encodes about 50 genes int gene, and the Po promoter, which makes
that are organized in functionally related another small RNA of unknown function.
clusters. At the left end of the chromosome
(Fig. A1) is a block of 10 genes that is re- Lytic Growth
quired for production of the phage head In the lytic response, l replicates its DNA,
followed by 11 genes needed to make the packages the new chromosomes into phage
phage tail. The central region of the chromo- heads, adds phage tails to these, and finally
some contains 19 nonessential genes that are lyses the cell. The genes for head and tail
also functionally clustered. Included in this production and cell lysis are contained in
region are the genetic elements of the site- the later operon, expressed from the PR0 pro-
specific (int, xis, and the att site) and hom- moter. However, essential control genes and
Fig. A1. Phage l genetic map. The details of the map are described in the appendix. Arrows indicate direction of transcriHption of the different operons.
GENETIC TRANSDUCTION 579

the DNA replication functions, located in chromosome. It is passively carried in this


the early leftward and rightward operons, latent state as a prophage until it becomes
must be expressed first. induced. The repressor that turns off all of
The leftward operon contains the essential the lytic genes is the product of the cl gene. It
N gene, which is an antiterminator of tran- binds to the phage chromosome at two
scription. Transcription termination signals regions: OL and OR (Fig. A1). Each of these
are located throughout the leftward and right- is a cluster of three operator sites and bind-
ward operons. There is a terminator immedi- ing of the l repressor causes repression of
ately after the N gene as well as at other transcription from the PL and PR promoters.
downstream sites in this operon. In the right- This in turn prevents all lytic gene expression
ward operon there are terminators after the by preventing expression of the control genes
cro and P genes. The N gene product prevents N and Q.
transcription from terminating at these sites Expression of the cl gene involves two
and thus allows expression of downstream promoters. The PRE promoter (for the estab-
genes. This provides a timing mechanism. lishment of repression) is used early in infec-
Genes preceding the terminators (N and cro) tion when the decision between the lytic and
are early genes, while the others are sometimes lysogenic choices has not been resolved. This
called delayed-early genes, since their expres- is a strong promoter that is stimulated by the
sion is delayed until N protein is available. product of the cll gene, located in the early
In the rightward operon there are several rightward operon. This promoter allows a
essential functions for lytic development. lot of repressor to be made quickly. Once a
First are the DNA replication genes O and lysogen has been established, PRE is not used
P and the origin of replication. These are and expression switches to the PRM pro-
located after the first transcription termin- moter (for maintenance of repression). This
ation signal in this operon. In addition is is a weaker promoter that produces enough
the Q gene, located after the second termin- repressor to maintain the repressed state. It
ator, which thus delays its expression fur- can be either stimulated or inhibited by l
ther. The Q gene product is required for the repressor bound at OR (depending on the
expression of all late proteins (i.e., produc- repressor concentration) and thus allows
tion of heads, tails, and cell lysis). This is for buffering of the amount of repressor pro-
because there is a transcription terminator duced in the lysogen.
just after the late promoter P0R and immedi- The other important control protein in the
ately before all of the late genes. The Q l system is the cro gene product, which
protein is a transcription antiterminator serves to counteract the effect of cl. The cro
that is specific for this site and thus controls protein is also a repressor and it also binds at
late gene expression. The delay in expression the sites at OR and OL . However, its binding
of Q contributes to the delay in expression of characteristics are different from cl and as a
the late genes and ensures that plenty of result it can antagonize cl binding and lead
DNA has accumulated before the packaging to a lytic response. The decision between the
and lysis processes begin. lytic and lysogenic response involves the
competition for control of the OR and OL
The Lysogenic Response sites by cl and cro. This competition is ultim-
In the lysogenic response l turns off the lytic ately decided by the physiology of the host,
genes by the action of a single repressor and which influences expression of these genes in
integrates its chromosome into the bacterial ways that are not at present clear.
isotope effect, reported by Baker (6, 7), ing of A-NADD to heart lactate de- 6. R. H. Baker, Jr., Biochemistry 1, 41 (1962).
7. H. R. Mahler, R. H. Baker, V. J. Shiner, Jr.,
for B-NADD of 0.50 + 0.10 based on hydrogenase (Kd = 0.69) and muscle ibid., p. 47.
kinetic measurements, it is in complete lactate dehydrogenase (Kd = 0.43). 8. D. Palm, T. Fielder, D. Ruhrseitz, Z.
Naturforsch. 23b, 623 (1968).
agreement with our most recent kinetic These various results in conjunction 9. K. Bush, H. R. Mahler, V. J. Shiner, Jr.,
results (9). Our observations also indi- with the absence of any real difference in preparation.
10. Abbreviations: LADH, horse liver alcohol
cate that the B-NADD preparation is in the binding constant between NADH dehydrogenase; A-NADD and B-NADD,
more sensitive to deterioration on stor- and B-NADD, which eliminates the NADH with deuterium in the 4-A and 4-B
nicotinamide positions, respectively; Kd, dis-
age than the usual NADH sample. This possibility of secondary interaction be- sociation constant.
is probably the cause of the discrepancy tween the B proton and the enzyme, 11. H. Theorell, A. Ehrenberg, C. de Zalenski.
Biochem. Biophys. Res. Commun. 27, 309
between Baker's results and those re- indicate that dehydrogenases are capa- (1967).
12. R. Pietruszko, H. J. Ringold, T. K. Li, B.
ported here. ble of recognizing and discriminating L. Vallee, A. Akeson, H. Theorell, Nature
The isotope effect for equilibrium against coenzymes isotopically substi- 221, 440 (1969).
13. S. Taniguchi, H. Theorell, A. Akeson, Acta
dissociation constants with A-NADD tuted in the appropriate position ("A" Chem. Scand. 21, 1903 (1967); H. Theorell,
was determined by both the Sephadex for A enzymes) even in the initial bind- A. Akeson, B. Liszka-Kopec, C. de Zalenski,
Arch. Biochem. Biophys. 139, 241 (1970).
and the fluorimetric methods, and the ing reaction. The properly deuterated 14. K. Dalziel, Acta Chem. Scand. 12, 459 (1958).
two results are identical within experi- coenzyme is bound less tightly than is 15. S. Taniguchi, ibid. 21, 1511 (1967).
16. S. P. Colowick and N. 0. Kaplan, Methods
mental error: its protonated counterpart, so that dur- Enzymol. 4, 840 (1957).
ing the binding reaction the equilibrium 17. A. D. Winer, J. Biol. Chem. 239, PC 3598
Kd -KdE-NADIT/KdE-(A-NADD) (1964).
(and perhaps also the rate) constant 18. J. P. Hummel and W. J. Dreyer, Biochim.
= 0.65 ± 0.14 (Sephadex method) already reflects the specificity of the Biophys. Acta 63, 530 (1962).
19. R. W. Green and R. H. McKay, J. Biol.
= 0.52 ± 0.07 (fluorimetric method) subsequent hydrogen transfer step. Chem. 244, 5034 (1969); H. Jornwall, Eur.
J. Biochem. 16, 25 (1970).
This kind of ("inverse") isotope effect KAREN BUSH, H. R. MAHLER 20. G. F. Fairclough, Jr., and J. S. Fruton, Bio-
indicates that A-NADD is bound less V. J. SHINER, JR. chemistry 5, 673 (1966).
20. H. Theorell and A. D. Winer, Arch. Bio-
tightly to the enzyme than is NADH: Department of Chemistry, chem. Biophys. 83, 291 (1959).
Indiana University, Bloomington 47401 22. S. R. Anderson and G. Weber, Biochemistry
ki 4, 1948 (1965).
LADH + NADH = LADH - NADH 23. K. Dalziel, Acta Chem. Scand. 17, S27 (1963).
References and Notes 24. H. Theorell and J. S. McKinley-McKee, ibid.
k2 15, 1811 (1961).
1. V. J. Shiner, Jr., H. R. Mahler, R. H. Baker, 25. We thank Mrs. Genevieve Adams for techni-
and that the zero point energies for the Jr., R. R. Hiatt, Ann. N.Y. Acad. Sci. 84, cal assistance in the preparation of the deu-
583 (1960). terated coenzymes and the laboratory of Dr.
isotopically sensitive vibrations are low- 2. H. R. Mabler and J. Douglas, J. Amer. Drew Schwartz for perforniing the starch-
er in the complex than in the free Chem. Soc. 79, 1159 (1957); D. Palm, Z. gel electrophoresis. We thank Dr. Henry
Naturforsch. 21b, 540, 547 (1966). Weiner of Purdue University who donated his
NADD. 3. J. F. Thomson, D. A. Bray, J. J. Bummert, laboratory facilities and computer program
Since Kd (observed) = KH/KD = Biochenm. Pharmacol. 11, 943 (1962). for the fluorimetric determinations. Supported
4. J. F. Thomson, J. J. Darling, L. F. Bordner, in part by grant RG-8502 from NIH and by
k2/k1; k, - klHIk1D, k2 -- k2E/k2D, Biochim. Biophys. Acta 85, 177 (1964); J. F.
Thomson and S. L. Nance, ibid. 99, 369
grant AT(11-1)-1008 from AEC (Document
No. COO-1008-17).
then if Kd < 1 and I2 > 1 it follows (1965).
that k1 > k2 > 1 (conversely for k2 < 1, 5. D. Palm, Euir. J. Biochem. 5, 270 (1968). 9 November 1970; revised 14 December 1970 U
k, < k2). Therefore if the isotope effect
on the dissociation reaction (rate con-
stant k2) is normal (k2 > 1), a possi-
bility suggested from kinetic data (6, Bacteriophage and the Toxigenicity of
7, 9), then the isotope effect for the Clostridium botulinum Type C
binding step (rate constant kl) must
also be normal and even larger, thus Abstract. Nontoxigenic and bacteriophage-sensitive bacterial cultures have been
indicating an ability of the enzyme to isolated from toxigenic Clostridium botulinum type C, strain 468C, after treat-
discriminate kinetically against the co- ment with either ultraviolet light or acridine orange. Two bacteriophages, desig-
enzyme deuterated at the transferable nated CE/3 and CE-y, were isolated from toxigenic strain 468C. Both of these
hydrogen.
Previously Mahler et al. (7) reported bacteriophages were capable of infecting the nontoxigenic type C cultures, but
Kd for the LADH-(A-NADD) dissocia- only bacteriophage CE/ was involved in the change from nontoxigenicity to
tion constant to be 1.31 + 0.64; how- toxigenicity.
ever, this value was based on an extrap- Inoue and lida (5) presented data which
olation from kinetic constants derived Nontoxigenic clostridia resembling
from initial rate measurement and as toxigenic Clostridium botulinum have strongly suggested that bacteriophage
indicated, exhibited a large experimental frequently been isolated from marine may be involved in the toxigenicity of C.
uncertainty. Isotope effects for the cor- and terrestrial environments (1); oc- botulinum type C. They were able to
responding equilibrium constants for casionally, pure cultures of toxigenic C. recover toxigenic isolates from nontoxi-
yeast alcohol dehydrogenase (1) and botulinum have also become nontoxi- genic cultures incubated in broth with
malate dehydrogenase (3) were reported genic (2). It has been suggested that the filtrates of a toxigenic type C (strain
to be of the same magnitude as that production of toxin by C. botulinum Stockholm). Marked lysis occurred in
for LADH. However, "inverse" deu- might be governed by the presence of the cultures; however, plaques were not
terium isotope effects similar to the one a specific converting bacteriophage in a demonstrated on solid medium.
reported here, but based on inverse system analogous to the production of In our studies, two bacteriophages,
rate effects for kl, were reported from diphtheria toxin by Corynebacterium designated CE,8 and CE-y, were isolated
Thomson's laboratory (4) for the bind- diphtheriae (3, 4). In a recent report, from toxigenic C. botulinum type C,
480 SCIENCE, VOL. 172
strain 468C (received from W. P. Seg- Table 1. Reinfecting cured nontoxigenic bac- of its prophage. After a 60-second treat-
ner, Continental Can Co., Chicago). terial cultures with bacteriophage CEj8 and the ment with ultraviolet light, 15 of the 106
effect on toxigenicity. UV, ultraviolet.
This report presents conclusive evidence surviving colonies were cured of their
for the involvement of CE/3 in the toxi- Nontoxi- Method of No. of cultures
genic obtaining prophages and concomitantly became
genicity of C. botulinum type C. Bac- cured toxigenic Toxi- Tested nontoxigenic. In comparison, only 2 of
teriophage-producing cultures were ob- culture cultures genic the 68 colonies surviving the AO treat-
tained from nontoxigenic type C cultures A028 Plaques* 40 40 ment were cured of their prophages and
that had been reinfected with either bac- A028 30-minute 4 40 toxigenic character. The cured cultures
teriophage. However, only bacteriophage contactt were subcultured ten times over a period
CE,8 caused the change from nontoxi- A028 240-minute 40 40 of 2 months, and all of them remained
contactt
genicity to toxigenicity, and these toxi- UV171 Plaques 20 20 nontoxigenic and sensitive to the bac-
genic isolates invariably carried bac- UV171 30-minute 2 40 teriophages of the toxigenic parent strain
teriophage CE,8. Cultivation of the contact 468C.
CE,8-infected isolates in medium con- UV171 240-minute 40 40 Each nontoxigenic culture cured of its
contact
taining antiserum against bacteriophage prophage was tested for its sensitivity to
* Material from plaques produced by bacteriophage
CE,B resulted in the loss of bacteriophage CEfl on culture A028 or culture UV171 was lysates of the other cured nontoxigenic
CE,8 and loss of toxigenicity. These iso- checked for toxigenicity and bacteriophage produc- cultures by the agar-layer procedure.
lates were then reinfected with bac- tion. t Cultures A028 and UV171 were ex-
posed to bacteriophage and isolated colonies were Only one culture, A028, was sensitive to
teriophage CE,8 and they again became checked for toxigenicity and bacteriophage pro- the lysates of the other nontoxigenic
duction.
toxigenic. cultures. Culture A028, therefore, had
Nontoxigenic cultures were isolated been cured of two of its prophages,
after treatment of toxigenic strain 468C whereas the other cultures continued to
with either ultraviolet irradiation or (pH 7.4) containing 20 ,tug of AO per carry a second prophage.
acridine orange (AO). Irradiation with milliliter and incubated at 37°C for 18 Bacteriophages were isolated from an
ultraviolet light is a standard method for hours. The AO culture was streaked 18-hour broth culture of the toxigenic
inducing lysis of bacteria that harbor onto TYG blood agar plates, and iso- parent strain 468C. The broth culture
prophages (the form in which bac- lated colonies were tested for toxigenicity was centrifuged at 6000g for 10 minutes
teriophages are perpetuated in lysogenic and sensitivity to bacteriophages by the and sterilized by filtration. Colony-
bacteria), whereas AO has been used same procedures that were described for centered plaques and turbid plaques pro-
to remove extra chromosomal elements the colonies treated with ultraviolet light. duced by bacteriophages designated as
from bacteria. Both procedures can re- Ultraviolet treatment was more effi- CE,B and CEy, respectively, were ob-
sult in bacterial isolates that are "cured" cient than AO was in curing strain 468C served when the filtrate was plated with
of either prophages in the former or
extra chromosomal elements in the
latter.
For irradiation, about 106 cells of
strain 468C in the logarithmic phase of
growth were spread onto the surfaces of
trypticase, yeast-extract, glucose (TYG)
blood agar plates and irradiated for 60
seconds at a distance of 20 cm (General
Electric germicidal lamp, 15 watts).
The agar plates were incubated anaero-
bically in Brewer jars for 48 hours at
37°C. Isolated colonies surviving the
treatment (approximately 102 cells)
were tested for toxigenicity by the mouse
assay (6) and for sensitivity to the
bacteriophages of the toxigenic parent
culture by the agar-layer procedure (7).
The base agar used in the agar-layer pro-
cedure was TYG agar, and the overlay
was soft agar (0.7 percent) prepared
from the filtered broth of Segner's forti-
fied egg meat (SFEM) medium (8).
The addition of blood (0.2 ml) to the
overlays was essential for confluent
growth. Cysteine hydrochloride (0.1
percent) was used as the reducing agent
in all media.
For curing with AO, about 105 cells
of strain 468C in the logarithmic phase Fig. 1. Bacteriophage CE,8, from lysate of Clostridium botulinum type C, strain 468C,
of growth were added to TYG broth which induces toxigenicity (X 247,500). Scale, 20 nm.
30 APRIL 1971 481
culture A028. These two different bac- These cultures were subcultured in
teriophages were purified by five succes- SFEM six times over a period of 2
sive single-plaque isolations on culture months, and all of them maintained
A028. their toxigenic and bacteriophage-pro-
Electron micrographs were prepared ducing characteristics.
of the two bacteriophages according to Experiments reported in Table 1 were
the procedure of Eklund et al. (9). Bac- repeated with bacteriophage CEy and
teriophage CE/? exhibited a hexagonal with bacterial culture A028 (data not
head 100 nm in diameter and a tail 400 shown). Colonies from a 240-minute
nm long and 11 nm in diameter sur- contact between bacteriophage CEy and
rounded by a contracted sheath 30 nm culture A028 were all bacteriophage-
in diameter (Fig. 1). Bacteriophages producers but nontoxigenic.
comparable to CE/3 in size and mor- There is little doubt that in our ex-
phology have been observed in lysates periments toxigenicity of C. botulinumn
of other strains of type C (5, 10). Bac- type C (strain 468C) is in some manner
teriophage CE-y exhibited a hexagonal dependent on the presence of bacterio-
head 60 nm in diameter and a tail 185 phage CE/B. Groman (4) also reported
nm long and 6 nm in diameter sur- that conversion of toxigenicity of
rounded by a contracted sheath 20 nm Corynebacterium diphtheriae is bac-
in diameter (Fig. 2). teriophage specific. The curing of CE,B-
Both bacteriophages were studied for infected cultures by antiserum against
their relation to the toxigenicity of type the bacteriophage indicates that this
C strain 468C. Bacteriophage stocks relation between bacteriophage CE/l
used in these studies were produced by Fig. 2. Bacteriophage CEy, from lysate of and host is probably pseudolysogeny,
propagating the purified bacteriophages Clostridium botulinium type C, strain 468C, similar to that which occurs with Bacil-
which does not induce toxigenicity (X
with culture A028 in TYG broth at 247,500). Scale, 20 nm. lus subtilis (11), instead of true
30(C. Bacteriophage stocks were lysogeny.
treated with 25 ug of crystalline deoxy- Some of the other types of C. botu-
ribonuclease II (Sigma) per milliliter streaked onto TYG blood agar plates, linum that we have studied have been
for 3 hours at 37°C and then sterilized and isolated colonies were transferred cured of their prophage but still remain
by filtration. This step was used to rule into TYG broth, incubated, assayed for toxigenic. It is possible either that they
out the possibility of a transformation toxin, and tested for bacteriophage pro- carry other bacteriophages or that not
principle of the deoxyribonucleic acid duction, with culture A028 as the indi- all C. botulinum toxins are induced by
type. Sterility checks were made by cator strain. bacteriophage.
inoculating several milliliters of filtrate After a 30-minute contact with bac- M. W. EKLUND
into SFEM broth and then incubating teriophage CEj/, 10 percent of the colo- F. T. POYSKY
the medium for several weeks at 37°C. nies of culture A028 and 5 percent of S. M. REED
Two techniques were used to test the the colonies of culture UV171 were C. A. SMITH
relation of the two bacteriophages to toxigenic (Table 1). The toxigenic cul- National Marine Fisheries Service
the toxigenicity of cultures A028 and tures were resistant to bacteriophage Technology Laboratory,
UV171 (which carries bacteriophage CE,I. The nontoxigenic cultures con- 2725 Montlake Boulevard East,
CEy). In the first, dilutions of bacterio- tinued to be sensitive to bacteriophage Seattle, Washington 98102
phage CE/I were plated with the recipi- CE/I? but when they were reinfected
ent nontoxigenic culture according to References and Notes
with this bacteriophage they also be-
the agar-layer procedure. Material from came toxigenic and produced bacterio- 1. C. E. Dolman, M. Tomsich, C. C. R. Camp-
the plaques was transferred into TYG bell, W. B. Laing, J. Infect. Dis. 106, 5
phage. After a 240-minute contact be- (1960).
broth and incubated at 30°C for 5 days. tween bacteriophage and bacteria, all 2. C. E. Dolman and L. Murakami, ibid. 109,
107 (1961).
Cultures arising from the plaques pro- of the isolates were toxigenic and pro- 3. V. J. Freeman, J. Bacteriol. 61, 675 (1951).
duced by CE/I on cultures A028 and duced bacteriophage (Table 1). 4. N. B. Groman, ibid. 69, 9 (1955).
5. K. Inoue and H. lida, Jap. J. Microbiol. 14,
UV171 produced bacteriophage and Several of the CE/I-infected cultures 87 (1970).
were toxigenic (Table 1). Cultures aris- were subcultured in TYG broth con- 6. M. W. Eklund, F. T. Poysky, D. I. Wieler,
Appl. Microbiol. 15, 1316 (1967).
ing from the plaques produced by CEy taining 50 percent antiserum against 7. M. H. Adams, Bacteriophage (Interscience,
on culture A028 also produced bac- bacteriophage CE,B (produced by im- New York, 1959).
8. W. P. Segner, C. F. Schmidt, J. K. Boltz,
teriophage but were nontoxigenic (data munizing rabbits with CE,/ bacterio- Report No. COO-1183-32 (U.S. Atomic Energy
not shown). phage). Four successive transfers in Commission, Washington, D.C., 1970).
9. M. W. Eklund, F. T. Poysky, E. S. Boatman,
In the second procedure, bacterio- antiserum resulted in the simultaneous J. Virol. 3, 270 (1969).
phage CE/I was added to actively grow- loss of bacteriophage CE,8 and toxi- 10. G. Vinet and V. Fredette, Rev. Ca,t. Biol. 27,
73 (1968).
ing nontoxigenic cultures of A028 and genicity. When these nontoxigenic cul- 11. K. Bott and B. Strauss, Virology 25, 212
UVI 71 at a multiplicity of infection tures were reinfected with bacterio- (1965).
12. We thank Dr. E. S. Boatman, University of
(ratio of adsorbed bacteriophage to phage CE,/, they again became toxi- Washington, Seattle, for preparation of the
bacteria) of 1.8 and 2.6, respectively. genic. electron micrographs, and Miss Doris Huff
for technical assistance. Supported by AEC
After 30 and 240 minutes of exposure The toxins of the different isolates contract No. AT(949-7)-2442.
to the bacteriophage, the cultures were were neutralized with type C antitoxin. 18 January 1971
482 SCIENCE, VOL. 172
INFECTION AND IMMUNITY, Aug. 2002, p. 3985–3993 Vol. 70, No. 8
0019-9567/02/$04.00⫹0 DOI: 10.1128/IAI.70.8.3985–3993.2002
Copyright © 2002, American Society for Microbiology. All Rights Reserved.

MINIREVIEW

Bacteriophage Control of Bacterial Virulence


Patrick L. Wagner and Matthew K. Waldor*
Howard Hughes Medical Institute and New England Medical Center, Boston, Massachusetts 02110

In 1930, Felix d’Herelle wrote “. . .the actions and reactions variable in bacterial pathogenesis has proven correct. How-
are not solely between these two beings, man and bacterium, ever, while d’Herelle emphasized the diminution of bacterial
for the bacteriophage also intervenes; a third living being and, virulence by bacteriophages (19), we have instead come to
hence, a third variable is introduced” (19). The contribution of learn that phages serve as a driving force in bacterial patho-

Downloaded from http://iai.asm.org/ on August 12, 2014 by guest


bacteriophages to the pathogenicity of their bacterial hosts genesis, acting not only in the evolution of bacterial pathogens
began to be uncovered as early as 1927, when Frobisher and through gene transfer, but also contributing directly to bacte-
Brown discovered that nontoxigenic streptococci exposed to rial pathogenesis at the time of infection. This review provides
filtered supernatants of toxigenic streptococcal cultures ac- a discussion of (i) the discovery of phage-encoded virulence
quired the ability to produce scarlatinal toxin (28). We now factors, (ii) bacterial virulence properties altered by phages,
know that these supernatants contained a bacteriophage en- (iii) regulation of phage-encoded virulence factors, and (iv) the
coding the scarlatinal toxin and that these investigators were role of in situ prophage induction in the control of bacterial
describing transduction, i.e., the transfer of genetic material to virulence.
a bacterial cell via phage infection. Although these early in-
vestigators lacked a mechanistic explanation for their observa-
DISCOVERY OF PHAGE-ENCODED
tions, they postulated that the bacterium was “within certain
VIRULENCE FACTORS
limits, perhaps a matter of secondary importance and that
toxicogenicity might be a property that could be acquired by Historically, the discovery that a virulence factor was phage
different types of organisms.” (28). Their hypothesis that bac- encoded relied upon observation of transmissibility of the fac-
teria acquire virulence properties has since gained widespread tor among bacterial strains, followed by demonstration, some-
acceptance, as many virulence genes have been shown to un- times much later, that the relevant genes were located on a
dergo transfer among bacteria by phages (via transduction) phage chromosome. The scarlatinal and diphtheria toxins, for
and other mobile genetic elements such as plasmids (via con- example, were proven to be phage encoded many years after
jugation). their production was initially reported to be transmissible (27,
Over time, a number of toxin genes were found to be phage 28, 38, 44, 90, 102, 108). Later, genetic tools such as trans-
encoded, and consideration of the role of phages in bacterial posons TnphoA and Tn10d-bla (4, 73) were developed to aid in
pathogenesis emphasized the dissemination of toxin genes the discovery of phage-encoded virulence factors. Since PhoA
among bacterial strains (10). However, it has become increas- and Bla must be secreted in order to be active, screening fusion
ingly clear that toxin genes are only a subset of the diverse libraries for transducible PhoA or Bla activity allowed for the
virulence factors encoded by bacteriophages. For example, rapid identification of secreted, phage-encoded putative viru-
some phages encode regulatory factors that increase expres- lence factors, including the ␭-encoded lom and bor (4) genes of
sion of virulence genes not encoded by the phage (84), while Escherichia coli and the phage K139-encoded glo gene of Vibrio
others encode enzymes that alter bacterial components related cholerae (73).
to virulence (31, 58). Furthermore, phages have unique prop- Phage-encoded genes are not always transmissible, however,
erties that enable them to contribute more directly to bacterial either due to technical limitations in detecting transduction or
virulence than via transduction. Structural components of due to the fact that integrated prophages frequently become
virion particles, for example, may be directly pathogenic (5, 6, defective. In the present era, analysis of the genome sequences
99). Additionally, phage-encoded genes frequently undergo of bacterial pathogens can expeditiously reveal whether viru-
replication and transcriptional activation following prophage lence factors are associated with phage-like DNA sequences
induction, a process that was speculated to have a role in the regardless of whether they are transmissible. For example, the
production of diphtheria toxin by Corynebacterium diphtheriae nontransmissible (86) stx genes in Shigella dysenteriae are ad-
as early as 1960 (3). jacent to lambdoid phage-like sequences interrupted by nu-
Since d’Herelle’s time, his notion of the phage as a third merous insertion sequences, suggesting that the toxin genes lie
in a prophage that has been rendered defective by the insertion
sequences (59). Moreover, transduction of a virulence gene is,
* Corresponding author: Division of Geographic Medicine and In- in and of itself, insufficient evidence that the gene resides in a
fectious Diseases, Tufts University School of Medicine and New En-
gland Medical Center, NEMC #041, 750 Washington St., Boston, MA
phage genome. Generalized transducing phages can package
02110. Phone: (617) 636-7618. Fax: (617) 636-5292. E-mail: mwaldor and transmit any chromosomal locus, and specific interactions
@lifespan.org. between generalized transducing phages and chromosomal vir-

3985
3986 MINIREVIEW INFECT. IMMUN.

ulence genes have been described. For example, the Staphylo- sule of GAS is composed solely of hyaluronic acid, the
coccus aureus pathogenicity island SapI1, which contains the hyaluronidase presumably benefits the phage by aiding in cap-
gene for toxic shock syndrome toxin (tst), is mobilized at high sule penetration during its infection of or release from strep-
frequency by the generalized staphylococcal transducing phage tococci. This phage-associated hyaluronidase activity may also
80␣ (76); a similar mechanism has not been ruled out for the aid the streptococci in their spread through human connective
transmission of the V. cholerae pathogenicity island (VPI), tissues. Although this hypothesis is untested, antibody to
which was reported to correspond to the genome of a phage phage-encoded hyaluronidase is detectable in sera of patients
(49). infected by GAS (32).
Currently, analysis of sequences surrounding virulence fac- The invasive properties of Salmonella enterica serovar Ty-
tor genes offers the most direct and sensitive method of deter- phimurium require the Salmonella pathogenicity island 1
mining whether virulence genes are associated with phage-like (SPI1)-encoded type III secretion system, which injects effec-
sequences. While this approach can reveal whether a virulence tor proteins directly into the cytoplasm of host cells (40).
gene is associated with phage sequences, it cannot reveal Among the numerous effector proteins exported via this secre-
whether the gene is part of a prophage capable of transducing tion system is SopE, which activates human Rho GTPases and
it or of influencing its expression. Mutational analyses can contributes to efficient entry of Salmonella into tissue culture

Downloaded from http://iai.asm.org/ on August 12, 2014 by guest


complement sequence analysis in this respect, revealing, for cells (33, 34, 105). The sopE gene is located on a temperate
example, the necessity of phage morphogenesis genes for Salmonella phage, designated SopE␾ (62). Since effector pro-
transduction (100) or the necessity of phage regulatory se- teins such as SopE interact intimately with mammalian intra-
quences for virulence factor production (98). cellular components, lysogeny thus aids in the adaptation of
Salmonella to its mammalian host. In this instance, two acces-
sory genetic elements (SPI1 and SopE␾) interact to enhance
BACTERIAL VIRULENCE PROPERTIES ALTERED
the virulence of a bacterial pathogen. Following epithelial
BY PHAGES
translocation, Salmonella preferentially localizes to Peyer’s
Historically, exotoxin production has been the most widely patches within the bowel wall. Stanley et al. identified a Sal-
recognized bacterial characteristic linked to bacteriophage in- monella gene, gipA, encoded by phage Gifsy-1, that is ex-
fection (10). However, phage infection of bacteria is increas- pressed specifically in the Peyer’s patch and is necessary for
ingly associated with additional effects on bacterial virulence. optimal survival in this environment (85). Therefore, two dif-
Phages can alter host bacterial properties relevant to all stages ferent phage-encoded genes facilitate two separate early steps
of the infectious process (Table 1), including bacterial adhe- during Salmonella infection, namely, uptake by intestinal epi-
sion, colonization, invasion, and spread through human tissues; thelial cells (sopE) and survival in the Peyer’s patch (gipA).
resistance to immune defenses; exotoxin production; sensitivity Phages enhance bacterial resistance to serum and phago-
to antibiotics; and transmissibility among humans. cytes. During infection, bacterial pathogens encounter the se-
Phages influence bacterial adhesion, colonization, and in- rum- and phagocyte-mediated elements of the innate immune
vasion. Early events during bacterial infection include adhe- system. Staphylococci produce a number of proteins involved
sion, colonization, and sometimes, invasion. Phages have been in phagocyte evasion, including a recently discovered chemo-
shown to be involved in each of these steps in different bacte- taxis inhibitory protein (CHIPS) that binds to and attenuates
rial pathogens. Phage-mediated adhesion is illustrated by the activity of the neutrophil receptors for complement and
Streptococcus mitis, a causative agent of infective endocarditis. formylated peptides (93; W. Van Wamel, A. Peshel, K. Van
Blood-borne streptococci adhere to platelets, fibrin, and ex- Kessel, and J. Van Strijp, Abstr. 101st Gen. Meet. Am. Soc.
posed valvular matrix components, giving rise to the vegeta- Microbiol. 2001, abstr. B-98, p. 62, 2001). This function is
tions that damage heart valves and provide a source of bacte- proposed to protect S. aureus from neutrophil-mediated killing
remia and septic emboli. Recently, Bensing et al. (7) used (93; Van Wamel et al., Abstr. 101st Gen. Meet. Am. Soc.
transposon mutagenesis to identify bacterial loci involved in Microbiol.), an important host defense against staphylococci.
platelet adherence. They identified a locus encoding two sur- The gene encoding CHIPS (chp) has been shown to reside on
face proteins, PblA and PblB, the gene sequences of which a functional phage that also transduces the staphylokinase
resembled phage capsid and tail fiber genes and were sur- (sak) and enterotoxin A (sea) genes and eliminates ␤-hemoly-
rounded by other phage-like gene sequences (7). These genes sin production, presumably by insertional inactivation (Van
reside on an inducible prophage, SM1 (8), and encode proteins Wamel et al., Abstr. 101st Gen. Meet. Am. Soc. Microbiol.
present in the SM1 phage particle (8). Disruption of either 2001). Staphylococci also produce the phage-encoded Panton-
pblA or pblB resulted in decreased platelet binding by S. mitis, Valentine leukocidin (PVL), a cytotoxin with direct activity
although polar effects on other loci were not strictly ruled out against human phagocytes (48, 92). Thus, by inhibiting phago-
(7). Whether and by what mechanisms the phage-particle-as- cytosis (CHIPS) and by directly attacking phagocytes (PVL),
sociated and/or the bacterial-membrane-bound fractions of two different phage gene products counteract phagocyte-me-
PblA and PblB contribute directly to vegetation formation by diated destruction of their staphylococcal hosts.
S. mitis remain unknown. Inside phagocytes, bacteria are subjected to oxidative stress
Bacterial invasion of human tissues involves the elaboration within organelles that produce superoxide radical and hydro-
of bacterial enzymes, including collagenases, hyaluronidases, gen peroxide. Bacterial survival in the presence of these mol-
and hemolysins. The group A streptococcal (GAS) hyaluroni- ecules is enhanced by enzymes such as superoxide dismutase,
dase is phage encoded (43), and hyaluronidase activity is as- which catalyzes the conversion of superoxide ion into hydrogen
sociated with phage particles themselves (5, 6). Since the cap- peroxide and molecular oxygen (21, 23). Figueroa-Bossi and
VOL. 70, 2002 MINIREVIEW 3987

TABLE 1. Bacterial virulence properties altered by bacteriophages


Bacterial property altered Mechanism Reference(s) or Source

Colonization/adhesion
E. coli The ␭-encoded lom gene promotes adhesion to buccal epithelial cells. 4, 69, 72
P. aeruginosa Phage FIZ15 promotes adhesion to buccal epithelial cells. 91
S. mitis The SM1-encoded PblA and PblB surface proteins promote adhesion to 7, 8
platelets.
V. cholerae The toxin-coregulated pilus may be phage encoded. 49

Invasion
S. enterica Phage SopE␾ transduces a type III secretion system effector that promotes 62
entry into epithelial cells.
Phage Gifsy-1 encodes gipA, a gene that enhances survival in the Peyer’s patch. 85
S. pyogenes Hyaluronidase is phage encoded. 43
S. aureus Fibrinolysin is phage encoded. 77

Resistance to serum/phagocytes

Downloaded from http://iai.asm.org/ on August 12, 2014 by guest


E. coli The ␭-encoded bor gene confers a survival advantage in animal serum. 4
P. aeruginosa Phages encode enzymes that alter the O antigen. 37
S. enterica Phage Gifsy-2 encodes SodC, a superoxide dismutase. 24
Phages encode enzymes that alter the O antigen. 74, 75, 106
S. dysenteriae Phages encode enzymes that alter the O antigen. 31, 58
S. aureus Phages encode CHIPS, a phagocytotoxin. Van Wamel et al.a
Phage ␾PVL encodes the Panton-Valentine leukocidin. 46
S. pyogenes Lysogeny up-regulates the antiphagocytic M protein. 84

Exotoxin production
B. avium Pertussis toxin is phage encoded in B. avium. van Horne et al.b
C. botulinum Botulinum toxin is phage encoded. 29
C. diphtheriae Diphtheria toxin is phage encoded. 38, 90
E. coli The Shiga toxins are phage encoded. 39, 66, 103
P. aeruginosa Pseudomonas cytotoxins are phage encoded. 35
S. dysenteriae The Shiga toxin genes are associated with phage sequences, probably a defective 59
prophage.
S. aureus Staphylococcal enterotoxins are phage encoded. Staphylococcal exfoliative toxins 9, 17, 18
are phage encoded. Toxic shock syndrome toxin is encoded by SapI, a mobile
pathogenicity island transduced at high frequency by phage 80␣.
107
55
S. pyogenes Streptococcal pyrogenic (erythrogenic, scarlatinal) exotoxins are phage encoded. 30, 44, 102
V. cholerae Cholera toxin is phage encoded. 100

Susceptibility to antibiotic
S. aureus Generalized transduction contributes to horizontal transmission of gram-positive 15, 26
antibiotic-resistance genes.
S. pyogenes

Transmission
V. cholerae Phage-encoded cholera toxin likely promotes transmission by stimulating 100
copious amounts of watery diarrhea.
a
Van Wamel et al., Abstr. 101st Gen. Meet. Am. Soc. Microbiol. 2001.
b
S. J. van Horne, D. Bjornsen, P. Carpentier, and L. M. Temple, Abstr. 101st Gen. Meet. Am. Soc. Microbiol. 2001, abstr. B-109, p. 64–65, 2001.

Bossi showed that a superoxide dismutase (SodC) of S. enterica work by a variety of mechanisms that have been the subject of
serovar Typhimurium is encoded by a functional bacterio- a previous review of the involvement of phages in bacterial
phage, Gifsy-2; isogenic Salmonella derivatives lacking Gifsy-2 pathogenesis (10).
were attenuated in a mouse infection model (24). Intriguingly, Phages alter bacterial susceptibility to antibiotics. Most mo-
hydrogen peroxide is a highly effective chemical inducer of bile antibiotic resistance genes are encoded on plasmids or
Gifsy-2, suggesting a possible relationship between SodC ac- transposons, and no examples of phage-encoded resistance
tivity, which results in hydrogen peroxide production, and in- genes are known. However, phages may play an important role,
duction of the Gifsy-2 prophage (24). via transduction, in the mobility of these resistance plasmids
Phages encode bacterial exotoxins. The most widely recog- among staphylococci (26) and streptococci (15). Streptococcal
nized examples of phage-encoded virulence factors are exotox- phages have transferred resistance to tetracycline, chloram-
ins. Exotoxin production is the major pathogenic mechanism of phenicol, macrolides, lincomycin, and clindamycin (89) and
several bacterial pathogens, including V. cholerae, C. diphthe- streptomycin (42), probably via generalized transduction of
riae, and Clostridium botulinum. Phage-encoded exotoxins non-phage-encoded resistance genes.
3988 MINIREVIEW INFECT. IMMUN.

Phage-encoded products enhance transmission of bacterial phase in culture (67). Since these examples of phage-encoded
pathogens. Cholera toxin up-regulates enterocyte adenylate virulence factors are coordinately regulated with other genes
cyclase activity, leading to profuse watery diarrhea (25) that is not encoded on phages and not solely involved in pathogenesis
widely assumed to contribute significantly to the fecal-oral per se, they are consequently not thought to be regulated in
transmission of V. cholerae (61). Since the cholera toxin genes concert with other phage-encoded genes.
are phage encoded (100), this system is an example of a bac- In contrast to these examples in which phages encode
teriophage contributing to the transmission of its bacterial host virulence factors but are thought to play little role in their
among humans. regulation, our recent work on the Shiga toxin-encoding
phages of E. coli has established the principle that the bac-
teriophage life cycle can exert control over virulence factor
REGULATION OF PHAGE-ENCODED
production by bacterial pathogens. The Shiga toxins (Stx1
VIRULENCE FACTORS
and Stx2) are the principal virulence factors of enterohem-
While phages encode virulence genes that influence virtually orrhagic E. coli (EHEC), accounting for the most-severe
all aspects of bacterial pathogenesis, phages have been thought consequences of EHEC infection, including hemorrhagic
to have little role in regulating the expression of these genes. colitis and hemolytic uremic syndrome (11, 68, 95). The stx

Downloaded from http://iai.asm.org/ on August 12, 2014 by guest


Three main observations account for the predominance of this genes in EHEC strains are located within prophages related
idea. First, virulence factors are often produced during lysog- to ␭. Like ␭, these prophages contain transcriptional units
eny, when most phage-encoded genes are not efficiently tran- for various functions, such as replication, morphogenesis,
scribed. Some virulence genes, including the ␭-encoded bor of lysis, etc., that are coordinately expressed during specific
E. coli and the phage-encoded vir of Mycoplasma arthritidis intervals following prophage induction due to the regulatory
(94), lie on the noncoding strand relative to lytic phage genes, influence of phage promoters, phage repressor, transcrip-
suggesting that phage transcriptional regulation has little, if tional terminators, and transcriptional antiterminators (Fig.
any, influence over virulence factor production. 1). The stx genes were found within the late operons of the
Second, many phage-encoded virulence genes are located Stx-encoding phages, suggesting that they could be tran-
near the attachment (att) sites of their respective phages, the scribed from the late phage promoter (pR⬘) along with lysis
portion of the integrated phage adjacent to the host chromo- and morphogenesis genes following prophage induction
some. This finding suggests that they were acquired from the (Fig. 1) (64, 71). Indeed, phage-inducing agents such as
ancestral bacterial chromosome via aberrant phage excision mitomycin C have been shown to increase Stx production by
events, perhaps as independently regulated operons. This E. coli (2, 41; A. E. Hull, D. W. K. Acheson, A. Donohue-
property is true of all of the gram-positive exotoxin-encoding Rolfe, G. T. Keusch, and P. Echeverria, Abstr. 91st Gen.
bacteriophages in which the att site is known, including diph- Meet. Am. Soc. Microbiol. 1991, abstr., p. 390, 1991), and
theria toxin (52), staphylococcal enterotoxin A (18), staphy- subsequently, it was demonstrated that overexpression of Q,
lokinase (18), PVL (47), staphylococcal exfoliative toxin (107), a gene that encodes an antiterminator necessary for efficient
CHIPS (Van Wamel et al., Abstr. 101st Gen. Meet. Am. Soc. late gene expression in ␭-like phages, resulted in increased
Microbiol. 2001), streptococcal erythrogenic (scarlatinal) toxin Stx production by E. coli strains harboring Stx-encoding
(45), and streptococcal pyrogenic exotoxin C (30). Interest- phages (65).
ingly, many phage-encoded virulence genes in gram-negative We characterized the mechanisms by which prophage in-
organisms are not located near the att site, including the Shiga duction augments Stx production by constructing a mutant
toxin genes of E. coli (64, 71), the cytotoxin gene of Pseudo- derivative of the Stx2-encoding phage ␾361 in which the pR⬘
monas aeruginosa (35), and the SodC (24) and SopE (62) genes promoter and a portion of the Q antiterminator gene nec-
of Salmonella. essary for efficient transcription from pR⬘ were deleted. A
Third, many phage-encoded virulence genes are regulated lysogen of the mutant phage was drastically impaired in Stx2
by chromosome-encoded transcription factors. For example, production both in vitro and in a mouse model of intestinal
the iron-dependent transcriptional repressor DtxR, encoded infection (98), implying that the primary regulatory element
on the C. diphtheriae chromosome, represses diphtheria toxin in Stx2 production is the late phage promoter pR⬘ and that
production by binding to an operator overlapping the toxin Stx2 production relies ultimately upon prophage induction.
gene promoter (12, 81, 83). Since DtxR is known to have The previously characterized pStx2 promoter adjacent to stx2
numerous chromosomal target genes involved in iron acquis- (87) is apparently not a dominant regulatory element in Stx2
tion (54, 78–80, 82), diphtheria toxin production is apparently production.
regulated as part of a global cellular response to environmental We also studied the Stx1-encoding phage H-19B. Like the
stimuli. Similarly, the cholera toxin genes (ctx) are up-regu- stx2 genes in ␾361, the stx1 genes in H-19B are located between
lated by the V. cholerae virulence transcriptional activators the late promoter pR⬘ and the lysis genes. However, the pR⬘
ToxR and ToxT (22, 60). While toxR appears to be a compo- promoter is not necessary for Stx1 production by lysogens of
nent of the ancestral V. cholerae genome, toxT is encoded on H-19B (97). Instead, multiple promoters contribute to stx1
VPI (36). Thus, the CTX␾-encoded ctx genes are regulated transcription, including the adjacent iron-regulated promoter
both by a chromosomal locus (toxR) and by a locus encoded on pStx1 (14), as well as upstream phage promoters (pR and pR⬘)
another presumedly mobile genetic element (toxT). Likewise, that are regulated by prophage induction. Other phage-related
phage-encoded staphylococcal toxins are regulated by the mechanisms besides control of stx1 transcription contribute to
chromosomal accessory gene regulatory locus (agr), which co- Stx1 production following H-19B induction. Replication of the
ordinates gene expression during the post-exponential growth H-19B genome, with the associated increase in stx1 copy num-
VOL. 70, 2002 MINIREVIEW 3989

Downloaded from http://iai.asm.org/ on August 12, 2014 by guest


FIG. 1. Phage control of Stx production and release by E. coli. (Top) A partial map (not to scale) of an integrated Stx-encoding prophage is
shown. Transcription from phage promoters is limited both by direct phage repressor activity at pL and pR and by transcriptional terminators
(downstream of pL, pR, and pR⬘). Following prophage induction, repressor cleavage allows for increased transcription from pL and pR and
production of antiterminators N and Q. These proteins facilitate transcription initiated at pR and pR⬘, respectively, by allowing readthrough at
several terminators, including tL, tR1, and tR⬘. (Bottom) Both the pR and pR⬘ promoters, as well as the toxin-associated promoter (pStx) can
contribute directly to stx transcription following prophage induction. Additionally, the phage replication gene products (O and P) contribute to Stx
production by amplifying stx copy numbers. The phage lysis gene products set a limit on the duration of Stx production and provide a mechanism
of toxin release from the cell (97).

ber, was the most quantitatively important mechanism of in- ROLE OF IN SITU PROPHAGE INDUCTION IN
creased Stx1 production. Phage-mediated lysis regulates the BACTERIAL VIRULENCE
quantity of Stx1 produced by limiting the duration of Stx1
accumulation following prophage induction and provides a Although prophage induction has been shown to contribute
mechanism for toxin release (97). In summary, by amplifying to the production of several virulence factors in vitro, we have
stx copy number, by contributing to stx transcription, and by little knowledge regarding the contribution of the phage life
allowing for Stx release, the Stx-encoding phages are intimately cycle to the regulation of virulence factor production during
involved in the regulation of Stx production and thereby the infection. It is conceivable that certain bacterial pathogens
pathogenicity of EHEC. exist in a less virulent state until they encounter a stimulus for
The Stx-encoding prophages are not the only prophages prophage induction within the human body, at which time
implicated in the regulation of virulence factor production. production of a virulence factor, regulated as part of the phage
Some 40 years ago, Barksdale et al. discovered that UV light, life cycle, could contribute to the pathogenesis of the bacte-
a potent prophage-inducing agent, greatly enhanced diphthe- rium. We propose that this in situ prophage induction could
ria toxin production, suggesting that prophage induction and help to explain when and where certain virulence factors are
replication could amplify toxin production (3). Similarly, UV produced during the course of bacterial infection. Moreover,
light enhances phage and streptococcal pyrogenic exotoxin A virion production and release within the human body, with
(scarlatinal toxin) production by GAS (108), as well as produc- subsequent phage infection of other resident bacteria, could
tion of the phage-encoded platelet-binding proteins of S. mitis contribute to pathogenesis by amplifying the number of viru-
(7, 8). Mitomycin C increases production of CTX␾ virions and lence gene-encoding organisms in the body during infection
cholera toxin by V. cholerae (B. Davis and M. Waldor, unpub- (1).
lished observations), and ToxR-independent cholera toxin pro- What are the stimuli for in situ prophage induction? Many
duction has been detected from the replicative form of CTX␾, prophages are induced by environmental conditions that lead
indicating the possible activity of alternative induction-related to bacterial DNA damage and, in the case of E. coli, activation
phage promoters for ctx transcription (53). Although muta- of RecA, which in turn catalyzes cleavage of phage repressors
tional analyses have not been carried out in these cases, the (56). Such conditions include exposure to agents produced by
results are highly suggestive that, as for Stx1 and Stx2, the human cells, including the reactive oxygen species generated
production of other virulence factors may be controlled, in and released by leukocytes, and to exogenous agents, such as
part, by prophage induction. antibiotics. Many antibiotics commonly used to treat diarrhea,
3990 MINIREVIEW INFECT. IMMUN.

for example, are known to induce Stx-encoding phages and Thus, enhanced mobility and persistence in the environment
therefore to promote toxin production by EHEC (51, 57, 101, may provide a selective advantage to virulence genes that are
109). Perhaps not coincidentally, numerous epidemiological phage encoded.
studies have detected an association between increased sever- This relationship may be of benefit to phages as well. For
ity of EHEC infection and treatment with antibiotics (13, 16, example, the hyaluronidase encoded by and incorporated into the
50, 70, 104). This observation has obvious clinical ramifica- particles of S. aureus phages may facilitate phage penetration of
tions, in that antibiotics may actually worsen the clinical course the S. aureus capsule during phage infection or release. Similarly,
of infection by bacteria such as EHEC. O-antigen alteration by phages of certain gram-negative patho-
Phage-inducing agents derived from human cells have been gens, which may aid in bacterial evasion of immunity, presum-
implicated in the pathogenesis of at least three organisms. We ably benefits the phage by conferring superinfection resistance
showed in EHEC that H2O2 (a known inducer of ␭-like on the bacterial host. Moreover, in some cases, virulence fac-
phages) and neutrophils, an endogenous source of H2O2, can tors may actually be integral components of the phage particle.
induce Stx-encoding prophages, which enhances toxin produc- The CTX␾-encoded Ace protein, reported to account in part
tion by EHEC cells (96). The superoxide dismutase (SodC)- for V. cholerae enterotoxicity (88), is thought to be a minor coat
encoding Salmonella phage Gifsy-2 (described above) is also protein of the CTX␾ virion (100). Thus, these putative viru-

Downloaded from http://iai.asm.org/ on August 12, 2014 by guest


induced by H2O2 (24). H2O2 encountered by Salmonella in the lence factors may ultimately be phage attributes that inciden-
phagosome could result in prophage induction and subsequent tally contribute to the pathogenicity of the bacterial host.
up-regulation of SodC production. In this way, prophage in- Other virulence factors may bestow upon phages unknown
duction could serve as an adaptive response by coupling pro- advantages, a possibility that merits future investigation.
duction of the protective SodC enzyme with exposure to reac- One paradoxical implication of in situ prophage induction in
tive oxygen species. In a third example, Broudy et al. bacterial pathogenesis is that virulence factor production could
discovered a soluble phage-inducing factor (SPIF), elaborated be linked to the death of the bacterial cell via phage-mediated
by human pharyngeal epithelial cells, which induced the strep- lysis. This consideration would not apply to pathogens such as
tococcal pyrogenic exotoxin C (SpeC)-encoding phage of GAS V. cholerae, since production of the filamentous CTX␾ does
and subsequent toxin production by this pathogen (12a). Al- not destroy its host bacterial cell. In fact, CTX␾ and cholera
though the SPIF molecule and its mechanism of prophage toxin share a mechanism of nonlethal secretion from the cell
induction remain to be characterized, these observations may (20). In contrast, we have learned that in E. coli lysogens of
reflect an important role for an epithelium-derived prophage- phage ␾361, Stx2 is apparently produced only when the late
inducing factor in the pathogenesis of GAS. phage promoter, responsible for lysis gene transcription, is
We believe that the examples described here demonstrate active (98). This finding implies that toxin production involves
the need for a systematic evaluation of the extent to which the simultaneous and obligatory phage-mediated cell death. One
induction of prophages influences expression of the virulence rationale for the tight linkage between Stx2 production and
factors that they encode. For example, consider the pathogen- phage induction is that production of Stx2 by a small number
esis of endocarditis caused by S. mitis. It is important to learn of doomed cells contributes in some way to the survival of the
whether the organisms encounter prophage-inducing agents in remaining lysogenic population whose prophages were not in-
the bloodstream or in the endocardium that lead to increased duced (98). Alternatively, if the unit of selection is ␾361 or the
expression of the phage-encoded platelet-binding factors. Such stx2 genes themselves, death of the host bacterium could be
information could have important clinical implications in the irrelevant.
treatment of bacterial diseases. For instance, patients suffering In summary, bacteriophages are intimately involved in bac-
from or at risk for diseases in which prophage induction plays terial pathogenesis, including adhesion and invasion, evasion
a pathogenic role might benefit from anti-inflammatory agents of immunity, and exotoxin production, largely by transducing a
that reduce the generation of prophage-inducing antibacterial diverse set of virulence genes. In some cases, the expression of
factors by human cells and from avoiding the use of prophage- these genes may be driven in part by induction of the proph-
inducing antibiotics. ages that encode them. Since prophage induction contributes
to virulence gene expression and since the human body is
EVOLUTIONARY CONSIDERATIONS replete with prophage-inducing chemicals, prophage induction
AND CONCLUSIONS in the human body may be an important, underrecognized
mechanism of bacterial pathogenesis.
Bacteriophages are intimately associated with bacterial vir-
ulence genes. What factors account for this association? This
relationship likely reflects the mobility that virulence genes ACKNOWLEDGMENTS
derive from being phage encoded, allowing for their wide-
We thank our collaborators David Friedman, Melody Neely, and
spread dissemination among bacterial populations. Further-
Jonathan Livny for educating us about lambdoid prophage induction
more, virulence genes encoded by phages may withstand envi- and David Acheson and Xiaoping Zhang for their insights into the
ronmental exposure better than those encoded by bacteria. For epidemiology and pathogenesis of EHEC. We also thank Brigid Davis
example, Muniesa et al. found that Stx2-encoding phages per- and Anne Kane for critical reading of the manuscript.
sisted in river water longer and were more resistant to chlori- P.L.W. was supported by the Harvard Medical School and the
Howard Hughes Medical Institute. M K.W.’s laboratory is supported
nation and pasteurization than Stx2-encoding bacteria, sug- by the National Institute of Allergy and Infectious Diseases, the
gesting that phage particles serve as a more durable Howard Hughes Medical Center, the Pew Foundation, and the New
environmental reservoir of the stx2 genes than bacteria (63). England Medical Center GRASP Center.
VOL. 70, 2002 MINIREVIEW 3991

REFERENCES spective, p. 155–187. In D. Barua and W. B. Greenough III (ed.), Cholera.


Plenum Press, New York, N.Y.
1. Acheson, D. W., J. Reidl, X. Zhang, G. T. Keusch, J. J. Mekalanos, and
26. Firth, N., and R. A. Skurray. 2000. Genetics: accessory elements and ge-
M. K. Waldor. 1998. In vivo transduction with Shiga toxin 1-encoding
netic exchange, p. 326–338. In V. A. Fischetti, R. P. Novick, J. J. Ferretti,
phage. Infect. Immun. 66:4496–4498.
D. A. Portnoy, and J. I. Rood (ed.), Gram-positive pathogens. ASM Press,
2. Al-Jumaili, I., D. A. Burke, S. M. Scotland, H. M. Al-Mardini, and C. O.
Washington, D.C.
Record. 1992. A method of enhancing verocytotoxin production by Esche-
27. Freeman, V. J. 1951. Studies on the virulence of bacteriophage-infected
richia coli. FEMS. Microbiol. Lett. 93:121–126.
strains of Corynebacterium diphtheriae. J. Bacteriol. 61:675–688.
3. Barksdale, L., L. Garmise, and K. Horibata. 1960. Virulence, toxinogeny
28. Frobisher, M., and J. Brown. 1927. Transmissible toxicogenicity of strep-
and lysogeny in Corynebacterium diphtheriae. Ann. N. Y. Acad. Sci. 88:
tococci. Bull. Johns Hopkins Hosp. 41:167–173.
1093–1098.
29. Fujii, N., K. Oguma, N. Yokosawa, K. Kimura, and K. Tsuzuki. 1988.
4. Barondess, J. J., and J. Beckwith. 1990. A bacterial virulence determinant Characterization of bacteriophage nucleic acids obtained from Clostridium
encoded by lysogenic coliphage ␭. Nature 346:871–874. botulinum types C and D. Appl. Environ. Microbiol. 54:69–73.
5. Benchetrit, L. C., E. D. Gray, R. D. Edstrom, and L. W. Wannamaker. 1978. 30. Goshorn, S. C., and P. M. Schlievert. 1989. Bacteriophage association of
Purification and characterization of a hyaluronidase associated with a tem- streptococcal pyrogenic exotoxin type C. J. Bacteriol. 171:3068–3073.
perate bacteriophage of group A, type 49 streptococci. J. Bacteriol. 134: 31. Guan, S., D. A. Bastin, and N. K. Verma. 1999. Functional analysis of the
221–228. O-antigen glucosylation gene cluster of Shigella flexneri bacteriophage SfX.
6. Benchetrit, L. C., E. D. Gray, and L. W. Wannamaker. 1977. Hyaluronidase Microbiology 145:1263–1273.
activity of bacteriophages of group A streptococci. Infect. Immun. 15:527– 32. Halperin, S. A., P. Ferrieri, E. D. Gray, E. L. Kaplan, and L. W. Wanna-
532. maker. 1987. Antibody response to bacteriophage hyaluronidase in acute
7. Bensing, B. A., C. E. Rubens, and P. M. Sullam. 2001. Genetic loci of glomerulonephritis after group A streptococcal infection. J. Infect. Dis.

Downloaded from http://iai.asm.org/ on August 12, 2014 by guest


Streptococcus mitis that mediate binding to human platelets. Infect. Immun. 155:253–261.
69:1373–1380. 33. Hardt, W. D., L. M. Chen, K. E. Schuebel, X. R. Bustelo, and J. E. Galan.
8. Bensing, B. A., I. R. Siboo, and P. M. Sullam. 2001. Proteins PblA and PblB 1998. S. typhimurium encodes an activator of Rho GTPases that induces
of Streptococcus mitis, which promote binding to human platelets, are en- membrane ruffling and nuclear responses in host cells. Cell 93:815–826.
coded within a lysogenic bacteriophage. Infect. Immun. 69:6186–6192. 34. Hardt, W. D., H. Urlaub, and J. E. Galan. 1998. A substrate of the centi-
9. Betley, M. J., and J. J. Mekalanos. 1985. Staphylococcal enterotoxin A is some 63 type III protein secretion system of Salmonella typhimurium is
encoded by phage. Science 229:185–187. encoded by a cryptic bacteriophage. Proc. Natl. Acad. Sci. USA 95:2574–
10. Bishai, W. R., and J. R. Murphy. 1988. Bacteriophage gene products that 2579.
cause human disease, p. 683–724. In R. Calendar (ed.), The bacteriophages. 35. Hayashi, T., T. Baba, H. Matsumoto, and Y. Terawaki. 1990. Phage-con-
Plenum Press, New York, N.Y. version of cytotoxin production in Pseudomonas aeruginosa. Mol. Microbiol.
11. Boerlin, P., S. A. McEwen, F. Boerlin-Petzold, J. B. Wilson, R. P. Johnson, 4:1703–1709.
and C. L. Gyles. 1999. Associations between virulence factors of Shiga 36. Heidelberg, J. F., J. A. Eisen, W. C. Nelson, R. A. Clayton, M. L. Gwinn,
toxin-producing Escherichia coli and disease in humans. J. Clin. Microbiol. R. J. Dodson, D. H. Haft, E. K. Hickey, J. D. Peterson, L. Umayam, S. R.
37:497–503. Gill, K. E. Nelson, T. D. Read, H. Tettelin, D. Richardson, M. D. Ermo-
12. Boyd, J., M. N. Oza, and J. R. Murphy. 1990. Molecular cloning and DNA laeva, J. Vamathevan, S. Bass, H. Qin, I. Dragoi, P. Sellers, L. McDonald,
sequence analysis of a diphtheria tox iron-dependent regulatory element T. Utterback, R. D. Fleishmann, W. C. Nierman, and O. White. 2000. DNA
(dtxR) from Corynebacterium diphtheriae. Proc. Natl. Acad. Sci. USA 87: sequence of both chromosomes of the cholera pathogen Vibrio cholerae.
5968–5972. Nature 406:477–483.
12a.Broudy, T. B., V. Pancholi, and V. A. Fischetti. 2001. Induction of lysogenic 37. Holloway, B. W., and G. N. Cooper. 1962. Lysogenic conversion in Pseudo-
bacteriophage and phage-associated toxin from group A streptococci dur- monas aeruginosa J. Bacteriol. 84:1321–1324.
ing coculture with human pharyngeal cells. Infect. Immun. 69:1440–1443. 38. Holmes, R. K., and L. Barksdale. 1969. Genetic analysis of tox⫹ and tox-
13. Butler, T., M. R. Islam, M. A. K. Azad, and P. K. Jones. 1987. Risk factors bacteriophages of Corynebacterium diphtheriae. J. Virol. 3:586–598.
for development of hemolytic uremic syndrome during shigellosis. J. Pedi- 39. Huang, A., S. De Grandis, J. Friesen, M. Karmali, M. Petric, R. Congi, and
atr. 110:894–897. J. L. Brunton. 1986. Cloning and expression of the genes specifying Shiga-
14. Calderwood, S. B., and J. J. Mekalanos. 1987. Iron regulation of Shiga-like like toxin production in Escherichia coli H19. J. Bacteriol. 166:375–379.
toxin expression in Escherichia coli is mediated by the fur locus. J. Bacteriol. 40. Hueck, C. J. 1998. Type III protein secretion systems in bacterial pathogens
169:4759–4764. of animals and plants. Microbiol. Mol. Biol. Rev. 62:379–433.
15. Caparon, M. 2000. Genetics of group A streptococci, p. 53–64. In V. A. 41. Hull, A. E., D. W. K. Acheson, P. Echeverria, A. Donohue-Rolfe, and G. T.
Fischetti, R. P. Novick, J. J. Ferretti, D. A. Portnoy, and J. I. Rood (ed.), Keusch. 1993. Mitomycin immunoblot colony assay for detection of Shiga
Gram-positive pathogens. ASM Press, Washington, D.C. like toxin-producing Escherichia coli in fecal samples: comparison with
16. Carter, A. O., A. A. Borczyk, J. A. Carlson, B. Harvey, J. C. Hockin, M. A. DNA probes. J. Clin. Microbiol. 31:1167–1172.
Karmali, C. Krishnan, D. A. Korn, and H. Lior. 1987. A severe outbreak of 42. Hyder, S. L., and M. M. Streitfeld. 1978. Transfer of erythromycin resis-
Escherichia coli O157:H7-associated hemorrhagic colitis in a nursing home. tance from clinically isolated lysogenic strains of Streptococcus pyogenes via
N. Engl. J. Med. 317:1496–1500. their endogenous phage. J. Infect. Dis. 138: 281–286.
17. Casman, E. P. 1965. Staphylococcal enterotoxin. Ann. N. Y. Acad. Sci. 43. Hynes, W. L., and J. J. Ferretti. 1989. Sequence analysis and expression in
128:124–131. Escherichia coli of the hyaluronidase gene of Streptococcus pyogenes bacte-
18. Coleman, D. C., D. J. Sullivan, R. J. Russell, J. P. Arbuthnott, B. F. Carey, riophage H4489A. Infect. Immun. 57:533–539.
and H. M. Pomeroy. 1989. Staphylococcus aureus bacteriophages mediating 44. Johnson, L. P., and P. M. Schlievert. 1984. Group A streptococcal phage
the simultaneous lysogenic conversion of ␤-lysin, staphylokinase and en- T12 carries the structural gene for pyrogenic exotoxin type A. Mol. Gen.
terotoxin A: molecular mechanism of triple conversion. J. Gen. Microbiol. Genet. 194:52–56.
135:1679–1697. 45. Johnson, L. P., M. A. Tomai, and P. M. Schlievert. 1986. Bacteriophage
19. d’Herelle, F. 1930. The bacteriophage and its clinical applications. Charles involvement in group A streptococcal pyrogenic exotoxin A production. J.
C. Thoams, Baltimore, Md. Bacteriol. 166:623–627.
20. Davis, B. M., E. H. Lawson, M. Sandkvist, A. Ali, S. Sozhamannan, and 46. Kaneko, J., K. Muramoto, and Y. Kamio. 1997. Gene of LukF-PV-like
M. K. Waldor. 2000. Convergence of the secretory pathways for cholera component of Panton-Valentine leukocidin in Staphylococcus aureus P83 is
toxin and the filamentous phage, CTX␾. Science 288:333–335. linked with lukM. Biosci. Biotechnol. Biochem. 61:541–544.
21. De Groote, M. A., U. A. Ochsner, M. U. Shiloh, C. Nathan, J. M. McCord, 47. Kaneko, J., T. Kimura, S. Narita, T. Tomita, and Y. Kamio. 1998. Complete
M. C. Dinauer, S. J. Libby, A. Vazquez-Torres, Y. Xu, and F. C. Fang. 1997. nucleotide sequence and molecular characterization of the temperate
Periplasmic superoxide dismutase protects Salmonella from products of staphylococcal bacteriophage ␾PVL carrying Panton-Valentine leukocidin
phagocyte NADPH-oxidase and nitric oxide synthase Proc. Natl. Acad. Sci. genes. Gene 215:57–67.
USA 94:13997–14001. 48. Kaneko, J., T. Kimura, Y. Kawakami, T. Tomita, and Y. Kamio. 1997.
22. DiRita, V. J., C. Parsot, G. Jander, and J. J. Mekalanos. 1991. Regulatory Panton-valentine leukocidin genes in a phage-like particle isolated from
cascade controls virulence in Vibrio cholerae. Proc. Natl. Acad. Sci. USA mitomycin C-treated Staphylococcus aureus V8 (ATCC 49775). Biosci. Bio-
88:5403–5407. technol. Biochem. 61:1960–1962.
23. Farrant, J. L., A. Sansone, J. R. Canvin, M. J. Pallen, P. R. Langford, T. S. 49. Karaolis, D. K. R., S. Somara, D. R. Maneval, J. A. Johnson, and J. B.
Wallis, G. Dougan, and J. S. Kroll. 1997. Bacterial copper- and zinc- Kaper. 1999. A bacteriophage encoding a pathogenicity island, a type-IV
cofactored superoxide dismutase contributes to the pathogenesis of sys- pilus and a phage receptor in cholera bacteria. Nature 399:375–379.
temic salmonellosis. Mol. Microbiol. 25:785–796. 50. Kimmit, P. T., C. R. Harwood, and M. R. Barer. 2000. Toxin gene expres-
24. Figueroa-Bossi, N., and L. Bossi. 1999. Inducible prophages contribute to sion by Shiga toxin-producing Escherichia coli: the role of antibiotics and
Salmonella virulence in mice. Mol. Microbiol. 33:167–176. the bacterial SOS response. Emerg. Infect. Dis. 6:458–465.
25. Finkelstein, R. A. 1992. Cholera enterotoxin (choleragen): a historical per- 51. Kohler, B., H. Karch, and H. Schmidt. 2000. Antibacterials that are used as
3992 MINIREVIEW INFECT. IMMUN.

growth promoters in animal husbandry can affect the release of Shiga-toxin- 77. Sako, T., S. Sawaki, T. Sakurai, S. Ito, Y. Yoshizawa, and I. Kondo. 1983.
2-converting bacteriophages and Shiga toxin 2 from Escherichia coli strains. Cloning and expression of the staphylokinase gene of Staphylococcus aureus
Microbiology 146:1085–1090. in Escherichia coli. Mol. Gen. Genet. 190:271–277.
52. Laird, W., and N. B. Groman. 1976. Prophage map of converting coryne- 78. Schmitt, M. P. 1997. Transcription of the Corynebacterium diphtheriae
bacteriophage beta. J. Virol. 19:208–219. hmuO gene is regulated by iron and heme. Infect. Immun. 65:4634–4641.
53. Lazar, S., and M. K. Waldor. 1998. ToxR-independent expression of chol- 79. Schmitt, M. P. 1997. Utilization of host iron sources by Corynebacterium
era toxin from the replicative form of CTX␾. Infect. Immun. 66:394–397. diphtheriae: identification of a gene whose product is homologous to eu-
54. Lee, J. H., T. Wang, K. Ault, J. Liu, M. P. Schmitt, and R. K. Holmes. 1997. karyotic heme oxygenases and is required for acquisition of iron from heme
Identification and characterization of three new promoter/operators from and hemoglobin. J. Bacteriol. 179:838–845.
Corynebacterium diphtheriae that are regulated by the diphtheria toxin re- 80. Schmitt, M. P., and R. K. Holmes. 1994. Cloning, sequence, and footprint
pressor (DtxR) and iron. Infect. Immun. 65:4273–4280. analysis of two promoter/operators from Corynebacterium diphtheriae that
55. Lindsay, J. A., A. Ruzin, H. F. Ross, N. Kurepina, and R. P. Novick. 1998. are regulated by the diphtheria toxin repressor (DtxR) and iron. J. Bacte-
The gene for toxic shock toxin is carried by a family of mobile pathogenicity riol. 176:1141–1149.
islands in Staphylococcus aureus. Mol. Microbiol. 29:527–543. 81. Schmitt, M. P., and R. K. Holmes. 1991. Iron-dependent regulation of
56. Little, J. W. 1996. The SOS regulatory system, p. 453–479. In E. C. C. Lynn diphtheria toxin and siderophore expression by the cloned Corynebacterium
and A. S. Lynch (ed.), Regulation of gene expression in Escherichia coli. diphtheriae repressor gene dtxR in C. diphtheriae C7 strains. Infect. Immun.
R. G. Landis Co., Georgetown, Tex. 59:1899–1904.
57. Matsushiro, A., K. Sato, H. Miyamoto, T. Yamamura, and T. Honda. 1999. 82. Schmitt, M. P., B. G. Talley, and R. K. Holmes. 1997. Characterization of
Induction of prophages of enterohemorrhagic Escherichia coli O157:H7 lipoprotein IRP1 from Corynebacterium diphtheriae, which is regulated by
with norfloxacin. J. Bacteriol. 181:2257–2260. the diphtheria toxin repressor (DtxR) and iron. Infect. Immun. 65:5364–
58. Mavris, M., P. A. Manning, and R. Morona. 1997. Mechanism of bacterio- 5367.

Downloaded from http://iai.asm.org/ on August 12, 2014 by guest


phage SfII-mediated serotype conversion in Shigella flexneri. Mol. Micro- 83. Schmitt, M. P., E. M. Twiddy, and R. K. Holmes. 1992. Purification and
biol. 26:939–950. characterization of the diphtheria toxin repressor. Proc. Natl. Acad. Sci.
59. McDonough, M. A., and J. R. Butterton. 1999. Spontaneous tandem am- USA 89:7576–7580.
plification and deletion of the Shiga toxin operon in Shigella dysenteriae 1. 84. Spanier, J. G., and P. P. Cleary. 1980. Bacteriophage control of anti-
Mol. Microbiol. 34:1058–1069. phagocytic determinants in group A streptococci. J. Exp Med. 152:1393–
60. Miller, V. L., R. K. Taylor, and J. J. Mekalanos. 1987. Cholera toxin 1406.
transcriptional activator toxR is a transmembrane DNA binding protein. 85. Stanley, R. L., C. D. Ellermeier, and J. M. Slauch. 2000. Tissue-specific
Cell 48:271–279. gene expression identifies a gene in the lysogenic phage Gifsy-1 that affects
61. Mintz, E. D., T. Popovic, and P. A. Blake. 1994. Transmission of Vibrio Salmonella enterica serovar typhimurium survival in Peyer’s patches. J.
cholerae O1, p. 345–356. In I. K. Wachsmuth, P. A. Blake, and O. Olsvik Bacteriol. 182:4406–4413.
(ed.), Vibrio cholerae and cholera: molecular to global perspectives. Amer- 86. Strockbine, N. A., M. P. Jackson, L. M. Sung, R. K. Holmes, and A. D.
ican Society for Microbiology, Washington, D.C. O’Brien. 1988. Cloning and sequencing of the genes for Shiga toxin from
62. Mirold, S., W. Rabsch, M. Rohde, S. Stender, H. Tschape, H. Russmann, E. Shigella dysenteriae type 1. J. Bacteriol. 170:1116–1122.
Igwe, and W. D. Hardt. 1999. Isolation of a temperate bacteriophage en- 87. Sung, L. M., M. P. Jackson, A. D. O’Brien, and R. K. Holmes. 1990.
coding the type III effector protein SopE from an epidemic Salmonella Transcription of the Shiga-like toxin type II and Shiga-like toxin type II
typhimurium strain. Proc. Natl. Acad. Sci. USA 96:9845–9850. variant operons of Escherichia coli J. Bacteriol. 172:6386–6395.
63. Muniesa, M., F. Lucena, and J. Jofre. 1999. Comparative survival of free 88. Trucksis, M., J. E. Galen, J. Michalski, A. Fasano, and J. B. Kaper. 1993.
Shiga toxin 2-encoding phages and Escherichia coli strains outside the gut. Accessory cholera enterotoxin (Ace), the third toxin of a Vibrio cholerae
Appl. Environ. Microbiol. 65:5615–5618. virulence cassette. Proc. Natl. Acad. Sci. USA 90:5267–5271.
64. Neely, M. N., and D. I. Friedman. 1998. Arrangement and functional iden- 89. Ubukata, K., M. Konno, and R. Fujii. 1975. Transduction of drug resistance
tification of genes in the regulatory region of lambdoid phage H-19B, a to tetracycline, chloramphenicol, macrolides, lincomycin and clindamycin
carrier of a Shiga-like toxin. Gene 223:105–113. with phages induced from Streptococcus pyogenes. J. Antibiot. (Tokyo)
65. Neely, M. N., and D. I. Friedman. 1998. Functional and genetic analysis of 28:681–688.
regulatory regions of coliphage H-19B: location of Shiga-like toxin and lysis 90. Uchida, T., D. M. Gill, and A. M. Pappenheimer. 1971. Mutation in the
genes suggest a role for phage functions in toxin release. Mol. Microbiol. structural gene for diphtheria toxin carried by temperate phage ␤. Nature
28:1255–1267. New Biol. 233:8–11.
66. Newland, J. W., N. A. Strockbine, S. F. Miller, A. D. O’Brien, and R. K. 91. Vaca, S., J. Arce, G. Oliver, D. Arenas, and F. Arguello. 1989. FIZ15
Holmes. 1985. Cloning of Shiga-like toxin structural genes from a toxin bacteriophage increases the adhesion of Pseudomonas aeruginosa to human
converting phage of Escherichia coli. Science 230:179–181. buccal epithelial cells. Rev. Lat. Am. Microbiol. 31:1–5.
67. Novick, R. P. 2000. Pathogenicity factors and their regulation, p. 392–407. 92. van der Vijver, J. C., M. van Es-Boon, and M. F. Michel. 1972. Lysogenic
In V. A. Fischetti, R. P. Novick, J. J. Ferretti, D. A. Portnoy, and J. I. Rood conversion in Staphylococcus aureus to leucocidin production. J. Virol.
(ed.), Gram-positive pathogens. ASM Press, Washington, D.C. 10:318–319.
68. Ostroff, S. M., P. I. Tarr, M. A. Neill, J. H. Lewis, N. Hargrett-Bean, and 93. Veldkamp, K. E., H. C. Heezius, J. Verhoef, J. A. van Strijp, and K. P. van
J. M. Kobayashi. 1989. Toxin genotypes and plasmid profiles as determi- Kessel. 2000. Modulation of neutrophil chemokine receptors by Staphylo-
nants of systemic sequelae in Escherichia coli O157:H7 infections. J. Infect. coccus aureus supernate. Infect. Immun. 68:5908–5913.
Dis. 160: 994–998. 94. Voelker, L. L., and K. Dybvig. 1999. Sequence analysis of the Mycoplasma
69. Pacheco, S. V., O. G. Gonzalez, and G. L. P. Contreras. 1997. The lom gene arthritidis bacteriophage MAV1 genome identifies the putative virulence
of bacteriophage ␭ is involved in Escherichia coli K12 adhesion to human factor. Gene 233:101–107.
buccal epithelial cells. FEMS Microbiol. Lett. 156:129–132. 95. Wadolkowski, E. A., L. M. Sung, J. A. Burris, J. E. Samuel, and A. D.
70. Pavia, A. T., C. R. Nichols, D. P. Green, R. V. Tauxe, S. Mottice, K. D. O’Brien. 1990. Acute renal tubular necrosis and death of mice orally in-
Greene, J. G. Wells, R. L. Siegler, E. D. Brewer, D. Hannon, et al. 1990. fected with Escherichia coli strains that produce Shiga-like toxin type II.
Hemolytic-uremic syndrome during an outbreak of Escherichia coli Infect. Immun. 58:3959–3965.
O157:H7 infections in institutions for mentally retarded persons: clinical 96. Wagner, P. L., D. W. K. Acheson, and M. K. Waldor. 2001. Human neu-
and epidemiologic observations. J. Pediatr. 116:544–551. trophils and their products induce Shiga toxin production by enterohem-
71. Plunkett, G., 3rd, D. J. Rose, T. J. Durfee, and F. R. Blattner. 1999. orrhagic Escherichia coli. Infect. Immun. 69:1934–1937.
Sequence of Shiga toxin 2 phage 933W from Escherichia coli O157:H7: 97. Wagner, P. L., J. Livny, M. N. Neely, D. W. K. Acheson, D. I. Friedman, and
Shiga toxin as a phage late-gene product. J. Bacteriol. 181:1767–1778. M. K. Waldor. 2002. Bacteriophage control of Shiga toxin 1 production and
72. Reeve, J. N., and J. E. Shaw. 1979. Lambda encodes an outer membrane release by Escherichia coli. Mol. Microbiol. 44:957–970.
protein: the lom gene. Mol. Gen. Genet. 172:243–248. 98. Wagner, P. L., M. N. Neely, X. Zhang, D. W. Acheson, M. K. Waldor, and
73. Reidl, J., and J. J. Mekalanos. 1995. Characterization of Vibrio cholerae D. I. Friedman. 2001. Role for a phage promoter in Shiga toxin 2 expression
bacteriophage K139 and use of a novel mini-transposon to identify a phage- from a pathogenic Escherichia coli strain. J. Bacteriol. 183:2081–2085.
encoded virulence factor. Mol. Microbiol. 18:685–701. 99. Waldor, M. K. 1998. Bacteriophage biology and bacterial virulence. Trends
74. Robbins, P. W., and T. Uchida. 1965. Chemical and macromolecular struc- Microbiol. 6:295–297.
ture of O antigens from Salmonella anatum strains carrying mutants of 100. Waldor, M. K., and J. J. Mekalanos. 1996. Lysogenic conversion by a
bacteriophage ε34. J. Biol. Chem. 240:375–383. filamentous phage encoding cholera toxin. Science 272:1910–1914.
75. Robbins, P. W., and T. Uchida. 1962. Studies on the chemical basis of the 101. Walterspiel, J. N., S. Ashkenazi, A. L. Morrow, and T. G. Cleary. 1992.
phage conversion of O-antigens in the E-group Salmonellae. Biochemistry Effect of subinhibitory concentrations of antibiotics on extracellular Shiga-
1:323–335. like toxin I. Infection 20:25–29.
76. Ruzin, A., J. A. Lindsay, and R. P. Novick. 2001. Molecular genetics of 102. Weeks, C. R., and J. J. Ferretti. 1984. The gene for type A streptococcal
SaPI1–a mobile pathogenicity island in Staphylococcus aureus. Mol. Micro- exotoxin (erythrogenic toxin) is located in bacteriophage T12. Infect. Im-
biol. 41:365–377. mun. 46:531–536.
VOL. 70, 2002 MINIREVIEW 3993

103. Willshaw, G. A., H. R. Smith, S. M. Scotland, and B. Rowe. 1985. Cloning bacteriophage ε34. J. Bacteriol. 105:927–936.
of genes determining the production of vero cytotoxin by Escherichia coli. 107. Yamaguchi, T., T. Hayashi, H. Takami, K. Nakasone, M. Ohnishi, K.
J. Gen. Microbiol. 131:3047–3053. Nakayama, S. Yamada, H. Komatsuzawa, and M. Sugai. 2000. Phage con-
104. Wong, C. S., S. Jelacic, R. L. Habeeb, S. L. Watkins, and P. I. Tarr. 2000. version of exfoliative toxin A production in Staphylococcus aureus. Mol.
The risk of the hemolytic-uremic syndrome after antibiotic treatment of Microbiol. 38:694–705.
Escherichia coli O157:H7 infections. N. Engl. J. Med. 342:1930–1936. 108. Zabriskie, J. B. 1964. The role of temperate bacteriophage in the production
105. Wood, M. W., R. Rosqvist, P. B. Mullan, M. H. Edwards, and E. E. Galyov. of erythrogenic toxin by group A streptococci. J. Exp. Med. 119:761–779.
1996. SopE, a secreted protein of Salmonella dublin, is translocated into the 109. Zhang, X., A. D. McDaniel, L. E. Wolf, G. T. Keusch, M. K. Waldor, and
target eukaryotic cell via a sip-dependent mechanism and promotes bacte- D. W. Acheson. 2000. Quinolone antibiotics induce Shiga toxin-encoding
rial entry. Mol. Microbiol. 22:327–338. bacteriophages, toxin production, and death in mice. J. Infect. Dis. 181:
106. Wright, A. 1971. Mechanism of conversion of the salmonella O antigen by 664–670.

Editor: D. A. Portnoy

Downloaded from http://iai.asm.org/ on August 12, 2014 by guest


CASE REPORT
published: 08 December 2015
doi: 10.3389/fmicb.2015.01375

Typing Discrepancy Between


Phenotypic and Molecular
Characterization Revealing an
Emerging Biovar 9 Variant of Smooth
Phage-Resistant B. abortus Strain
8416 in China
Edited by: Yao-Xia Kang 1,2 , Xu-Ming Li 1 , Dong-Ri Piao 3 , Guo-Zhong Tian3 , Hai Jiang 3 , En-Hou Jia 3 ,
Yongqun “Oliver” He, Liang Lin 2 , Bu-Yun Cui 3*, Yung-Fu Chang 4* , Xiao-Kui Guo 1* and Yong-Zhang Zhu1,4*
University of Michigan, USA 1
Department of Immunology and Microbiology, Institutes of Medical Sciences, Shanghai Jiao Tong University School of
Reviewed by: Medicine, Shanghai, China, 2 Baotou Municipal Center for Disease Control and Prevention, Baotou, China, 3 State Key
Jianguo Jian Zhu, Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of
Shanghai Jiao Tong University, China Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Beijing, China, 4 Department of
Menachem Banai, Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
Kimron Veterinary Institute, Israel
Jiabo Ding,
China Institute of Veterinary Drugs A newly isolated smooth colony morphology phage-resistant strain 8416 isolated from a
Control, China 45-year-old cattle farm cleaner with clinical features of brucellosis in China was reported.
*Correspondence: The most unusual phenotype was its resistance to two Brucella phages Tbilisi and
Bu-Yun Cui
cuibuyun@icdc.cn; Weybridge, but sensitive to Berkeley 2, a pattern similar to that of Brucella melitensis
Yung-Fu Chang biovar 1. VITEK 2 biochemical identification system found that both strain 8416 and
yc42@cornell.edu;
Xiao-Kui Guo
B. melitensis strains shared positive ILATk, but negative in other B. abortus strains.
microbiology@sjtu.edu.cn; However, routine biochemical and phenotypic characteristics of strain 8416 were most
Yong-Zhang Zhu similar to that of B. abortus biovar 9 except CO2 requirement. In addition, multiple PCR
yzhzhu@hotmail.com
molecular typing assays including AMOS-PCR, B. abortus special PCR (B-ab PCR) and
Specialty section: a novel sub-biovar typing PCR, indicated that strain 8416 may belong to either biovar
This article was submitted to
3b or 9 of B. abortus. Surprisingly, further MLVA typing results showed that strain 8416
Infectious Diseases,
a section of the journal was most closely related to B. abortus biovar 3 in the Brucella MLVA database, primarily
Frontiers in Microbiology differing in 4 out of 16 screened loci. Therefore, due to the unusual discrepancy between
Received: 30 June 2015 phenotypic (biochemical reactions and particular phage lysis profile) and molecular
Accepted: 19 November 2015
Published: 08 December 2015
typing characteristics, strain 8416 could not be exactly classified to any of the existing
Citation:
B. abortus biovars and might be a new variant of B. abortus biovar 9. The present
Kang Y-X, Li X-M, Piao D-R, Tian G-Z, study also indicates that the present phage typing scheme for Brucella sp. is subject to
Jiang H, Jia E-H, Lin L, Cui B-Y,
variation and the routine Brucella biovar typing needs further studies.
Chang Y-F, Guo X-K and Zhu Y-Z
(2015) Typing Discrepancy Between Keywords: B. abortus, smooth phage-resistant (SPR), MLVA typing, unusual biochemical reactions
Phenotypic and Molecular
Characterization Revealing an
Emerging Biovar 9 Variant of Smooth
Brucellosis is one of the most common zoonotic infectious diseases, causing enormous economic
Phage-Resistant B. abortus Strain
8416 in China.
loss in domestic animals and public health problems worldwide (Adone and Pasquali, 2013; Van der
Front. Microbiol. 6:1375. Henst et al., 2013). Transmission from animals to human occurs primarily through direct contact
doi: 10.3389/fmicb.2015.01375 with infected animals and ingestion of raw milk or unpasteurized cheese. On the basis of obviously

Frontiers in Microbiology | www.frontiersin.org 1 December 2015 | Volume 6 | Article 1375


Kang et al. Identification of an Unusual B. abortus SPR Strain

different phenotypic characteristics, host preference, growth of Baotou Municipal Center for Disease Control and Prevention,
and biochemical characteristics including CO2 requirement, the Inner Mongolia Autonomous Region of China. However,
substrate utilization and growth on dyes and agglutination with only one blood sample from a 45-year-old male janitor yielded a
monospecific sera as well as Brucella phage lysis profiles, four positive culture result. The isolated strain 8416 displayed smooth,
main Brucella pathogenic species including Brucella melitensis tiny, white, shiny and translucent colonies on solid agar after
(sheep and goat), B. suis (pigs), B. abortus (cattle), and B. canis 3 days of incubation. The strain 8416 was sub-cultured on blood
(dogs), a taxonomic scheme can be defined and further divided plate with 5% CO2 and displayed typical colonies with small
into multiple biovars. For example, B. abortus is subdivided into Gram-negative coccobacilli. The strain was sent to department
eight biovars (biovar 1–7 and 9) (Van der Henst et al., 2013). of brucellosis, Chinese Communicable Disease Control and
Because of unstable phenotypic characteristics among Brucella Prevention (Chinese CDC) for further analysis and identification.
strains, it is somewhat difficult to define atypical strains into The reference strains including B. abortus biovar 1 to 7 and
standard biovars. For instance, the susceptibility of smooth 9, strains: 544A (ATCC 23448), 86/8/59 (ATCC 23449), Tulya
B. abortus strains to lysis by most of brucella phages, such (ATCC 23450), 292 (ATCC 23451), B3196 (ATCC 23452), 870
as Tbilisi (Tb), Firenze (Fi), Weybridge (Wb), and Berkeley 2 (ATCC 23453), 63/75, and C68 (ATCC 23455), B. melitensis
(BK2 ), is commonly regarded as one of the routine criteria to biovar 1 to 3, strains: 16M (ATCC 23456), 63/9 (ATCC 23457)
differentiate this organism from other Brucella species. However, and Ether (ATCC 23458)), B. suis biovar 1 to 5, strains: 1330S
the majority of B. abortus strains resistant to Brucella phage (ATCC 23444), Thomsen (ATCC 23445), 686 (ATCC 23446), 40
have been currently reported primarily due to variation from (ATCC 23447), and 513, B. neotomae RM6/66 (ATCC 23365),
smooth to rough form during normal in vitro culture. Since the B. ovis 63/290 (ATCC 25840), and B. canis 5K33 (ATCC 23459)
first smooth phage-resistant strain (SPR) of B. abortus isolated were used as controls for phenotype typing, biochemical and/or
from bovine tissue was reported in 1973 (Corbel and Morris, molecular analysis.
1974, 1975), a similar study describing SPR strains has not been
reported yet. In this study, we report a newly isolated SPR Analysis of Phenotypic Characteristics
strain, strain 8416 from a patient with brucellosis in the Inner At first, to exclude mixed cultures of different biovars and phage
Mongolia Autonomous Region of China on 2012. Actually, it carrier state, the strain used in this study was subjected to a single
was the only B. abortus strain among a total of 197 Brucella cloned isolation for successive three times to confirm no variable
strains isolated and authenticated by Chinese CDC during this colonial morphology as described by Jones et al. (1962). The
year. The Inner Mongolia Autonomous Region has the highest strain was further characterized by using the classical Brucella
incidence, responsible for about more than 40% of reported cases phenotypic identification procedures, such as CO2 requirement,
in China (Zhang et al., 2010; Chen et al., 2013). Interestingly, the H2 S production, dye sensitivity by basic fuchsin and thionin,
unique phenotypical characteristics of the B. abortus SPR strain agglutination with monospecific antisera, and phage typing as
8416, determined by routine biotyping for the identification of described by Alton GG (Alton et al., 1975). Brucella monospecific
Brucella species and biovars, did not completely fit into any antisera to A, M, and R (rough) and Brucella phages Tb, Wb,
of the recognized classification biovars, indicating the potential and Bk2 were used according to standard protocol of the Chinese
presence of a new variant of B. abortus biovar 3. CDC (Jiang et al., 2013) to characterize this strain. All of
phenotypic characterizations in this study were repeated at least
three times to make sure the results are repeatable.
MATERIALS AND METHODS
Bacterial Isolation and Used Strains Molecular Typing Identification
Brucella strains were inactivated by suspending one loop from
The protocol for this study was approved by ethics committee of
a solid bacterial culture in 200 µl DNA storage buffer. Total
local disease control and Prevention Research Center of the Inner
genomic DNA was extracted using the DNeasy Blood &
Mongolia Autonomous Region and Baotou Municipal Center
Tissue Kit (Qiagen China Ltd., Beijing, China) following the
for Disease Control and Prevention. In June 2012, two workers
manufacture’s instruction. The PCR assay targeting bcsp31, was
from a cattle farm in Sichuan province, presenting fever, night
performed to confirm the Brucella genus as previously described
sweat and soreness of waist, arthralgia and muscle weakness,
(Bounaadja et al., 2009), and species-level using the routine
were admitted to one local hospital in the Inner Mongolia
Abortus-Melitensis-Ovis-Suis PCR (AMOS-PCR) (Bricker and
Autonomous Region. The serum samples from these two patients
Halling, 1994). Furthermore, B. abortus B-ab PCR and a novel
were strongly positive to Brucella by both Rose-Bengal-plate-
PCR to differentiate B. abortus biovar 3a, 3b, 5, 6, and 9 were
agglutination-test (RBPT) and Serum Agglutination Test (SAT)
performed as previously described (Ocampo-Sosa et al., 2005;
with titers of 1/320 according to standard procedures. Moreover,
Huber et al., 2009).
the two serum samples were also confirmed by positive ELISA
results with Brucella IgG (>150 U/ml) and IgM (>60 U/ml)
(Brucella IgG and IgM ELISA kits, IBL Germany). At the same Multiple Locus Variable Number Tandem
time, the blood culture of the two patients were inoculated in Repeat Analysis (MLVA) Genotyping
a dual-phase coloration blood culture bottle (BioMerieux Inc., Multiple locus variable number tandem repeat analysis (MLVA)
Durham, USA) at 37◦ C for 2–3 weeks at the diagnostic laboratory was performed as previously described by Le Fleche et al. (2006)

Frontiers in Microbiology | www.frontiersin.org 2 December 2015 | Volume 6 | Article 1375


Kang et al. Identification of an Unusual B. abortus SPR Strain

and by Jiang et al. (2013), respectively. The 16 primer pairs

B. abortus biovar 9 variant


comprised three main groups: panel 1 including bruce06, 08, 11,

B. melitensis biovar 1
B. melitensis biovar 3
B. abortus biovar 1
B. abortus biovar 3
B. abortus biovar 6
B. abortus biovar 9
12, 42, 43, 45, and 55 for species identification, panel 2A (bruce18,

B. suis biovar 1
Interpretation 19, and 21), and panel 2B (bruce04, 07, 09, 16, and 30) for further
subspecies differentiation were used.

Biochemical Identification by VITEK 2


bruce30 System
3
5
5
3
3
6
3
3
A total of 47 biochemical reactions of the Brucella strains

11
bruce16
were analyzed using the standard Gram-negative bacteria
3
4

3
2
3
3
5
bruce09 identification card on automatic VITEK 2 system according to
7
3
3
2
2
8
1
5
the manufacturer’s instructions.
TABLE 1 | Comparison of phenotypic characteristics and Brucella phage lysis profiles of Brucella abortus strain 8416 and other Brucella reference strains.

bruce07
6
5
5
2
2
5
1
6
bruce04
6
3
6
3
3
2
3
6
bruce21 RESULTS
8
8
8
8
8
6
8
9
Routine Phenotypic Typing
42
21
20
42
42
18
42
brucel9 19
Brucella MLVA16

bruce18 Characteristics
6
5
8
7
7
5
7
4

bruce55 According to routine phenotypic analysis, strain 8416 was anti-


2
3
3
3
3
3
9
2

R negative and H2 S positive, agglutination with anti-M serum


but not anti-A serum and grew in the presence of thionine and
12

bruce45
12

13
3
3
3

bruce43 fuchsin dyes (Table 1). Moreover, it was not lysed by Tb and
2
2
2
3
3
2
3
1

Wb phages both in 1× RTD (Routine Test Dilution) and 104 ×


bruce42
RTD, but lysed by BK2 phage both in 1× RTD and 102 × RTD
12
2
2
2
3
3
4

bruce12 (Figure 1A). Thus, the particular phenotypic profiles of the strain
12
12
11

13

10
5
5

8416 were more similar to that of the classic characteristics of


bruce11
B. abortus biovars 9.
4
4
4
6
6
2
5
6

bruce08
5
5
5
3
3
4
3
3

bruce06
Biochemical Identification of Automatic
4
4
3
3
6
3
7
2

VITEK 2 System
Four biochemical indicators ProA (L-pyrrolydonyl-arylamidase),
BK2
Mono specific phage at

+
+
+
+
+
+
+
+

TyrA (tyrose arylamidase), URE (urease), and GlyA could be


RTD

used to distinguish Brucella species. All of eight B. abortus


Wb



+
+
+
+

reference strains and 21 field strains were positive in ILATk (L-


Tb

lactate alkalization), but it was negative in strain 8416, three





+
+
+
+

B. melitensis reference strains and 92 field strains (Cui BuYun’s


M




+

+
+
+
Sera

unpublished data). This result indicated that strain 8416 showed


special biochemical characteristics distinct from that of B. abortus


A

+
+
+

+
+
+

strains.
Fuschin


+
+
+
+
+
+
+

Molecular Typing Identification


Strain 8416 was identified as B. abortus by the combination of
Thionin

bcsp31 PCR (223-bp, data not shown) and B-ab PCR (370-bp)
Growth characteristics


+

+
+
+
+
+
+

(Figure 1B) but not as biovar 1, 2, and 4 of B. abortus according


to AMOS-PCR (Figure 1C). The novel PCR assay was used to
production

compare strain 8416 to B. abortus biovar 3b, 5, 6, and 9, and


H2 S

found that the PCR product of 1.7 kb from strain 8416 was similar
++



+
+
+

to B. abortus biovar 3b, 5, 6, and 9, but not to other B. abortus


biovars (Figure 1D).
requirement

MLVA Genotyping
±

±






CO2

According to Brucella MLVA typing database (Grissa et al., 2008),


16 loci of MLVA matching results displayed that strain 8416
was closely related to B. abortus biovar 3 (Jiang et al., 2013),
1330S
Strain

but primarily different in four variable loci, bruce04, bruce07,


544A

Ether
8416

Tulya

16M
C68
870

bruce11, and bruce55 (Table 2).

Frontiers in Microbiology | www.frontiersin.org 3 December 2015 | Volume 6 | Article 1375


Kang et al. Identification of an Unusual B. abortus SPR Strain

FIGURE 1 | (A) The lysis patterns of phage Tb, Wb and Bk2 to Brucella abortus strain 8416, B. abortus biovar 1 strain 544A (A is indicated as B. abortus),
B. melitensis biovar 1 strain 16M (M is indicated as B. melitensis), and B. suis biovar 1 strain 1330S (S is indicated as B. suis) as well as B. abortus biovar 6 strain
870 and biovar 9 strain C68; (B) Amplification of DNA fragments from different Brucella strains. Genomic DNA was amplified by the B-ab PCR assay. 1: strain 8416;
2–5: four B. melitensis field strains; 104 M: B. melitensis biovar 1 strain 104M; 544A: B. abortus biovar 1 strain 544A; (C) Amplification of DNA fragments from
different Brucella strains. Genomic DNA was amplified by AMOS-PCR assay. 1: strain 8416; 2–5: four B. melitensis field strains; 104 M: B. melitensis biovar 1 strain
104M; 544A: B. abortus biovar 1 strain 544A; (D) Amplification of DNA fragments from different Brucella strains. Genomic DNA was amplified by new PCR assay
identifying B. abortus biovar 3b, 5, 6, and 9. 1: B. melitensis biovar 1 strain 16M; 2: B. abortus biovar 1 strain 544A; 3: B. suis biovar 1 strain 1330S; 4: B. abortus
biovar 9 strain C68; 5: B. abortus biovar 3a strain Tulya; 6: strain 8416.

Finally, based on these typing results, strain 8416 might be a FS showed no differences in virulence, morphological, cultural,
new variant of B. abortus biovar 9. biochemical or metabolic, and serological reactions, but with an
altered phage resistance profile (Corbel and Morris, 1974). The
potential mechanism of the phage resistance may be due to its
DISCUSSION failure to penetrate the FS cell wall since the strain FS is more
resistant to lysis by phage lysozymes than that of the phage-
Until now, the phage resistance mechanism from Brucella SPR sensitive parent strain 544 (Corbel and Morris, 1975). Strain
strains was poorly understood. In this study, a natural SPR strain 544-FS showed a complete resistance to lysis by many Brucella
of B. abortus isolated from a patient in China was identified. phages except Bk2 at 1× RTD and 104 × RTD. Subsequently,
Although SPR strains of B. abortus were rarely isolated from another B. abortus SPR strain with resistance to phage Tb, was
patients, a SPR strain was isolated from a B. abortus phage isolated from a supramammary lymph node of a cow and it is
sensitive parent strain 544 in 1974 and a SPR variant of B. abortus virulent to guinea-pigs (Harrington et al., 1977). Interestingly,
strain 19 was identified in 1976 through the manipulation these B. abortus SPR strains mentioned above belonging to
of laboratory cultures (Corbel and Morris, 1974; Corbel and B. abortus biovar 1 were identified. However, strain 8416 was
Thomas, 1976). Compared to the parent strain 544, the SPR strain significantly different from all of B. abortus biovars by using

Frontiers in Microbiology | www.frontiersin.org 4 December 2015 | Volume 6 | Article 1375


TABLE 2 | Comparison of Brucella MLVA typing results of B. abortus strain 8416 and the most closely related B. aborus biovar 3 field strains in the Brucella MLVA database.
Kang et al.

Distance
MLVA8
MLVA11
MLVA16
Bruce06- 1322
Bruce08- 1134
Bruce11- 211
Bruce12 - 73
Bruce42-424
Bruce43-379
Bruce45-233
Bruce55-2066
Bruce18-339
Bruce19-324
Bruce21-329
Bruce04-1543
Bruce07-1250
Bruce09-588
Bruce16-548
Bruce30-1505

Strain name BaseView Strain Host Isolated_in Species- Contact Group Year
biovar

8416 8416 Human Inner Mongolia, 2012 4 5 4 12 2 2 3 2 6 42 8 6 6 7 3 3


China
2013Jiang#095 0 Brucella2013 Human Inner Mongolia, B. abortus Buyun Cui 2012 30 4 5 4 12 2 2 3 2 6 42 8 6 6 7 3 3
China biovar 3

Frontiers in Microbiology | www.frontiersin.org


2013Garofolo_ 3 Brucella_ 9263 Bufalo Albanella,Italy B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 5 6 7 3 3
9263 ITALIA_1 biovar3 Garofolo
2013Jiang#108 3 Brucella2013 NM1068 Cattle Inner Mongolia, B. abortus Buyun Cui 1985 36 4 5 3 12 2 2 3 1 6 42 8 6 7 7 3 3
China biovar 3
2013Jiang#105 3 Brucella2013 NM1065 Cattle Inner Mongolia, B. abortus Buyun Cui 1985 117 4 5 3 12 2 2 3 2 6 42 8 6 4 4 3 3
China biovar 3
2013Jiang#131 3 Brucella2013 NM1158 Cattle Inner Mongolia, B. abortus Buyun Cui 1988 36 4 5 3 12 2 2 3 1 6 42 8 6 4 7 3 3
China biovar 3
2013Jiang#140 3 Brucella2013 NM1175 Cattle Inner Mongolia, B. abortus Buyun Cui 1990 36 4 5 3 12 2 2 3 1 6 42 8 6 4 7 3 3
China biovar 3
2013Garofolo_ 3 Brucella_ 3636 Cattle Monte San B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 4 6 7 3 3
3636 ITALIA_1 Giacomo,Italy biovar3 Garofolo

5
2013Garofolo_ 3 Brucella_ 3916 Cattle Monte San B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 4 6 7 3 3
3916 ITALIA_1 Giacomo,Italy biovar3 Garofolo
2013Garofolo_ 3 Brucella_ 3920 Cattle Teggiano,Italy B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 4 6 7 3 3
3920 ITALIA_1 biovar3 Garofolo
2013Garofolo_ 3 Brucella_ 4363 Cattle Laurenzana,Italy B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 4 6 7 3 3
4363 ITALIA_1 biovar3 Garofolo
MLVA16

2013Garofolo_ 3 Brucella_ 12183 Cattle Corleto B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 4 6 7 3 3


12183 ITALIA_1 Monforte,Italy biovar3 Garofolo
2013Garofolo_ 3 Brucella_ 12185 Cattle San Rufo,Italy B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 4 6 7 3 3
12185 ITALIA_1 biovar3 Garofolo
2013Garofolo_ 3 Brucella_ 21571 Cattle Monte San B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 4 6 7 3 3
21571 ITALIA_1 Giacomo,Italy biovar3 Garofolo
2013Garofolo_ 3 Brucella_ 21675 Cattle Monte San B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 4 6 7 3 3
21675 ITALIA_1 Giacomo,Italy biovar3 Garofolo
2013Garofolo_ 3 Brucella_ 22839 Bufalo Albanella,Italy B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 4 6 7 3 3
22839 ITALIA_1 biovar3 Garofolo
2013Garofolo_ 3 Brucella_ 22842 Cattle Teggiano,Italy B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 4 6 7 3 3
22842 ITALIA_1 biovar3 Garofolo
2013Garofolo_ 3 Brucella_ 5362 Bufalo Monte San B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 4 6 7 3 3
5362 ITALIA_1 Giacomo,Italy biovar3 Garofolo
2013Garofolo_ 3 Brucella_ 8980 Cattle Teggiano,Italy B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 4 6 7 3 3
8980 ITALIA_1 biovar3 Garofolo

(Continued)

December 2015 | Volume 6 | Article 1375


Identification of an Unusual B. abortus SPR Strain
TABLE 2 | Continued
Kang et al.

Distance
MLVA8
MLVA11
MLVA16
Bruce06-1322
Bruce08-1134
Bruce11-211
Bruce12-73
Bruce42-424
Bruce43-379
Bruce45-233
Bruce55-2066
Bruce18-339
Bruce19-324
Bruce21-329
Bruce04-1543
Bruce07-1250
Bruce09-588
Bruce16-548
Bruce30-1505

Strain name BaseView Strain Host Isolated_in Species- Contact Group Year
biovar

2013Garofolo_ 3 Brucella_ 8984 Cattle Sassano,Italy B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 4 6 7 3 3


8984 ITALIA_1 biovar3 Garofolo
2013Jiang#093 3 Brucella2013 2011166 Human Chongqing, B. abortus Buyun Cui 2011 36 4 5 3 12 2 2 3 1 6 42 8 4 6 7 3 3
China biovar 3

Frontiers in Microbiology | www.frontiersin.org


2013Jiang#094 3 Brucella2013 YLQ Human Zhejiang, China B. abortus Buyun Cui 2006 36 4 5 3 12 2 2 3 1 6 42 8 4 6 7 3 3
biovar 3
2013Jiang#104 4 Brucella2013 NM1061 Cattle Inner Mongolia, B. abortus Buyun Cui 1984 112 4 5 3 12 2 2 3 3 6 42 8 5 4 7 3 3
China biovar 3
2013Jiang#092 4 Brucella2013 2011165\’ Human Chongqing, B. abortus Buyun Cui 2011 36 4 5 3 12 2 2 3 1 6 42 8 4 7 7 3 3
China biovar 3
2012 4 Brucella2012 LNIV- Alentejo, B. abortus 3 Cristina B. abortus 2006 36 72 4 5 3 12 2 2 3 1 6 42 8 4 7 7 3 3
Ferreira#146 328Ba3-06 Portugal Ferreira
2013Garofolo_ 4 Brucella_ 3272 Cattle Apricena,Italy B. abortus_ Giuliano 2011 4 5 3 12 2 2 3 3 6 42 8 7 6 3 3 3
3272 ITALIA_1 biovar3 Garofolo
2013Garofolo_ 4 Brucella_ 18081 Cattle Apricena,Italy B. abortus_ Giuliano 2011 4 5 3 12 2 2 3 3 6 42 8 7 6 3 3 3

6
18081 ITALIA_1 biovar3 Garofolo
2006 4 Brucella2012 BCCN#99- Cattle Mongolia B. abortus 7 Gilles B. abortus 1999 36 72 4 5 3 12 2 2 3 1 6 42 8 5 6 4 3 3
LeFlèche#119 98 Vergnaud
2013Jiang#089 4 Brucella2013 Cattle Hebei, China B. abortus Buyun Cui 2011 36 4 5 3 12 2 2 3 1 6 42 8 4 5 7 3 3
biovar 3
2013Jiang#090 4 Brucella2013 Cattle Hebei, China B. abortus Buyun Cui 2011 36 4 5 3 12 2 2 3 1 6 42 8 4 5 7 3 3
biovar 3
2006 4 Brucella2012 BCCN#94- Cattle Limoges, B. abortus 3 Gilles B. abortus 1994 36 72 4 5 3 12 2 2 3 1 6 42 8 4 5 7 3 3
LeFlèche#112 18 France Vergnaud
2013Jiang#100 4 Brucella2013 NM1051 Cattle Inner Mongolia, B. abortus Buyun Cui 1984 36 4 5 3 12 2 2 3 1 6 42 8 6 4 8 3 3
China biovar 3
2006 4 Brucella2012 REF 292 Cattle England B. abortus 4 Gilles B. abortus 30 78 4 5 4 12 2 2 3 2 6 42 8 3 4 3 3 5
LeFlèche#005 Vergnaud
2009Her#004 4 Brucella2012 KRef04 Cattle England B. abortus 4 Moon Her B. abortus 30 78 4 5 4 12 2 2 3 2 6 42 8 3 4 3 3 5
2012 4 Brucella2012 REF 292 B. abortus 4 Cristina B. abortus 30 78 4 5 4 12 2 2 3 2 6 42 8 3 4 3 3 5
Ferreira#213 Ferreira
2013Jiang#127 4 Brucella2013 NM1147 Cattle Inner Mongolia, B. abortus Buyun Cui 1988 117 4 5 3 12 2 2 3 2 6 42 8 5 4 3 3 3
China biovar 3
2013Jiang#130 4 Brucella2013 NM1156 Sheep Inner Mongolia, B. abortus Buyun Cui 1988 36 4 5 3 12 2 2 3 1 6 42 8 6 4 4 3 3
China biovar 3
2013Jiang#125 4 Brucella2013 NM1140 Cattle Inner Mongolia, B. abortus Buyun Cui 1988 36 4 5 3 12 2 2 3 1 6 42 8 5 4 7 3 3
China biovar 3

(Continued)

December 2015 | Volume 6 | Article 1375


Identification of an Unusual B. abortus SPR Strain
Kang et al.

TABLE 2 | Continued

Frontiers in Microbiology | www.frontiersin.org


Distance
MLVA8
MLVA11
MLVA16
Br uc e06- 1322
Br uc e08- 1134
Bruce11-211
Bruce12-73
Bruce42-424
Bruce43-379
Bruce45-233
Bruce55-2066
Bruce18-339
Bruce19-324
Bruce21-329
Bruce04-1543
Bruce07-1250
Bruce09-588
Bruce16-548
Bruce30-1505

Strain name BaseView Strain Host Isolated_in Species- Contact Group Year
biovar

2013Jiang#126 4 Brucella2013 NM1146 Cattle Inner Mongolia, B. abortus Buyun Cui 1988 36 4 5 3 12 2 2 3 1 6 42 8 5 4 7 3 3
China biovar 3
2013Jiang#128 4 Brucella2013 NM1148 Cattle Inner Mongolia, B. abortus Buyun Cui 1988 36 4 5 3 12 2 2 3 1 6 42 8 5 4 7 3 3
China biovar 3
2013Jiang#141 4 Brucella2013 NM1176 Cattle Inner Mongolia, B. abortus Buyun Cui 1990 36 4 5 3 12 2 2 3 1 6 42 8 5 4 7 3 3
China biovar 3
2013Jiang#150 4 Brucella2013 NM1215 Cattle Inner Mongolia, B. abortus Buyun Cui 1994 36 4 5 3 12 2 2 3 1 6 42 8 5 4 7 3 3
China biovar 3

7
2013Jiang#151 4 Brucella2013 NM1218 Cattle Inner Mongolia, B. abortus Buyun Cui 1995 36 4 5 3 12 2 2 3 1 6 42 8 5 4 7 3 3
China biovar 3
2013Jiang#146 4 Brucella2013 NM1185 Cattle Inner Mongolia, B. abortus Buyun Cui 1990 36 4 5 3 12 2 2 3 1 6 42 8 6 4 5 3 3
China biovar 3
2013Jiang#152 4 Brucella2013 NM1219 Cattle Inner Mongolia, B. abortus Buyun Cui 1995 36 4 5 3 12 2 2 3 1 6 42 8 5 5 7 3 3
China biovar 3
2013Jiang#113 4 Brucella2013 NM1075 Cattle Inner Mongolia, B. abortus Buyun Cui 1985 117 4 5 3 12 2 2 3 2 8 42 8 5 6 3 3 3
China biovar 3
2006 4 Brucella2012 BfR 95 Mouse ? B. abortus 1 Gilles B. abortus 28 82 4 5 4 12 2 2 3 3 6 42 8 3 6 3 3 5
LeFlèche#135 Vergnaud
2009Her#011 4 Brucella2012 KRef15 Cattle USA B. abortus 1 Moon Her B. abortus 28 82 4 5 4 12 2 2 3 3 6 42 8 3 6 3 3 5
2013Jiang#083 4 Brucella2013 Cattle Xinjiang, China B. abortus Buyun Cui 2011 36 4 5 3 12 2 2 3 1 6 42 8 4 6 5 3 3
biovar 3
2013Garofolo_ 4 Brucella_ 3921 Cattle San Gregorio B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 4 6 6 3 3
3921 ITALIA_1 Magno,Italy biovar3 Garofolo
2013Garofolo_ 4 Brucella_ 5007 Cattle San Gregorio B. abortus_ Giuliano 2011 36 72 4 5 3 12 2 2 3 1 6 42 8 4 6 6 3 3
5007 ITALIA_1 Magno,Italy biovar3 Garofolo

December 2015 | Volume 6 | Article 1375


Identification of an Unusual B. abortus SPR Strain
Kang et al. Identification of an Unusual B. abortus SPR Strain

phenotypic and molecular typing method. However, it shared origins, suggesting that more B. abortus strains phenotypically
the same phage lysis profiles to that of B. melitensis biovar 1. In identified as biovar 3 are required for the comparison. The MLVA
conclusion, strain 8416 is the only SPR strain isolated from the assay confirmed that B. abortus biovar 3 is a heterogeneous group
infected human thus far with a similar phage lysis pattern with (Le Fleche et al., 2006), and in agreement with the B. abortus
B. melitensis 16 M. However, despite same resistant to phage Tb, biovar 3 divided into two sub-biovar 3a and 3b (Huber et al.,
we could not comprehensively compare with phage lysis profiles 2009).
of the three reported SPR strains due to different Brucella phages In this study, an atypical B. abortus strain displaying a phage
tested among them. lysis profile similar to B. melitensis biovars 1 was identified.
Currently, MLVA has been mainly used for tracking the Most importantly, the lysis pattern by bacteriophages observed
variances of the bacterial genus with a high homology, such as in this newly uncovered B. abortus SPR strain. Although phage
Brucella genus (Haguenoer et al., 2011). The MLVA-16 (panel typing in general can successfully classify Brucella species, our
1, 2A and 2B) assay was widely used for molecular typing of research calls for attention as to conclusions on SPR strains.
a larger collection of isolates at both species and biovars level. Further investigation focusing on the strain 8416’s whole genomic
The panel 1 comprised eight minisatellite markers for species variations associated with phage resistance is needed.
identification (Le Fleche et al., 2006) and the panel 2 markers were
found with a higher biovar discriminatory power. Surprisingly,
the MLVA-16 typing results showed that strain 8416 was clustered ACKNOWLEDGMENTS
into the Chinese B. abortus biovar 3 strains (Jiang et al., 2013)
with four variable loci (bruce04, 07, 11, and 55). Actually, among This study was supported by the National Nature Science
the four known panel 1 genotypes (28, 30, 112, 116), strain 8416 Foundation (81360444, 81460319) and the Inner Mongolia
(genotype 30) was distinct from other 65 Chinese B. abortus Autonomous Region of Nature Science Foundation
biovar 3 strains isolated previously from different geographic (2013MS1105).

REFERENCES Huber, B., Scholz, H. C., Lucero, N., and Busse, H. J. (2009). Development of a PCR
assay for typing and subtyping of Brucella species. Int. J. Med. Microbiol. 299,
Adone, R., and Pasquali, P. (2013). Epidemiosurveillance of brucellosis. Rev. Sci. 563–573. doi: 10.1016/j.ijmm.2009.05.002
Tech. 32, 199–205. Jiang, H., Wang, H., Xu, L., Hu, G., Ma, J., Xiao, P., et al. (2013). MLVA genotyping
Alton, G. G., Jones, L. M., and Pietz, D. E. (1975). Laboratory techniques in of Brucella melitensis and Brucella abortus isolates from different animal species
brucellosis. Monogr. Ser. No.55 World Health Organ. 1975, 1–163. and humans and identification of Brucella suis vaccine strain S2 from cattle in
Bounaadja, L., Albert, D., Chenais, B., Henault, S., Zygmunt, M. S., Poliak, S., China. PLoS ONE 8:e76332. doi: 10.1371/journal.pone.0076332
et al. (2009). Real-time PCR for identification of Brucella spp.: a comparative Jones, L. M., McDuff, C. R., and Wilson, J. B. (1962). Phenotypic alterations in the
study of IS711, bcsp31 and per target genes. Vet. Microbiol. 137, 156–164. doi: colonial morphology of Brucella abortus due to a bacteriophage carrier state.
10.1016/j.vetmic.2008.12.023 J. Bacteriol. 83, 860–866.
Bricker, B. J., and Halling, S. M. (1994). Differentiation of Brucella abortus bv. 1, 2, Le Fleche, P., Jacques, I., Grayon, M., Al Dahouk, S., Bouchon, P., Denoeud, F.,
and 4, Brucella melitensis, Brucella ovis, and Brucella suis bv. 1 by PCR. J. Clin. et al. (2006). Evaluation and selection of tandem repeat loci for a Brucella MLVA
Microbiol. 32, 2660–2666. typing assay. BMC Microbiol. 6:9. doi: 10.1186/1471-2180-6-9
Chen, Y., Ke, Y., Wang, Y., Yuan, X., Zhou, X., Jiang, H., et al. (2013). Ocampo-Sosa, A. A., Aguero-Balbin, J., and Garcia-Lobo, J. M. (2005).
Changes of predominant species/biovars and sequence types of Brucella isolates, Development of a new PCR assay to identify Brucella abortus biovars 5, 6
Inner Mongolia, China. BMC Infect. Dis. 13:514. doi: 10.1186/1471-2334-1 and 9 and the new subgroup 3b of biovar 3. Vet. Microbiol. 110, 41–51. doi:
3-514 10.1016/j.vetmic.2005.06.007
Corbel, M. J., and Morris, J. A. (1974). Studies on a smooth phage-resistant Van der Henst, C., De Barsy, M., Zorreguieta, A., Letesson, J. J., and De Bolle, X.
variant of Brucella abortus I. Immunological properties. Br. J. Exp. Pathol. 55, (2013). The Brucella pathogens are polarized bacteria. Microbes Infect. 15,
78–87. 998–1004. doi: 10.1016/j.micinf.2013.10.008
Corbel, M. J., and Morris, J. A. (1975). Studies on a smooth phage resistant Zhang, W. Y., Guo, W. D., Sun, S. H., Jiang, J. F., Sun, H. L., Li, S. L., et al. (2010).
variant of Brucella abortus II. Mechanism of phage resistance. Br. J. Exp. Pathol. Human brucellosis, Inner Mongolia, China. Emerg. Infect. Dis. 16, 2001–2003.
56, 1–7. doi: 10.3201/eid1612.091081
Corbel, M. J., and Thomas, E. L. (1976). The in vivo activity of a smooth
phage-resistant variant of Brucella abortus strain 19. Br. Vet. J. 132, Conflict of Interest Statement: The authors declare that the research was
121–123. conducted in the absence of any commercial or financial relationships that could
Grissa, I., Bouchon, P., Pourcel, C., and Vergnaud, G. (2008). On-line resources for be construed as a potential conflict of interest.
bacterial micro-evolution studies using MLVA or CRISPR typing. Biochimie 90,
660–668. doi: 10.1016/j.biochi.2007.07.014 Copyright © 2015 Kang, Li, Piao, Tian, Jiang, Jia, Lin, Cui, Chang, Guo and Zhu.
Haguenoer, E., Baty, G., Pourcel, C., Lartigue, M. F., Domelier, A. S., Rosenau, A., This is an open-access article distributed under the terms of the Creative Commons
et al. (2011). A multi locus variable number of tandem repeat analysis (MLVA) Attribution License (CC BY). The use, distribution or reproduction in other forums
scheme for Streptococcus agalactiae genotyping. BMC Microbiol. 11:171. doi: is permitted, provided the original author(s) or licensor are credited and that the
10.1186/1471-2180-11-171 original publication in this journal is cited, in accordance with accepted academic
Harrington, R. Jr., Bond, D. R., and Brown, G. M. (1977). Smooth phage-resistant practice. No use, distribution or reproduction is permitted which does not comply
Brucella abortus from bovine tissue. J. Clin. Microbiol. 5, 663–664. with these terms.

Frontiers in Microbiology | www.frontiersin.org 8 December 2015 | Volume 6 | Article 1375


REVIEW ARTICLE

Cues and regulatory pathways involved in natural competence


and transformation in pathogenic and environmental
Gram-negative bacteria
Patrick Seitz & Melanie Blokesch
Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland

Correspondence: Melanie Blokesch, EPFL- Abstract


SV-UPBLO/Station 19, Ecole Polytechnique
Fédérale de Lausanne (EPFL), CH-1015 Bacterial genomics is flourishing, as whole-genome sequencing has become
Lausanne, Switzerland. Tel.: +41 21 693 affordable, readily available and rapid. As a result, it has become clear how fre-
0653; fax: +41 21 693 7210; e-mail: quently horizontal gene transfer (HGT) occurs in bacteria. The potential impli-
melanie.blokesch@epfl.ch cations are highly significant because HGT contributes to several processes,
including the spread of antibiotic-resistance cassettes, the distribution of
Received 7 May 2012; revised 27 July 2012;
toxin-encoding phages and the transfer of pathogenicity islands. Three modes
accepted 21 August 2012. Final version
published online 20 September 2012.
of HGT are recognized in bacteria: conjugation, transduction and natural
transformation. In contrast to the first two mechanisms, natural competence
DOI: 10.1111/j.1574-6976.2012.00353.x for transformation does not rely on mobile genetic elements but is driven
solely by a developmental programme in the acceptor bacterium. Once the
Editor: Hauke Hennecke bacterium becomes competent, it is able to take up DNA from the environ-
ment and to incorporate the newly acquired DNA into its own chromosome.
Keywords The initiation and duration of competence differ significantly among bacteria.
horizontal gene transfer; DNA uptake; In this review, we outline the latest data on representative naturally trans-
carbon catabolite repression; quorum
formable Gram-negative bacteria and how their competence windows differ.
sensing; DNA repair; type IV pilus.
MICROBIOLOGY REVIEWS

We also summarize how environmental cues contribute to the initiation of


competence in a subset of naturally transformable Gram-negative bacteria
and how the complexity of the niche might dictate the fine-tuning of the
competence window.

acquired genetic elements. Horizontal gene transfer


Introduction
(HGT) is important in rendering harmless bacteria into
In 2011, the spread of food-borne gastroenteritis in major human pathogens, as exemplified by the commen-
Europe, most notably in Germany, was widely reported. sal bacterium E. coli mentioned above. Indeed, Rasko
The pathogen caused severe bloody diarrhoea, often et al. (2011) concluded from their WGS findings that
accompanied by haemolytic uremic syndrome. The causa- ‘horizontal genetic exchange allowed for the emergence of
tive agent of the outbreak was Escherichia coli serotype the highly virulent Shiga-toxin–producing enteroaggrega-
O104:H4. The strain was unique in that it combined the tive E. coli O104:H4 strain that caused the German out-
‘potentials of two different pathogens: Shiga-toxin- break’. Recent reviews on this topic concluded that there
producing E. coli and enteroaggregative E. coli’ (Bielaszewska is substantial evidence that HGT contributes to the acqui-
et al., 2011). Multiple isolates of this outbreak were fully sition, integration and maintenance of pathogenicity
sequenced for molecular epidemiological analysis (Rasko islands (PAIs) in bacterial genomes (Hacker et al., 1997,
et al., 2011; Grad et al., 2012). Such whole-genome 2003; Dobrindt et al., 2004, 2010; Ahmed et al., 2008). In
sequencing (WGS) studies are extremely helpful in analy- general, it is widely accepted that HGT significantly
sing single-nucleotide polymorphisms (SNPs) and allow impacts the evolution of bacterial genomes and their
us to understand patterns of genetic diversity, which can adaptation to new environments (a nonexhaustive list of
help in the identification of transmission routes. Further- reviews on this topic published within this century would
more, WGS sheds light on the acquisition of horizontally be Eisen, 2000; Ochman et al., 2000; Boucher et al., 2001;

ª 2012 Federation of European Microbiological Societies FEMS Microbiol Rev 37 (2013) 336–363
Published by Blackwell Publishing Ltd. All rights reserved
Regulation of natural competence in Gram-negative bacteria 337

Koonin et al., 2001; Boucher et al., 2003; Thomas & Niel- that has been recently reviewed by Claverys et al., 2006)
sen, 2005; Davidsen et al., 2007; Cohan & Koeppel, 2008; are highlighted throughout the text.
Koonin & Wolf, 2008; Ambur et al., 2009; Popa & Dagan, Finally, although this review emphasizes on pathogens
2011; Stokes & Gillings, 2011; Wiedenbeck & Cohan, 2011). (with or without a niche outside of humans), a plethora
HGT in bacteria occurs through three main mecha- of nonpathogenic bacteria are known to be naturally
nisms: transduction, conjugation, and natural transforma- transformable. The rationale behind our focus on patho-
tion. The latter mechanism is based on the uptake of free gens is based on two factors: (1) Research on HGT has
DNA from the environment and therefore does not rely long been influenced by medically relevant questions,
on mobile genetic elements such as phages and plasmids; such as how virulence factors are acquired by pathogens
instead, it is solely encoded by the acceptor bacterium. and how antibiotic-resistance genes spread among micro-
Natural competence is the developmental state of the bac- organisms. In fact, the discovery of natural transforma-
terium in which it is able to take up external DNA and tion in bacteria was based on the seminal work by
to recombine this DNA into the chromosome, thereby Frederick Griffith. Griffith was interested in understand-
undergoing natural transformation. Natural competence ing the difference between virulent and nonvirulent
and transformation are common to a wide variety of bac- strains of Streptococcus pneumoniae and how such strains
terial species and have been extensively reviewed (Lorenz can interconvert (Griffith, 1928). A more recent example
& Wackernagel, 1994; Dubnau, 1999; Chen & Dubnau, would be the opportunistic pathogen Porphyromonas
2004). The composition and dynamics of the DNA- gingivalis. Porphyromonas gingivalis belongs to the phylum
uptake complexes (Box 1) have also been the focus of bacteroidetes and contributes to periodontal disease upon
several excellent recent reviews (Averhoff & Friedrich, stable colonization of the human oral cavity. Recent data
2003; Allemand & Maier, 2009; Claverys et al., 2009; Bur- demonstrated that natural competence and transforma-
ton & Dubnau, 2010; Allemand et al., 2012). However, tion is the major driving force behind DNA exchange in
less is known about the initiation of competence, particu- this organism (Tribble et al., 2012). The resulting genetic
larly in Gram-negative bacteria. It has become clear in variability is most likely the reason for P. gingivalis’
recent years that there are major differences in the regula- survival in the human host and its evasion from human
tory network involved in competence induction in immune defences (Tribble et al., 2012). (2) The second
Gram-negative bacteria. Thus, the present review aims to reason that pathogenic bacteria are sometimes considered
provide an overview of how environmental signals drive more appropriate for the study of HGT is the somewhat
natural competence and transformation in these organ- biased sequencing effort. Whereas often only one or very
isms. Differences and similarities in the conditions that few whole-genome sequences of nonpathogenic bacteria
induce competence in Gram-positive bacteria (a topic are available, a trend towards sequencing many different

Box 1 . A hypothetical mode for DNA update in V. cholerae

FEMS Microbiol Rev 37 (2013) 336–363 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
338 P. Seitz & M. Blokesch

isolates of human pathogens has recently emerged enough, another naturally competent e-proteobacterium,
(Tettelin et al., 2005; Croucher et al., 2011; Mutreja et al., Campylobacter jejuni, does not seem to share this depen-
2011). The use of information from multiple isolates dency on a transformation-related T4SS; instead DNA
facilitates the identification of horizontally acquired uptake and transformation is dependent on genes encod-
regions and the underlying regulatory circuits driving ing components of a putative type II secretion or type IV
HGT. pilus-like system (Wiesner et al., 2003). More information
on natural competence of C. jejuni has been recently
reviewed by Young et al. (2007). And although DNA
Caught in competence – constitutively
uptake in H. pylori is exceptional, recent studies indicate
competent Gram-negative bacteria
that certain steps might be conserved in DNA transport.
Whereas the uptake of DNA into the periplasm by a
Helicobacter pylori responds to DNA damage
T4SS is specific to H. pylori, transport across the inner
Helicobacter pylori is an extremely successful human path- membrane involves homologous proteins found in both
ogen. This bacterium colonizes the gastric epithelia of naturally competent Gram-negative and Gram-positive
more than half of the world’s population (Suerbaum & bacteria (Stingl et al., 2010; Box 1).
Michetti, 2002) and has probably done so since humans DNA uptake is not the only ‘exception that proves the
migrated out of Africa roughly 60 000 years ago (Falush rule’ in H. pylori. An even more striking difference to
et al., 2003; Linz et al., 2007). Helicobacter pylori is also most other naturally transformable bacteria occurs at the
one of the most diverse bacterial species (Achtman et al., regulatory level of competence. In contrast to those bacte-
1999). This bacterium’s diversity has been studied for ria, H. pylori does not limit its competence window in
almost two decades, and it is clear that every person response to environmental stimuli. Instead, this bacterium
infected by H. pylori carries a unique strain (Langenberg is naturally competent during all growth phases (Israel
et al., 1986; Majewski & Goodwin, 1988; Achtman et al., et al., 2000; Baltrus & Guillemin, 2006). Because transfor-
1999). Extremely frequent recombination events are key mation frequencies specifically peak within certain growth
to this extraordinary genetic diversity (Suerbaum et al., phases during in vitro growth (Baltrus & Guillemin,
1998; Kersulyte et al., 1999; Falush et al., 2001; Morelli 2006), a regulatory system must exist to at least fine-tune
et al., 2010). This high recombination frequency and DNA uptake. The mediators involved in this process
frequent horizontal flow of genetic material in H. pylori is remain unknown. The only stimulus experimentally pro-
directly linked with its ability to undergo natural trans- ven to increase natural transformation in H. pylori is
formation, the topic of this review. However, in terms of
natural competence, H. pylori differs from other Gram-
negative bacteria in two important respects: the mecha-
nism of the DNA-uptake process and the constitutive
competence state of the bacterium.
Most naturally competent bacteria rely on type IV
pilus-related DNA-uptake machineries (Hobbs & Mattick,
1993 and for recent review see Chen & Dubnau, 2004;
Allemand & Maier, 2009; Burton & Dubnau, 2010; Box
1). In H. pylori, however, the components involved in the
translocation of transforming DNA resemble type IV
secretion systems (T4SS; Hofreuter et al., 1998, 2000;
Karnholz et al., 2006), such as the archetypical VirB/
VirD4 system of Agrobacterium tumefaciens (for recent
review see Alvarez-Martinez & Christie, 2009). Indeed,
many bacteria use similar systems to export both protein
and DNA substrates. In addition to this competence- Fig. 1. The most prominent environmental cues involved in
related T4SS, known as the comB system, many H. pylori competence induction or the fine-tuning of competence. These signals
strains harbour a ‘classical’ T4SS encoded on a PAI. This include genotoxic stresses causing DNA damage, such as UV light or
certain antibiotics (in red); bacterial cell-cell communication systems
system is used to inject the virulence factor CagA into
(e.g. quorum-sensing; in green); the starvation of preferred carbon
gastric epithelial cells (Odenbreit et al., 2000). These sources, leading to the accumulation of the intracellular secondary
two T4SS are independent of each other. Moreover, the messenger cAMP (carbon catabolite repression; in blue); and the
Cag-PAI is fully dispensable in natural transformation presence or absence of certain carbon sources (such as the GlcNAc
(Hofreuter et al., 2000; Israel et al., 2000). Interestingly polymer chitin for Vibrio cholerae). Details are provided in the text.

ª 2012 Federation of European Microbiological Societies FEMS Microbiol Rev 37 (2013) 336–363
Published by Blackwell Publishing Ltd. All rights reserved
Regulation of natural competence in Gram-negative bacteria 339

DNA damage (Dorer et al., 2010; Fig. 1). Indeed, Dorer prior to unwinding and perhaps extracellularly’ (Dorer
et al. (2010) showed that mutations in a DNA repair pro- et al., 2011). The idea that such foreign species-derived
tein, as well as treatment with the DNA-damaging agent DNA degradation could occur outside the cells might
ciprofloxacin, led to the upregulation of transcription of very well be possible as DNA damage-induced lysis of
many genes involved in DNA uptake and to increased neighbouring cells would not only release these cells’
recombination. Consequently, transformation frequencies DNA but also their REases.
increased four to fivefold. Among the upregulated genes Nuclease-mediated degradation of extracellular DNA
was a homologue of a T4 phage-derived lysozyme that has also been described for nontransformable variants of
had previously been characterized for its lytic activity C. jejuni (Gaasbeek et al., 2009, 2010). More specifically,
(Marsich et al., 2002). Therefore, DNA damage might these authors showed that acquisition of prophage-
lead not only to increased DNA uptake but also to the encoded endonucleases lead to an inhibition of natural
enhanced lysis of neighbouring cells and ultimately to transformability of C. jejuni (Gaasbeek et al., 2009, 2010).
high levels of natural transformation (for recent review This is in excellent agreement with an earlier publication
see Dorer et al., 2011). The induction of competence on another naturally competent Gram-negative bacterium,
upon DNA damage, coupled with fratricide, has also been Vibrio cholerae: in this study Blokesch and Schoolnik dem-
observed in the Gram-positive bacterium S. pneumoniae onstrated that an extracellular nuclease Dns degrades
(Guiral et al., 2005; Havarstein et al., 2006; Prudhomme external and potentially transforming DNA thus inhibiting
et al., 2006). Although the two bacterial species are only transformation of this organism (Blokesch & Schoolnik,
distantly related, they share another characteristic: 2008). However, repression of nuclease production at high
S. pneumoniae and H. pylori both lack a bona fide SOS cell density occurs via a quorum-sensing regulatory circuit
response system. Competence induction in response to and thus still allows natural transformation to occur in
DNA damage might thus represent a product of conver- V. cholerae (see below; Blokesch & Schoolnik, 2008; Lo
gent evolution in distantly related bacteria that lack an Scrudato & Blokesch, 2012).
SOS response system (Dorer et al., 2010). This same con-
dition holds for the Gram-negative pathogen Legionella
The competence-induced uptake and secretion
pneumophila. In this organism natural competence is also
of DNA – the unique case of Neisseria
induced upon antibiotics- and UV-induced DNA damage
(Charpentier et al., 2011). Neisseria spp. have been known to be naturally competent
Some naturally competent bacteria are biased towards for more than half a century (Alexander & Redman,
the uptake of genetic material derived from closely related 1953; Catlin & Cunningham, 1961). Recent research on
neighbours. Various strategies have evolved to this end the natural competence and transformation of Neisseria
(Fig. 2). Helicobacter pylori is believed to be able to take has primarily focused on the human pathogens Neisseria
up any kind of DNA, independent of its source, but meningitidis and Neisseria gonorrhoeae. These species are
recent results indicated that incoming DNA can be con- very similar with respect to competence regulation and to
trolled by restriction-modification (R-M) systems (Aras their DNA-uptake mechanisms (Koomey, 1998). Thus,
et al., 2002; Humbert & Salama, 2008; Humbert et al., many findings derived from one organism can readily be
2011). R-M systems are based on restriction endonucleas- applied to the other species.
es (REases), which cleave DNA at specific sequence Like H. pylori, Neisseriae are competent for natural
motifs, and corresponding DNA methyltransferases, transformation during all growth phases (Sparling, 1966).
which inhibit the DNA cleavage of self DNA by methyla- Environmental stimuli or secreted factor responsible for
tion of the respective recognition sites. This mechanism the induction or enhancement of competence and trans-
recognizes and degrades foreign genetic material (Fig. 2). formation have not been identified to date, except for a
Indeed, R-M systems were traditionally associated with minor effect of growth temperature (Sparling, 1966). No
protection against bacteriophages or conjugative plasmids further studies have been conducted on growth tempera-
(for review see Kessler & Manta, 1990). It is not entirely ture, most likely because it only reflects elevated metabolic
clear how the REases destroy unmethylated DNA to activity. Natural transformation peaked at 37 °C for
reduce transformation frequencies in H. pylori (Humbert N. gonorrhoeae (Sparling, 1966), which corresponds to the
et al., 2011) as current models suggest that only single- constant ambient temperature of its niche, which is the
stranded DNA enters the cytoplasm (as reviewed by Chen mucosal tissue of the human urogenital tract. As for
& Dubnau, 2004; Allemand & Maier, 2009; Burton & H. pylori, no environmental reservoir is known for
Dubnau, 2010; Allemand et al., 2012; Box 1). Thus, it N. gonorrhoeae. Within the human host, the immune sys-
was concluded in a recent review: ‘Because restriction tem puts Neisseriae under strong selective pressure. Thus,
enzymes prefer double-stranded DNA, they likely act the extensive horizontal flow of genetic material mediated

FEMS Microbiol Rev 37 (2013) 336–363 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
340 P. Seitz & M. Blokesch

Fig. 2. Different strategies have evolved to enhance the probability of integrating species-specific DNA. Certain Gram-negative bacteria, such as
Haemophilus influenzae and Neisseria gonorrhoeae, are notably fastidious about the kind of DNA that they absorb. To sort species-specific from
nonspecies-specific DNA, these organisms have evolved specific DNA uptake sequences (DUS) that are overrepresented in their genomes and
recognized by naturally competent members of the same species through as-yet-unknown receptor proteins. These receptors are most likely
surface-exposed and initiate the ingestion of species-specific DNA into the cytoplasm of the bacterium (I). An alternative strategy to increase the
chances of taking up species-specific DNA is to link the regulation of natural competence and transformation to species-specific autoinducer
synthesis and recognition (II). The involvement of a ‘competence pheromone’ is a common strategy of Gram-positive bacteria and has also been
recently suggested for the Gram-negative bacterium Vibrio cholerae. Other bacteria, such as Helicobacter pylori, have evolved specific restriction-
modification (R-M) systems, which assure that species-nonspecific DNA is degraded and thus cannot recombine with the chromosome. The exact
mechanism is so far unknown for this mode of restricting nonrelated DNA. One possible mechanism could be that the restriction enzymes
(REases) are released from lysed bacteria (together with the DNA) and degrade species-nonspecific dsDNA extracellularly. Finally, as in the Gram-
positive bacterium Streptococcus pneumoniae, certain naturally competent Gram-negative bacteria initiate the donation of DNA from their
siblings. This process may be mediated either by autolysis, by the intentional killing of a subpopulation (e.g. the lysis of neighbouring cells) or by
the active secretion of DNA through a T4SS, as in a subset of Neisseria gonorrhoeae strains.

by natural transformation might provide an important siblings via autolysis or actively exported via a T4SS
means of adaptation. Indeed, natural transformation (Hamilton et al., 2005; Hamilton & Dillard, 2006; Fig. 2).
might contribute significantly to the high genomic diver- Although the T4SS involved in DNA secretion is found
sity of Neisseria (Viscidi & Demma, 2003; Hanage et al., only in a subset of N. gonorrhoeae strains (Dillard &
2005; Maiden, 2008). Researchers provided evidence to Seifert, 2001) and does not contribute to DNA secretion
support extensive interspecies recombination and inter- in N. meningitidis (Woodhams et al., 2012), all investi-
generic transfer of DNA in the late 1990s (Feil et al., 1996, gated Neisseria strains can undergo autolysis. Little is
1999; Kroll et al., 1998). Only recently have genotyping known about the regulation of either of these processes
techniques made it possible to determine the extent of or whether they are driven by environmental stimuli;
diversity in Neisseria (for recent review see Maiden, 2008). however, both mechanisms promote genetic transfer
As described earlier, H. pylori actively secretes a lyso- within bacterial communities and might be beneficial for
zyme-like protein to increase the concentration of naked genome maintenance (Fig. 2).
DNA in the environment (Dorer et al., 2010). Neisseria Whereas H. pylori limits interspecies transformation via
gonorrhoeae facilitates the release of DNA by two different intracellular R-M systems, Neisseria spp. discriminate
mechanisms. In this organism, DNA is either donated to between self and foreign DNA at the level of the

ª 2012 Federation of European Microbiological Societies FEMS Microbiol Rev 37 (2013) 336–363
Published by Blackwell Publishing Ltd. All rights reserved
Regulation of natural competence in Gram-negative bacteria 341

DNA-uptake machinery. In fact, the finding that N. menin- and H. pylori, the primary ecological niche of this organism
gitidis and N. gonorrhoeae preferentially take up DNA from is the human body, specifically, the upper respiratory tract.
closely related species is one of the strongest arguments Haemophilus influenzae also causes ear infections and men-
supporting the ‘DNA for repair’ hypothesis. More precisely, ingitis in children and respiratory disease in the elderly, as
for Neisseria spp., as well as for Haemophilus influenzae, well as in patients with cystic fibrosis and AIDS.
species-specific uptake of genetic material is dependent on The environmental signal that triggers the onset of natu-
so-called DNA-uptake sequences (DUS; Goodman & Scoc- ral competence in H. influenzae is not known; however,
ca, 1988; Elkins et al., 1991), which are conserved short because ‘the human respiratory tract is a hostile environ-
DNA sequence motifs (Danner et al., 1980; Fitzmaurice ment for bacteria’, and ‘nutrients and energy sources are
et al., 1984; Elkins et al., 1991; Aas et al., 2002). DUS limited’ (Murphy & Brauer, 2011), it is not surprising that
motifs may serve as a binding target for a still unidentified a link between starvation for preferred carbon sources and
and hypothetical receptor protein localized on the outside competence induction has been discovered in vitro for this
of DUS-containing Gram-negative bacteria (Fig. 2). organism. Indeed, in the laboratory, H. influenzae becomes
Furthermore, DUS motifs are highly overrepresented in the naturally transformable when the culture reaches stationary
bacterial genomes. Early estimates of roughly one DUS phase (Redfield, 1991) or when the cells are transferred
motif per kb of DNA (Goodman & Scocca, 1991) were vali- from rich to starvation medium (Herriott et al., 1970). The
dated when the first two Neisseria genomes were made addition of the secondary messenger cyclic adenosine
publicly available [GenBank numbers AE002098 (Tettelin monophosphate (cAMP) to exponentially growing bacteria
et al., 2000) and AE004969]. Interestingly, further analysis also induces competence (Wise et al., 1973). As a conse-
revealed a strong bias in DUS frequencies towards genome quence it was shown that cAMP receptor protein (CRP)
maintenance genes (Davidsen et al., 2004). DUS motifs and adenylate cyclase (CyaA) are essential for the develop-
provide yet another mechanism to support species-specificity ment of natural transformation in this organism (Chandler,
in transformation (Fig. 2) and may play a role in ‘recom- 1992; Dorocicz et al., 1993).
binational repair’ (Michod et al., 2008). Recent work by Rosemary Redfield’s group and others
Recent findings, however, call into question both the have provided further insight into the induction of
nature and function of DUS (Ambur et al., 2007; Duffin competence in H. influenzae. These researchers identified
& Seifert, 2010). DNA uptake can occur in the absence of the gene sxy (Redfield, 1991; homologue in other compe-
DUS, and the effect of DUS on transformation frequen- tent bacteria is tfoX), which is essential for natural
cies varies significantly among strains of N. gonorrhoeae competence. Furthermore, Sxy induced constitutive com-
(Duffin & Seifert, 2010). Because the effect of DUS on petence upon overexpression from a multi-copy plasmid
natural transformation efficiency was not proportional to (Williams et al., 1994). The expression of sxy/tfoX as an
its effect on DNA uptake, the authors proposed an addi- early competence gene was increased upon the addition
tional role for DUS downstream of the DNA-uptake pro- of cAMP, a phenomenon consistent with the increase in
cess (Duffin & Seifert, 2010). In addition, Duffin & transformability (Zulty & Barcak, 1995). Sxy may act as a
Seifert (2012) demonstrated that DUS sequences can positive regulator of late competence-specific genes;
enhance the somewhat less efficient transformation by however, the mechanism of this positive regulation has
single-stranded DNA and that this phenomenon shows long been debated because Sxy lacks recognizable DNA-
strand preference. The identification of a human gene binding motifs (Macfadyen, 2000).
fragment in the genome of N. gonorrhoeae provides fur- The competence regulon in H. influenzae was identified
ther evidence for DUS-independent DNA uptake (Ander- in 2005 using microarray expression profiling (Redfield
son & Seifert, 2011). This finding highlights the et al., 2005). The regulon consisted of 25 genes in 13
evolutionary potential of N. gonorrhoeae and the impor- transcriptional units, each containing a characteristic
tance of HGT in host-pathogen interactions. promoter-associated 22-bp element, the competence regu-
latory element (CRE; Karudapuram & Barcak, 1997; Mac-
fadyen, 2000; Redfield et al., 2005). In these studies,
Competence induction based on
competence-induced transcription was again strongly
starvation signals
dependent on cAMP and the competence regulator Sxy.
Based on this information, it was proposed that Sxy
Haemophilus influenzae requires a nutrient
might act as an accessory factor, directing CRP to CRE
downshift to induce competence
sites and/or stabilizing the transcription-initiation com-
Haemophilus influenzae is yet another Gram-negative bac- plex (Macfadyen, 2000; Redfield et al., 2005). The CRE
terium in which natural competence and transformation elements were later renamed as CRP-S sites, describing a
has been studied for many years. As in the Neisseriaceae binding site for CRP; these sites, unlike canonical

FEMS Microbiol Rev 37 (2013) 336–363 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
342 P. Seitz & M. Blokesch

CRP-binding sites (CPR-N), are dependent on Sxy (Cam- versatile group of environmental bacteria that live pre-
eron & Redfield, 2006, 2008). What is the exact nature of dominantly in soil, sediment, and water, occurring less
the link between cAMP/CRP, Sxy and competence induc- commonly as opportunistic human pathogens (for recent
tion? Although there is no definitive answer at present, review on Pseudomonas taxonomy and isolation of type
the current model (based on work by the Redfield labora- strains see Peix et al., 2009). Many Pseudomonas spp.
tory) is as follows: (1) CRP and cAMP are required to undergo natural transformation, including P. mendocina,
strongly induce the transcription of sxy (Cameron et al., P. alcaligenes, P. pseudoalcaligenes, P. fluorescencs and
2008) via the binding of the cAMP/CRP complex to a P. stutzeri (Carlson et al., 1983; Demaneche et al., 2001);
CRP-binding site in the sxy promoter region (Zulty & to date, there is no evidence for natural competence of
Barcak, 1995; Cameron et al., 2008). (2) A stem loop P. aeruginosa. Thus, most of the research on natural com-
structure is formed within the 5′ UTR of the sxy mRNA, petence has focused on the ubiquitous soil bacterium
which is involved in controlling both the amount and P. stutzeri. This bacterium has a notably complex metabo-
translation efficiency of the sxy mRNA (Cameron et al., lism, which allows it not only to grow on a wide variety
2008). The authors of this study suggested that the latter of carbon sources but also to degrade environmental
was caused by the sequestration of the SD site by the pollutants and to fix nitrogen (as recently reviewed by
mRNA stem loop structure, which limited the binding of Lalucat et al., 2006).
ribosomes. Thus, the increase in Sxy protein following Most of the research on the regulation and mechanism
the transfer of the H. influenzae cells to starvation medium of natural transformation in P. stutzeri was performed in
might occur because the secondary structure of the sxy Wilfried Wackernagel’s laboratory, and a seminal review,
mRNA has ‘the potential to play a sensory role’ (Cameron ‘Bacterial Gene Transfer by Natural Genetic Transforma-
et al., 2008). Such a sensory function might be based on tion in the Environment’, was published by Lorenz &
the speed of transcription; ribosomes may bind before the Wackernagel (1994). This group showed, for example,
5′ UTR stem loop is properly folded, ‘thus making the that as with most other Gram-negative bacteria, the trans-
progress of RNA polymerase a potential transducer of formability of P. stutzeri correlates with the expression of
nutritional signals’ (Cameron et al., 2008). Alternatively, type IV pilus structural or assembly genes (Graupner
the 5′ UTR of the sxy mRNA might be directly involved in et al., 2000), indicating that DNA uptake in P. stutzeri
regulation, perhaps by a riboswitch-like mechanism. This might be similar to the mechanism proposed for other
hypothesis is supported by the recently described ribo- Gram-negative bacteria (as reviewed by Chen & Dubnau,
switch, which was identified in association with a sxy 2004 and described in Box 1). There are, however, several
homologue in V. cholerae (tfoXGEMM, Sudarsan et al., interesting differences. For example and in contrast to
2008; details below). In conclusion, cAMP and CRP, as the H. influenzae, transformation by single-stranded DNA
major players in carbon-catabolite repression (CCR), are (ssDNA) is efficient in P. stutzeri, reaching up to 5%
important in the natural transformation of H. influenzae compared with double-stranded DNA (dsDNA; Meier
because they are involved in the transcription of both et al., 2002). Notably, transformation with ssDNA was
early- (sxy) and late- (e.g. comA) competence genes. This dependent on the same components identified for
feature couples the nutritional state of the bacterium to dsDNA, indicating that both substrates follow identical
the induction of competence. routes into the cell (Meier et al., 2002). Furthermore, and
Apart from its cAMP-dependent competence induction, again in contrast to N. gonorrhoeae or H. influenzae,
H. influenzae has a preference for species-specific DNA. DNA uptake was not dependent on the presence of
The species-specificity of the DNA is discriminated at the DNA-uptake motifs. However, early competition experi-
level of the DNA-uptake process through the recognition ments with either homologous or heterologous DNA sug-
of specific DUS sequences, as described above for Neisseria gested that P. stutzeri might still discriminate between self
species (Danner et al., 1980; Fitzmaurice et al., 1984; and foreign DNA at an early step of transformation
Smith et al., 1999). through an unknown mechanism (Carlson et al., 1983;
Lorenz & Wackernagel, 1990). It was also shown that
homology-facilitated illegitimate recombination is much
Environmental cues involved in
less efficient in P. stutzeri than in other naturally trans-
competence induction in bacteria
formable bacteria, such as S. pneumoniae or Acinetobacter
commonly found outside a host
(Meier & Wackernagel, 2003). Finally, DNA restriction
has also been proposed as a barrier to natural transforma-
Natural transformation in Pseudomonadaceae
tion in P. stutzeri (Berndt et al., 2003). All of these fac-
In contrast to the highly adapted human pathogens tors result in a limited exchange of heterologous DNA
discussed earlier, the pseudomonads are an extremely (Fig. 2), which has important implications for the spread

ª 2012 Federation of European Microbiological Societies FEMS Microbiol Rev 37 (2013) 336–363
Published by Blackwell Publishing Ltd. All rights reserved
Regulation of natural competence in Gram-negative bacteria 343

of alleles in populations of different Pseudomonas species recently ‘emerged from an organism of questionable path-
and strains. ogenicity to an infectious agent of importance to hospi-
Transformation levels not only varied significantly tals worldwide’ (Munoz-Price & Weinstein, 2008). More
among strains of P. stutzeri (Lorenz & Sikorski, 2000; importantly, this bacterium readily acquires resistance to
Sikorski et al., 2002), but they also varied across growth antibiotics, another trait linked with HGT.
phases (Carlson et al., 1983; Lorenz & Wackernagel, With respect to natural competence and transforma-
1990). Specifically, transformation frequencies for growth tion, most studies have been performed on Acinetobacter
in rich media peaked at the onset of the stationary phase, baylyi BD413 (Carr et al., 2003; representative strains of
when nutrients start to become limited (Lorenz & Wack- this species were earlier named Acinetobacter calcoaceticus
ernagel, 1990). Thus, to mimic the natural habitat of this BD4, A. calcoaceticus BD413, A. calcoaceticus ADP1, and
bacterium and to study nutrient situation and its effect Acinetobacter spp. strain ADP1; Young et al., 2005). The
on competence induction in P. stutzeri, Lorenz & Wack- transformation efficiency of A. baylyi BD413 (formerly
ernagel (1991) tested growth and transformation on soil- named A. calcoaceticus BD413) was shown to be notably
extract-containing medium. These authors demonstrated high, ranging from 0.1% to 0.7% of all cells in an auto-
that nutrients in those extracts were limited, allowing for trophy/prototrophy transformation assay (Juni & Janik,
the growth of only one or two generations. By supple- 1969). Furthermore, this bacterium is not fastidious about
menting the soil media with different combinations of the source of DNA; no species-specificity was observed
carbon sources, phosphate and ammonium, these with respect to DNA uptake (Lorenz et al., 1992; Palmen
researchers determined the effect of each nutrient on et al., 1993; Palmen & Hellingwerf, 1997).
growth and transformation. Limiting N, C or P stimu- Acinetobacter baylyi BD413 is always transformable,
lated transformation; this effect was most pronounced in though with large variations in efficiency throughout the
soil extracts spiked with pyruvate and phosphate but growth cycle (Cruze et al., 1979). Reports on the time of
lacking additional ammonium (Lorenz & Wackernagel, peak transformability are conflicting. The initial study
1991). The underlying regulatory mechanism remains suggested that ‘a peak of competency is found as the cul-
unknown. ture enters the stationary phase’ (Juni, 1978) and that
Soil microcosm experiments demonstrated that condi- transformation was less efficient in the exponential phase
tions supporting the efficient transformation of P. stutzeri (Juni & Janik, 1969). This finding was confirmed by
are readily encountered in the environment (Sikorski others who found that the appearance of transformants
et al., 1998). The same phenomenon was observed in was maximal at the end of the exponential growth phase
P. fluorescens. Interestingly, transformation frequencies for the soil bacteria Bacillus subtilis and A. baylyi BD413
were significantly higher in nonsterile soil samples than in (Lorenz et al., 1991). These authors also demonstrated
gamma-irradiated microcosms, possibly ‘due to the pres- that there was no active DNA release by competent
ence of an organic compound in soil that is in part Acinetobacter cells. Other groups provided evidence that
destroyed by soil sterilization’ (Demaneche et al., 2001). the highest transformation frequencies occurred early in
Even more strikingly, none of the in vitro conditions tested the exponential phase, which was consistent with rapid
induced competence in P. fluorescens (Demaneche et al., growth (Cruze et al., 1979). The latter finding was also
2001), illustrating once more that most of the environ- confirmed by several groups (Palmen et al., 1993, 1994;
mental signals that foster natural competence and transfor- Porstendörfer et al., 2000), who showed that natural
mation have not yet been identified. Thus, many bacteria transformation was maximally induced after the dilution
used in the laboratory might have the potential for compe- of an overnight culture into fresh medium or ‘after an
tence, although the trigger has not yet been elucidated. increase in nutrient availability, preceded by cessation of
growth’ (Palmen et al., 1994). In an earlier review, these
contradictory findings were resolved with the conclusion
Competence of Acinetobacter – is a nutrient
that competence ‘reaches its maximum during early to
boost required?
late log phase in A. calcoaceticus’ (Lorenz & Wackernagel,
Another well-studied, naturally competent, Gram-negative 1994). Based on the results of a study on early compe-
bacterium is Acinetobacter. Acinetobacter spp. belong to tence induction, Palmen et al. (1994) concluded that ‘the
the c-proteobacteria, order Pseudomonadales. Acinetobac- biological function of natural transformation in A. calco-
ter strains are abundant in soil and water (Warskow & aceticus is not to provide the cell with nutrients’ and that
Juni, 1972), where they have been determined to make ‘the regulation of competence development as a function
up at least 0.001% of the total culturable aerobic bacterial of the stringency of carbon, nitrogen, and phosphate limi-
population (Baumann, 1968). Acinetobacter strains, how- tation was just opposite to what one would expect if the
ever, are also frequently isolated from patients and have ‘nutrient supply hypothesis’ would apply’.

FEMS Microbiol Rev 37 (2013) 336–363 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
344 P. Seitz & M. Blokesch

Beate Averhoff and collaborators demonstrated that the tute a hotspot for host/pathogen HGT (Pontiroli et al.,
transcription of at least two of the competence genes, 2009). In summary, the induction of transformability of
comP and comB, did not correlate with the transforma- Acinetobacter by an upshift in nutrients may behave in a
tion pattern (Herzberg et al., 2000; Porstendörfer et al., manner contrary to what was observed for many other nat-
2000). Instead comP expression peaked in the late station- urally transformable bacteria in which competence and
ary phase, a finding that was also confirmed at the transformation are often linked with starvation signals.
protein level (Porstendörfer et al., 2000). ComB transcrip-
tional activation also turned out as growth phase depen-
Natural transformation in aquatic Vibrio
dent: expression slightly peaked after dilution of the
spp., with a focus on V. cholerae
bacteria in fresh medium followed by an immediate
decreased of comB transcription. Within mid-exponential Vibrio cholerae is a human pathogen and the causative
phase expression of comB resumed up to a constant and agent of cholera; however, this bacterium’s main niches
high level of comB transcription in late stationary phase are aquatic environments, such as rivers, estuaries, and
(Herzberg et al., 2000). Based on these data Herzberg coastal regions. Within these environments, V. cholerae is
et al. (2000) concluded that the DNA machinery is often found in association with chitinous zooplankton or
already present in late stationary phase; diluting the cells their molts (Colwell, 1996; Lipp et al., 2002; Pruzzo et al.,
into fresh medium then provides the required energy to 2008). This association results from the ability of
allow efficient DNA uptake and consequently transforma- V. cholerae and other aquatic Vibrio species to degrade
tion. This conclusion is consistent with other reports on the chitinous exoskeletons of zooplankton and use them
competence induction and transformation (Palmen et al., as a carbon and nitrogen source (for review see Keyhani
1994; Porstendörfer et al., 2000) and the finding that & Roseman, 1999). Because the abundance of plankton
transformation of A. baylyi BD413 is not inhibited by the changes significantly between plankton blooms, Vibrio
protein synthesis inhibitor chloramphenicol (Palmen & species regulate gene expression in accordance with nutri-
Hellingwerf, 1997). The group of B. Averhoff has also ent availability. Whether such environmental changes also
conducted work on the natural transformation of an unu- influence competence induction is discussed below.
sual Gram-negative bacterium, the thermophile Thermus
thermophilus. This bacterium is a record-holder with
HGT within the species V. cholerae
respect to natural transformability (Koyama et al., 1986)
and a detailed review on T. thermophilus transformation Comparative genomic hybridization experiments per-
was recently published (Averhoff, 2009). The impact and formed by John Mekalanos’s group revealed a high degree
basis of natural competence and transformation for sur- of conservation among V. cholerae strains (Dziejman
vival of Thermus in and adaptation to hot environments et al., 2002). This study was based on microarray hybrid-
has recently been analysed (Omelchenko et al., 2005; ization experiments and compared the strains’ genomes
Averhoff & Müller, 2010). to the first sequenced strain of V. cholerae, which was a
Based on these in vitro studies described earlier, it is dif- pandemic O1 El Tor strain (N16961; Heidelberg et al.,
ficult to determine the environmental signals that could 2000). Only presence/absence data about genes and ope-
trigger competence in Acinetobacter. Nielsen et al. (1997) rons were collected; no information about novel genetic
performed soil microcosm studies (‘in situ’) and observed material was retrievable with this experimental approach.
the induction of competence by nutrient upshifts, as well as The authors of this study identified a limited number of
a transformation-enhancing effect of phosphate. These genomic islands that were specific to the seventh pan-
authors concluded that ‘poorly transformable A. calcoaceti- demic strain and absent in earlier isolates, such as the
cus cells can be induced to undergo natural transformation representative strains of the classical biotype (the causa-
with chromosomal DNA in soil’ and that ‘their level of tive agent of the sixth and, likely earlier, cholera pandem-
competence is influenced by their metabolic state’ (Nielsen ics; Dziejman et al., 2002). Furthermore, this initial study
et al., 1997). Such transformability was maintained for sev- focused mainly on patient isolates and only investigated
eral hours after induction. A recent in situ study confirmed representatives of the serogroups O1 and O139. A follow-
that A. baylyi entered competence soon after the inocula- up study by Dziejman et al. broadened this analysis to
tion of leaves, as evaluated by the re-isolation of bacteria include pathogenic non-O1 and non-O139 V. cholerae
from the leaves and in vitro provision of transforming isolates. These strains were ‘quite divergent’ from the
DNA (Pontiroli et al., 2009). Furthermore, these authors V. cholerae representatives belonging to the O1 and O139
directly visualized transformants in situ using gfp reporter serogroups (Dziejman et al., 2005).
fusion and transforming DNA derived from transgenic Technologically advanced, large-scale WGS studies
plants. They concluded that the phytosphere might consti- made it possible to generate a clearer picture of SNPs and

ª 2012 Federation of European Microbiological Societies FEMS Microbiol Rev 37 (2013) 336–363
Published by Blackwell Publishing Ltd. All rights reserved
Regulation of natural competence in Gram-negative bacteria 345

horizontally acquired genetic material; these data enabled spread of cholera-toxin prophages, most notably from
the construction of phylogenetic trees from which to non-O1/non-O139 strains. These authors provided evi-
derive information about the evolutionary relationships dence that strains that were unable to produce infectious
among strains. For example, recent WGS studies on cholera-toxin phage particles could still undergo horizontal
V. cholerae suggested that several transcontinental trans- transfer of the prophage using natural transformation
mission events have occurred (Mutreja et al., 2011), sig- (Udden et al., 2008). Apart from these virulence-associated
nificantly contributing to the understanding of the recent gene clusters, the transfer of genes involved in sugar
emergence of this pathogen in an enormous cholera out- metabolism via chitin-induced natural transformation has
break in Haiti (Chin et al., 2010; Hendriksen et al., 2011; also been demonstrated for V. cholerae isolates from the
Mutreja et al., 2011). Rita Colwell and collaborators also Californian coast (Miller et al., 2007). Thus, natural com-
used WGS to understand the genetic diversity of 23 dif- petence and transformation of V. cholerae provide a
ferent V. cholerae strains isolated over the past 98 years. newly recognized mechanism by which this organism can
These authors identified 73 genomic islands (containing effectively acquire new genes, thereby adapting to chang-
five or more ORFs) in their study and thus provided ing environmental conditions. Ways in which the natural
strong evidence for ‘extensive lateral gene transfer in competence programme is regulated in V. cholerae are
V. cholerae’ (Chun et al., 2009). summarized below.
Natural competence has only recently been described
in V. cholerae (Meibom et al., 2005). The authors demon-
Competence induction in some bacteria
strated that during growth on chitinous surfaces – a com-
involves more than just one signal – the three
mon environmental niche for this bacterium (Lipp et al.,
interlinked regulatory pathways of V. cholerae
2002), V. cholerae can take up naked DNA and recom-
bine it into its genome (Meibom et al., 2005). As a mech-
Sensing the carbon source chitin and colonizing
anism of HGT, natural competence enables V. cholerae to
chitinous surfaces
acquire new genes, including those that specify patho-
genic features. Indeed, Blokesch and Schoolnik demon- The resolution of the genome sequence of V. cholerae
strated experimentally that the O1-to-O139 serogroup (Heidelberg et al., 2000) provided the starting point for
conversion could occur in a single step through a natural the investigation of regulatory circuits using microarray
transformation-mediated exchange of the large O-antigen expression profiling. This approach enabled the identifica-
cluster (> 32 kb exchanged for > 42 kb; Blokesch & tion of genes that were specifically activated during
Schoolnik, 2007). The transformants displayed all pheno- V. cholerae’s association with chitinous crab-shell frag-
types associated with the acquired gene cluster, including ments or purified soluble chitin oligosaccharides, the
the changed O-antigen chain of the LPS and the capsular so-called chitin-utilization programme (Meibom et al.,
material surrounding the cells (Blokesch & Schoolnik, 2004). Genes that specifically responded to chitin oligo-
2007). A stretch of homologous DNA located within the saccharides included those genes predicted to encode
O-antigen cluster of many different V. cholerae strains components for the biogenesis and function of a type IV
(the IS1358 element) enabled transformants with only pilus (Fullner & Mekalanos, 1999; Meibom et al., 2004),
partial exchange of the cluster (only the upstream or although this pilus has not yet been visualized. Compo-
downstream region of IS1358) to be obtained (Blokesch nents of type IV pilus complexes in naturally competent
& Schoolnik, 2007). In this way, natural transformation bacteria are hypothesized to participate in the transport
may contribute to a ‘reshuffling’ of O-antigen genes of DNA through the outer membrane and the peptidogly-
and thus to the creation of new serogroups of V. cholerae. can (reviewed by Chen & Dubnau, 2004; Box 1), but ‘it
That serogroup conversion of V. cholerae occurs fre- is currently unclear how the components interact to gen-
quently in nature was also supported by comparative erate movement of the incoming DNA through the
genomics (Chun et al., 2009). A study notably similar to cell envelope’ (Allemand & Maier, 2009). The chitin-
that conducted by Blokesch & Schoolnik (2007) was per- dependent induction of type IV pilus-encoding genes
formed 4 years later, and the authors demonstrated a cohered with natural competence and transformation,
carbotype conversion of Vibrio vulnificus based on chitin- demonstrating for the first time that V. cholerae is a natu-
induced transformation (Neiman et al., 2011). In this rally transformable bacterium (Meibom et al., 2005).
case, the transformation event led to the exchange of a But how is the chitinous surface sensed, and how is the
capsular polysaccharide locus in this organism (Neiman signal transmitted to induce gene-specific expression? In
et al., 2011). Udden et al. (2008) used a similar experi- this context, the induction of genes involved in chitin
mental approach to show that chitin-induced natural degradation is strictly dependent on a unique two-
competence of V. cholerae may be responsible for the component signalling system composed of a chitin hybrid

FEMS Microbiol Rev 37 (2013) 336–363 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
346 P. Seitz & M. Blokesch

Fig. 3. A complex, threefold regulatory network is involved in the natural transformation of Vibrio cholerae. As shown in Fig. 1, the induction of
natural competence and transformation in V. cholerae is linked with three of the common environmental cues: (1) the chitinous surface as the
sole carbon source for this bacterium and other Vibrio spp. (brown pathway); (2) CCR and the induction of competence after starvation for
preferred PTS-transported sugars (blue pathways); and (3) cell-to-cell communication, such as that exerted by the quorum-sensing systems (green
pathway). Left – the chitin pathway. Extracellular chitinase (e.g. ChiA-1 and ChiA-2 directly secreted by V. cholerae or chitinases from other
chitinolytic aquatic bacteria, which are abundant in this environment) degrades the insoluble N-acetylglucosamine (GlcNAc) polymer chitin and
releases diacetylchitobiose units as the major product (together with some triacetylchitotriose). These chitin oligomers are recognized by a sensory
histidine kinase, ChiS, and a signal transduction pathway subsequently leads to the induction of tfoX and tfoR expression. tfoR encodes for a
small regulatory RNA, which enhances the translation of the tfoX mRNA by freeing the Shine-Dalgarno (SD) site, as well as the start codon
(AUG) from a secondary stem loop structure formed by the 5′ UTR of the tfoX mRNA. TfoX, as the major regulator of transformation, is involved
in the induction of competence genes. This induction most likely involves pairing with the cAMP-CRP complex (grey double arrow), as TfoX itself
does not contain any obvious DNA-binding domain. Middle –CCR. In the absence of preferred PTS-transported sugars (such as glucose or the
chitin monomer GlcNAc), the PTS systems are not saturated, and the phosphoryl group is kept by enzyme II (EII). EII–P is a direct activator of the
cAMP-producing enzyme CyaA. Accumulating cAMP, together with its binding protein (CRP), leads to the induction of the competence genes.
This complex might interact with TfoX to allow for the binding of cAMP-CRP to competence-specific CRP-S sites. Right – high cell density, as
measured by QS, is required for competence induction and transformation. V. cholerae produces two main autoinducers: CAI-1 and autoinducer
2 (AI-2). At high cell density (HCD), the binding of the autoinducer to the respective receptors initiates a signalling cascade (simplified), which
conclusively results in the production of the major regulator of QS, HapR. HapR acts as repressor of the extracellular nuclease gene dns and as a
positive regulator of the competence genes comEA and comEC. The encoded proteins of all three genes are expected to be in direct contact
with transforming DNA; these proteins either induce the degradation of external DNA at low cell density (Dns) or are part of the DNA-uptake
machinery (ComEA and ComEC; see Box 1). Thus, QS fine-tunes the fate of the surrounding DNA. References on which the scheme is based on
are mentioned in the text.

sensor/kinase ChiS (Li & Roseman, 2004) and a thus far chitin-dependent competence-inducing conditions (M. Blok-
unidentified response regulator tentatively named ChiR esch, unpublished data).
(Fig. 3). Li & Roseman (2004) speculated that ChiS works The V. cholerae chitin utilization programme also indi-
in conjunction with an N,N′-diacetylchitobiose ABC-type cated that a gene encoding a protein with similarity to
transporter and that ChiS is activated upon the transport the transformation regulators of other naturally compe-
of the chitin degradation product (GlcNAc)2. The con- tent bacteria (TfoX/Sxy) was induced during growth on
nection between ChiS and the expression of the type IV chitin (Meibom et al., 2004). TfoX was later shown to be
pilus-encoding genes became obvious when the latter essential for natural transformation of V. cholerae (Mei-
genes were not upregulated in a V. cholerae chiS mutant bom et al., 2005). There is substantial evidence that TfoX
during growth on chitin (Meibom et al., 2004). Notewor- is downstream of ChiS in the chitin-dependent signalling
thy, a chiS mutant is not naturally transformable under cascade (Fig. 3). Expression of tfoX in trans or in cis

ª 2012 Federation of European Microbiological Societies FEMS Microbiol Rev 37 (2013) 336–363
Published by Blackwell Publishing Ltd. All rights reserved
Regulation of natural competence in Gram-negative bacteria 347

(Meibom et al., 2005; Lo Scrudato & Blokesch, 2012) transported sugars, including the chitin monomer GlcNAc,
relieved the requirement for chitin to induce the pilus- interfere with natural transformation (Blokesch, 2012).
specific genes and other genes involved in DNA uptake Such carbon sources are known to counteract against the
(Box 1), henceforth referred to as competence genes. Fur- intracellular accumulation of the secondary messenger
thermore, the transformation-negative phenotype of a cAMP (for review see Deutscher et al., 2006). The author
chiS mutant can be overcome by chitin-independent and of this study also demonstrated that the lack of cAMP or
low-level expression of tfoX (Lo Scrudato & Blokesch, of the CRP changes at least three levels of chitin-depen-
2012), which restores natural transformability in this dent natural competence induction in V. cholerae: chitin
strain (M. Blokesch, unpublished data). surface colonization, chitin degradation/metabolism and
Haruo Watanabe’s group demonstrated another link competence gene expression (Blokesch, 2012). Indeed,
between chitin-dependent transformation and TfoX pro- with respect to chitin metabolism, V. cholerae strains defi-
duction in V. cholerae. These researchers first confirmed cient in CCR showed significantly lower expression of
the transcriptional data of Meibom et al. (2004): in con- chiA-2 (Blokesch, 2012). ChiA-2 and chiA-1 encode two
trast to (GlcNAc)n oligomers (n  2), the chitin mono- redundant extracellular chitinases, and at least one of these
mer GlcNAc is not sufficient to induce tfoX expression chitinases is required for growth on chitin (Meibom et al.,
(Yamamoto et al., 2010). In addition, Watanabe et al. 2004). The lack of expression of such chitin degradation
discovered a translational component of chitin-induced enzymes might explain why V. cholerae mutants that are
TfoX production (Yamamoto et al., 2010). The latter defective for CCR, such as a crp minus strain, are unable
finding was supported by the discovery of a chitin- to grow with GlcNAc oligomers as the sole carbon source
induced small RNA (named TfoR), which positively con- and are unable to colonize the chitinous surface (Blokesch,
tributes to the translation of the tfoX mRNA (Yamamoto 2012).
et al., 2011; Fig. 3). These authors showed in vitro that The expression of the competence genes also being
the 102-nucleotide sRNA TfoR activates the translation of directly dependent on CCR was further proven by uncou-
the tfoX mRNA, most likely by freeing the SD motif and pling natural competence induction from chitin surface
the start codon from a secondary structure formed by the colonization and chitin metabolism (Blokesch, 2012;
5′ UTR of the tfoX mRNA in the absence of TfoR. This Lo Scrudato & Blokesch, 2012). Using different chitin-
mechanism mimics the analogous mechanism in H. influ- independent competence-inducing conditions and differ-
enzae (Cameron et al., 2008): a 5′ stem loop structure ent readouts (reverse transcription followed by PCR and
formed within the tfoX/sxy mRNA is significant in initiat- transcriptional reporter fusions for Blokesch, 2012 and Lo
ing translation, but the release of this stem loop by base Scrudato & Blokesch, 2012, respectively), a change in
pairing with the sRNA TfoR seems to be unique to competence gene expression became obvious when com-
V. cholerae and, possibly, other Vibrio spp. (Yamamoto paring wild-type V. cholerae with CCR-deficient strains.
et al., 2011). Furthermore, a V. cholerae strain that lacked the cAMP-
degrading enzyme cAMP phosphodiesterase (cpdA)
showed enhanced frequencies of natural transformation
CCR and the requirement for cAMP and CRP
(Lo Scrudato & Blokesch, 2012). Based on these and
in V. cholerae
other findings, Lo Scrudato & Blokesch (2012) concluded
When chitin-induced natural competence was first that an imbalanced intracellular cAMP pool affects com-
described, the authors demonstrated that glucose petence induction at the transcriptional level. How these
prevented natural transformation. They hypothesized two regulatory circuits – chitin-dependent induction of
therefore that CCR might play a role in competence TfoX and CCR – are connected is not fully understood.
induction (Meibom et al., 2005; Figs 1 and 3). Vibrio However, a current hypothesis is that the transformation
cholerae takes advantage of the abundance of zooplankton regulator TfoX can act only in conjunction with CRP.
in its natural environment. Zooplankton have chitinous This speculation is based on the suggested CRP-Sxy inter-
exoskeletons, which provide a nutritious surface that can action as described earlier for H. influenzae. Indeed, based
serve as the sole carbon source for V. cholerae and can on published expression data for V. cholerae grown on
induce natural competence (Fig. 1). Therefore, the link chitin surfaces or tfoX overexpression (Meibom et al.,
between CCR and chitin-induced natural competence was 2004, 2005), competence-specific CRP-S sites have also
further investigated (Blokesch, 2012). In this study, the been predicted in silico by Cameron and Redfield for
hypothesis that CCR is involved in the regulation of V. cholerae (Cameron & Redfield, 2006). It might be
natural competence was further strengthened by showing interesting to note that cAMP-CRP also positively regu-
that not only glucose but also other phosphoenolpyr- late the expression of the integron-specific integrase gene
uvate : carbohydrate phosphotransferase system (PTS)- in V. cholerae (Baharoglu et al., 2012), which might avoid

FEMS Microbiol Rev 37 (2013) 336–363 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
348 P. Seitz & M. Blokesch

the need to homologously recombine the incoming trans- More importantly, the genes that are indeed linked with
forming DNA into the chromosome; instead, the newly QS (comEA and comEC) are all extremely relevant for
acquired DNA could be integrated into the gene-captur- transformation because the encoded proteins are hypothe-
ing integron island (Cambray et al., 2010). sized to directly interact with the transforming DNA (Box
1). Indeed, Dns degrades the DNA at low cell density,
whereas ComEA and ComEC are produced at high cell
Bacterial communication is key for competence
density and present major components of the DNA-
induction in V. cholerae
uptake machinery (Box 1; Suckow et al., 2011), thereby
The first suggestion of the involvement of quorum sensing directly interacting with the incoming DNA in V. chole-
(QS) in natural competence and transformation (Fig. 3) rae. Thus, in conclusion, QS acts as a switch from extra-
was based on the finding that V. cholerae was transform- cellular DNA degradation to DNA uptake in V. cholerae
able in a more efficient manner after an extended growth (Lo Scrudato & Blokesch, 2012).
period on chitin surfaces, which is in accordance with Based on HapR being involved in the natural compe-
increased cell densities (Meibom et al., 2005). Blokesch & tence and transformation of V. cholerae, several research
Schoolnik (2008) followed up on this finding and groups became interested in the question of how the
described the connection between QS and natural compe- autoinducers of V. cholerae contribute to this regulatory
tence and transformation in further detail. They demon- circuit. Based on work by Bonnie Bassler and collabora-
strated that the main regulator of QS, HapR, which is tors, two main autoinducers have been identified and have
only produced at high cell density, represses the gene been allocated a specific role in V. cholerae. Cholera
encoding the extracellular nuclease Dns (Blokesch & autoinducer 1 (CAI-1), a (S)-3-hydroxytridecan-4-one
Schoolnik, 2008; Fig. 3). This repression is an essential (Higgins et al., 2007), is thought to play a role as an intra-
step in the regulation of natural transformation because species communication agent. By contrast, AI-2, a furano-
the nuclease Dns degrades surrounding DNA, thereby syl borate diester (Chen et al., 2002), serves as a ‘universal
destroying any potential transforming material (Blokesch signal’ in bacteria as this molecule is produced and sensed
& Schoolnik, 2008). Recently, other researchers also con- by many bacteria and therefore allows interspecies com-
firmed the HapR-dependent down-regulation of dns munication (Xavier & Bassler, 2003). Comparing V. cholerae
(Seper et al., 2011). mutants devoid of the capacity to synthesize CAI-1, AI-2
HapR also acting as a positive regulator of at least a or both autoinducers, the group of Melanie Blokesch
subset of competence genes, such as the periplasmic showed that in the absence of CAI-1, natural transforma-
DNA-binding protein encoding gene comEA, became tion is (almost) completely abolished in experiments
obvious early on (Meibom et al., 2005; Blokesch & mimicking the natural chitinous environment (Suckow
Schoolnik, 2008). This speculation was initially based on et al., 2011) and that transformation was consistently
microarray expression data (Meibom et al., 2005) and undetectable under homogenous competence-inducing
V. cholerae hapR/dns double knockout mutants being conditions (Lo Scrudato & Blokesch, 2012). Furthermore,
transformable at an c. 100-fold lower frequency than a the authors of these studies were never able to detect any
dns single knockout mutant, even though the degradation transformants in the absence of both autoinducers (CAI-1
of extracellular DNA was fully abolished (Blokesch & and AI-2), a phenotype that mirrors hapR minus strains
Schoolnik, 2008). The expression of comEA being indeed (Suckow et al., 2011; Lo Scrudato & Blokesch, 2012). Not
regulated in a QS-dependent manner was nicely demon- surprisingly no HapR protein was detectable in CAI-1/
strated using transcriptional fusion with a luciferase-based AI-2 synthase-deficient cells (Lo Scrudato & Blokesch,
readout (Antonova & Hammer, 2011) and also more 2012). This transformation-negative phenotype of autoin-
recently by detecting comEA expression using transcrip- ducer-deficient V. cholerae cells was not restorable by
tional fusions with fluorescent proteins (Lo Scrudato & cross-feeding of solely AI-2 from co-cultured bacteria, in
Blokesch, 2012). The latter study significantly extended contrast to the efficient restoration of transformation by
this finding by not only monitoring the expression of cross-fed CAI-1 (Suckow et al., 2011). Suckow et al.
comEA but also other competence genes, such as pilA therefore concluded that the intraspecies autoinducer
(Lo Scrudato & Blokesch, 2012; Fig. 3). Furthermore, Lo CAI-1 might be similar to competence pheromones
Scrudato and Blokesch investigated a plethora of compe- described in Gram-positive bacteria and that such a
tence genes with respect to any potential QS-dependent regulation might increase the chances of taking up
regulation. The results of this study were remarkable species-specific DNA (Fig. 2). However, even though
because they provided good evidence for only a small another study illustrated the same tendency, it did not
subset of competence genes of V. cholerae being coregu- show such a strict CAI-1-dependency. More specifically,
lated by TfoX and QS (Lo Scrudato & Blokesch, 2012). Antonova & Hammer (2011) concluded in their study

ª 2012 Federation of European Microbiological Societies FEMS Microbiol Rev 37 (2013) 336–363
Published by Blackwell Publishing Ltd. All rights reserved
Regulation of natural competence in Gram-negative bacteria 349

that ‘Vibrio-specific CAI-1 appears to play a major role and transformation in V. cholerae. This process also con-
and interspecies AI-2 a minor role, suggesting that nects a process indicative of preferred carbon source star-
induction of DNA uptake may not be restricted exclu- vation, or CCR, to the induction of competence. This
sively to a response to autoinducers produced by Vibrio link might reflect a similarity to the function of natural
species, but that HGT may also be promoted by AI-2 transformation in S. pneumoniae ‘as a rescue process’
derived from non-Vibrio members of a biofilm’. These (Prudhomme et al., 2006).
authors also showed that strains lacking both autoinducers In a recent review by Ng and Bassler, the authors wrote
(CAI-1 and AI-2) were readily transformable and thereby ‘One oddity is that homologues of cqsA and cqsS, called
significantly above the detection level of a nontransform- lqsA and lqsS respectively, exist in the distantly related bac-
able hapR mutant strain (Antonova & Hammer, 2011). terium L. pneumophila. LqsA produces 3-hydroxypentad-
Based on these data, it can be hypothesized that the dis- ecan-4-one; a molecule with a longer hydrocarbon chain
crepancy in autoinducer dependency in these different than CAI-1 (Spirig et al., 2008)’ (Ng & Bassler, 2009).
V. cholerae strains is not due to a different connection Interestingly, a recent analysis of six fully sequenced
between the QS regulator HapR and natural transforma- L. pneumophila genomes suggested that the genomes were
tion but most likely reflects a difference in the QS circuit- highly dynamic, as a result of extensive HGT and recombi-
ries. Further studies will be required to confirm or refute nation (Gomez-Valero et al., 2011). Furthermore, ‘L. pneu-
this hypothesis. mophila ranks as the prokaryote with the widest variety of
eukaryotic-like proteins’, all of which may have been
acquired from the host by HGT (Cazalet et al., 2004). At
The induction of competence in V. cholerae
this point, it is tempting to speculate that a-hydroxyketone
compared with the other naturally
signalling molecules are commonly involved in the regula-
transformable bacteria
tion of natural competence. Thus, it will be interesting to
Based on the results of CAI-1 dependency for natural see whether the Legionella autoinducer-1 (LAI-1; Spirig
transformation in V. cholerae, CAI-1 can be considered a et al., 2008) is also required for natural competence and
competence pheromone (Suckow et al., 2011; Lo Scruda- transformation in this bacterium. It had long been appreci-
to & Blokesch, 2012). Indeed, this molecule might per- ated that L. pneumophila is naturally transformable (Stone
form a function similar to a function proposed for & Kwaik, 1999), and recent studies have provided evidence
Gram-positive bacteria (for a seminal review on compe- that competence might fulfil a similar function in this
tence induction in Gram-positive bacteria see Claverys organism as in S. pneumoniae, replacing the absent SOS
et al., 2006). For example, competence induction in response (Charpentier et al., 2011).
S. pneumoniae is not solely dependent on the cell density
and thereby on the passive accumulation of the compe-
The induction of natural competence
tence-stimulating peptide (CSP), a competence phero-
in noncholera Vibrio species
mone. Instead, the production of CSP varies with
changing environmental conditions (Claverys et al., 2000, Studies in the 1990s had already suggested that ‘marine
2006). Claverys et al. showed that DNA-damaging antibi- Vibrio spp.’ were naturally transformable (Frischer et al.,
otics induce the expression of the competence regulon in 1990, 1993; Jeffrey et al., 1990; Paul et al., 1992). These
a CSP-dependent manner in S. pneumoniae. These aquatic isolates were assigned to the genus Vibrio and
authors argued that CSP serves as a stress signal and can were most similar to V. campbelli based on the biochemi-
therefore be considered an alarmone rather than a quo- cal features described in Bergey’s manual; however, fol-
rum-sensing effector (Prudhomme et al., 2006). Prud- low-up studies on this phenomenon provided evidence
homme et al. (2006) therefore concluded that ‘CSP could that the isolates belonged to the genus Pseudomonas, a
thus play a crucial role in generating genetic diversity finding based on 16S-rRNA gene analysis (Frischer et al.,
under stress conditions for a species that seems unable to 1996). Thus, the natural transformability of Vibrio spp.
rely on inducible mutagenic repair (such as the SOS had not been demonstrated before Meibom et al. (2005)
response)’. The hypothesis that CAI-1 in V. cholerae is described chitin-induced natural transformation in
similar to competence pheromones is supported by the V. cholerae.
fact that the synthesis of CAI-1 by the synthase CqsA is Following the first studies on natural competence in
also not constitutive in V. cholerae. In fact, Liang et al. V. cholerae (Meibom et al., 2005; Blokesch & Schoolnik,
(2007, 2008) demonstrated that CCR is involved in the 2007, 2008), other researchers have found that V. cholerae
post-transcriptional regulation of cqsA expression in is not the only representative of the genus Vibrio to be
V. cholerae, providing another connection among the naturally transformable; natural transformation has also
three regulatory pathways that drive natural competence been demonstrated in V. vulnificus (Gulig et al., 2009),

FEMS Microbiol Rev 37 (2013) 336–363 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
350 P. Seitz & M. Blokesch

Vibrio parahaemolyticus (Chen et al., 2010) and Vibrio with respect to chitin-dependent induction and the
fischeri (Pollack-Berti et al., 2010). The first two studies requirement for TfoX. The QS circuits, by contrast, differ
used a modified version of the natural transformation among these organisms, and not all Vibrio spp. contain a
protocol initially described for V. cholerae (Meibom et al., CqsA/CqsS system (for recent review see Milton, 2006).
2005; Blokesch & Schoolnik, 2007) to modify the bacteria Additionally, not all Vibrio spp. that do contain a CqsA/
genetically (Gulig et al., 2009; Chen et al., 2010). Because CqsS system produce the same autoinducer. For example,
this protocol is based on competence induction on chitin- work by Bonnie Bassler’s group demonstrated that the
ous surfaces, a common theme of chitin-based induction Vh-CAI-1 autoinducer of Vibrio harveyi differs slightly
seems clear. Indeed, it was experimentally shown that from its counterpart in V. cholerae in that the former
many other species of the genus Vibrio grow on chitin contains an 8-carbon tail instead of a 10-carbon tail
and that most of them contain proteins homologous to (Ng et al., 2011). An 8-carbon tail CAI-1 was also present
those identified in the V. cholerae chitin utilization pro- in spent supernatants of other Vibrio spp., including
gramme (Meibom et al., 2004 and as summarized in V. parahaemolyticus, Vibrio alginolyticus, Vibrio anguilla-
Hunt et al., 2008). Thus, the chitinous surface may be a rum and Vibrio furnissii (Ng et al., 2011). The authors of
common niche for Vibrio spp., and chitin-induced natu- this study concluded that ‘different Vibrio species display
ral competence may be widespread. unique production and detection profiles for the CAI-1
This finding was further emphasized by the group of family of molecules’. One could hypothesize that ‘the
Ned Ruby, who described the induction of natural com- forces that drove these two species [= V. cholerae and
petence in the symbiotic Vibrio, V. fischeri (Pollack-Berti V. harveyi] to evolve different signalling specificities’ (Ng
et al., 2010). These researchers provided evidence that as et al., 2011) might be linked with natural transformation
in V. cholerae, natural competence in V. fischeri is and species-specificity of the system (Fig. 2). Further
induced in the presence of chitin oligomers and in a studies will be required to elucidate the connection
TfoX-dependent manner. Interestingly, Pollack-Berti et al. between QS and the natural transformability in nonchol-
identified a second TfoX-like protein, named TfoY, in era Vibrios and especially in those Vibrios that do not
V. fischeri. Indeed, all fully sequenced Vibrionaceae con- contain CqsA/CqsS systems.
tain these two paralogues of TfoX (Pollack-Berti et al.,
2010). The authors of this study also demonstrated that
The induction of natural transformation
the two paralogs were unable to compensate fully for the
in plant pathogenic bacteria
loss of the counterpart and accordingly that they influ-
ence natural transformation in distinct manners. Interest-
Ralstonia solanacearum – a large host range
ingly, the corresponding TfoY counterpart of V. cholerae
plant pathogen takes up DNA
had been discovered earlier during a search for RNA
motifs and riboswitches in bacteria because it contains a Ralstonia solanacearum is a representative bacterium of
Genes for the Environment, for Membranes, and for the b-proteobacteria class, family Burkholderiaceae. Most
Motility (GEMM) motif (Weinberg et al., 2007; Sudarsan importantly, R. solanacearum is a major plant pathogen
et al., 2008). In this context, Weinberg et al. described ‘because of its aggressiveness, large host range, broad
that based on the conserved domain database (CDD), geographical distribution and long persistence in soil and
V. cholerae contains two genes belonging to the COG3070 water environments’ as recently reviewed (Genin, 2010).
family (encompassing TfoX-domains). As TfoY (named After the genome of R. solanacearum was sequenced
tfoXGEMM in Weinberg et al., 2007) but not tfoX (Salanoubat et al., 2002 and reviewed in Genin & Boucher,
contained this structured GEMM motif, the authors con- 2004), analysis revealed some interesting features. First, as
cluded ‘in V. cholerae and related bacteria, GEMM might in V. cholerae (Heidelberg et al., 2000), the genome of
participate in chitin-induced competence, or even regu- R. solanacearum is split into two chromosomes of
late competence in environments not containing elevated unequal sizes (3.7 and 2.1 megabases). Second, the gen-
chitin concentrations’. In line with the latter hypothesis, ome displayed a mosaic structure with respect to the
TfoY does not play an obvious role in chitin-induced nat- G+C content throughout several kilobases of both chro-
ural competence of V. cholerae. Indeed, a V. cholerae mosomes (7% of the total genomes differed). Together
knockout strain of tfoY is fully transformable during with the fact that the encoded genes differed in codon
growth on chitinous surfaces and artificial expression of usage from the rest of the bacterium, this difference was
tfoY does not render V. cholerae naturally competent indicative of HGT (Genin & Boucher, 2004). The authors
(M. Blokesch, unpublished data). of this review concluded ‘that [these regions] could play
In conclusion, the regulation of natural competence an important role in the rapid adaptation of the bacte-
and transformation in Vibrio spp. seems to be consistent rium to the change of ecological niche’ (Genin & Bou-

ª 2012 Federation of European Microbiological Societies FEMS Microbiol Rev 37 (2013) 336–363
Published by Blackwell Publishing Ltd. All rights reserved
Regulation of natural competence in Gram-negative bacteria 351

cher, 2004). Because R. solanacearum is naturally trans- 2011). This bacterium belongs to the class c-proteobacte-
formable, this mode of HGT might have contributed sig- ria, family Xanthomonadaceae. The lifestyle of X. fastidi-
nificantly to the mosaic structure of the genome. Indeed, osa has recently been reviewed and the authors
two recent studies provided evidence that large regions of mentioned that ‘strains of X. fastidiosa have been associ-
DNA (from 30 to 90 kb and up to 80 kb, respectively) ated with a large number of diseases, many causing great
can be integrated into the recipient’s genome (Coupat economic losses’ (Chatterjee et al., 2008). This bacterium
et al., 2008; Coupat-Goutaland et al., 2011). causes disease in many plants, including grapes and citrus
With respect to the regulation of natural competence fruits. Transmission of this bacterium is mediated by
in R. solanacearum, transformation frequencies peaked in insects, such as sharpshooters (Chatterjee et al., 2008).
the middle of the exponential phase and dropped quickly The ability to exchange genes horizontally may benefit
afterwards (Bertolla et al., 1997). Furthermore, as is the X. fastidiosa bacteria and ‘potentially permit them to
case for many other naturally competent bacteria, growth explore a wider variety of host plants’ (Kung & Almeida,
on minimal medium reflecting limiting growth conditions 2011).
permitted higher transformation efficiencies than those The information regarding when and how natural
observed in rich medium (Bertolla et al., 1997). Bertolla competence in Xylella is induced is still limited; however,
et al. (1999) also demonstrated that natural transforma- Kung & Almeida (2011) demonstrated that cell growth
tion occurs in situ (e.g. in planta). During cell-host inter- affects the number of transformants, and transformants
action, natural transformation was most efficient while were obtained more efficiently if the transforming DNA
R. solanacearum cells were multiplying within the plant was provided upon entry into exponential phase, with the
vessels (Bertolla et al., 1999). efficiency declining significantly afterwards. These authors
Ralstonia solanacearum also harbours more than one also demonstrated that methylated plasmid DNA served
quorum-sensing system (Whitehead et al., 2001). Specifi- better as transforming DNA than its unmethylated coun-
cally, this species contains a LysR-type regulator, PhcA, terpart, suggesting that R-M systems might play a role in
which regulates the production of extracellular polysac- the transformation of X. fastidiosa (Kung & Almeida,
charide (EPS) and extracellular enzymes (Brumbley et al., 2011) comparable with what was demonstrated for
1993). PhcA is subject to regulation by the two-component H. pylori (as described above; Humbert & Salama, 2008;
system, which responds to the autoinducer 3-hydroxypalmitic Humbert et al., 2011). Finally, the authors observed a
acid methyl ester (3OH PAME; Clough et al., 1997; medium-specificity of natural transformation because
Flavier et al., 1997). This autoinducer is most likely syn- transformation occurred only in nutrient-limited medium
thesized by the synthase PhcB, which exhibits homology (Kung & Almeida, 2011). Thus, the induction of natural
to small-molecule SAM-dependent methyltransferases competence seems to be linked with the nutritional status
(Flavier et al., 1997). Interestingly, the CAI-1 synthase of the cell in X. fastidiosa. Chitin has been proposed as a
CqsA of V. cholerae also uses SAM as a substrate (as do carbon source for X. fastidiosa upon entry into insect vec-
many other autoinducer synthases; Wei et al., 2011), tors (e.g. leafhoppers), and chitinolytic activity has been
and, at first glance, CAI-1 does show similarities to the demonstrated in vitro (Killiny et al., 2010). Thus, it is
3OH PAME of R. solanacearum. Evidence that, as in tempting to speculate that a co-regulation between chitin
V. cholerae, this quorum-sensing system is also involved utilization and CCR, as described earlier for V. cholerae,
in the regulation of natural transformation was generated might also be involved in competence induction in
by a recent study by Kang et al., who demonstrated that X. fastidiosa.
the gene encoding the major pilin subunit of a type
IV pilus, pilA, is essential for natural transformation of
Evolution in an evolutionary system –
R. solanacearum and is regulated in a cell density-dependent
what benefits does natural
manner. Specifically, these authors demonstrated that the
transformation bring to an organism?
expression of pilA decreases at high cell density and that
this downregulation is dependent on the major regulator The question of why natural transformation exists
of QS, PhcA (Kang et al., 2002). remains the subject of ongoing debate. Three hypotheses
are frequently discussed: ‘DNA for food’, ‘DNA for
repair’, and ‘DNA for evolution’. These hypotheses are
Another plant pathogen, Xylella fastidiosa,
not necessarily mutually exclusive, but they are not
was only recently discovered to be naturally
equally supported by the data. This topic has recently
transformable
been extensively reviewed elsewhere (Michod et al., 2008;
Another plant pathogen, X. fastidiosa, has also been iden- Vos, 2009); however, due to the strong link between the
tified as being naturally transformable (Kung & Almeida, induction of natural competence by environmental signals

FEMS Microbiol Rev 37 (2013) 336–363 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
352 P. Seitz & M. Blokesch

and the role transformation may play in bacteria, we however, this ability was disrupted in E. coli mutants
summarize some of the major arguments below. lacking genes that encode for potential competence-
related proteins. The authors of this and a follow-up
study identified eight genes encoding proteins that are
DNA for food
between 12% and 74% identical to the H. influenzae
The role of DNA as a source of nutrients was initially counterparts involved in natural competence (Finkel &
proposed by Redfield (2001). She wrote, ‘the simplest Kolter, 2001; Palchevskiy & Finkel, 2006). In the absence
model thus might be that the nutrients that transforma- of these genes, most of these strains encountered a sta-
tion reliably provides pay the evolutionary bills (and have tionary phase competition defect during co-culture with
been responsible for the evolution of its regulation), and the parental wild-type bacteria. The authors of this study
as a bonus the cell gets the occasional benefits of recom- concluded that taking up DNA for nutritional purposes,
bination and repair’ (Redfield, 1993b). Among others, the ‘particularly when that DNA is heterologous and less
main arguments put forward by Redfield for a role of likely to recombine onto the chromosome’ (Finkel &
transformation as ‘Genes for Breakfast’ were as follows: Kolter, 2001; Palchevskiy & Finkel, 2006), might confer a
(1) DNA as food provides a direct short-term advantage, significant advantage even over the acquisition of a bene-
whereas a role of novel genes in evolution might be ficial gene by HGT. However, because most of the
selected only in the long run. (2) H. influenzae and homologous proteins identified in E. coli and other proteo-
B. subtilis react to nutritional limitations when inducing bacteria (Palchevskiy & Finkel, 2006) resemble the type
competence (Redfield, 1993a, b). Notably, this is not the IV pilus part of the DNA-uptake machinery, the question
case for S. pneumoniae. In this species, nutrient starvation arises as to whether such DNA indeed reaches the cyto-
has never been observed to induce competence (Claverys plasm as linear ssDNA, as occurs in naturally competent
et al., 2006); instead, during growth in rich medium the bacteria. Alternatively, the type IV pilus-like structure
bacteria acquire competence in early log phase and main- may assist in recruiting free dsDNA into the periplasm
tain it only over a short period of time or differentially and thus facilitate transport across the outer membrane
said ‘competence is induced in times of feast rather than through the secretin PilQ/HofQ. DNA might be further
famine’ (Claverys & Havarstein, 2007). (3) Neither H. degraded in the periplasm into nucleosides, which are
influenzae nor B. subtilis induce competence upon DNA subsequently taken up into the cytoplasm by specific
damage. (4) Unrelated DNA can be used as a nutrient nucleoside transporters to serve as a source of carbon and
source, especially because it could not recombine homol- energy. Thus, whereas the first part of this process would
ogously into the chromosome. (5) The addition of ribo- resemble natural competence-induced DNA uptake and
nucleotide monophosphates, most notably AMP and involve type IV pilus-like protein components, DNA
GMP, to competence-inducing starvation medium transport across the inner membrane with concomitant
reduced the natural transformation in H. influenzae by degradation of one strand might not be identical in
more than two orders of magnitude and significantly ‘nutritional competence’ (Palchevskiy & Finkel, 2006).
reduced the expression of competence genes in this Indeed, DNA uptake is a 2-step process in naturally
organism (MacFadyen et al., 2001). These authors argued competent H. pylori (Stingl et al., 2010); however, as dis-
that the depletion of purines within the cell induces com- cussed earlier, H. pylori uses a T4SS, not a type IV
petence and that the incoming DNA could subsequently pilus-like structure, to shuffle the DNA across the outer
replenish the purine pool. Interestingly, this effect was membrane. But there are good indications that this 2-step
not observable for desoxyribonucleotide monophosphates, DNA-uptake process also holds true for other Gram-
triphosphates, or the free bases (MacFadyen et al., 2001). negative bacteria, as demonstrated in a recent review
(6) The poor-quality DNA derived from dead cells might (Kru ger & Stingl, 2011). Furthermore, V. cholerae regu-
not be suitable for transformation-mediated evolution lates the competence genes required for DNA movement
(Redfield, 1988; Redfield et al., 1997). But certain competent across the outer membrane differentially from the compe-
bacteria kill their (non-or not yet competent) siblings tence genes whose products are involved in DNA translo-
within the population, whereas other bacteria actively cation across the periplasmic space and the inner
donate DNA through a T4SS (Hamilton et al., 2005; membrane (Lo Scrudato & Blokesch, 2012).
Hamilton & Dillard, 2006) or through a currently Other findings also oppose the ‘DNA for food’ hypoth-
unknown mechanism (Stewart et al., 1983), suggesting esis. For example, David Dubnau stated in a review from
that not all transforming DNA is of poor quality. 1999 that B. subtilis possesses a powerful extracellular
Recent studies on E. coli support Redfield’s work. nuclease and adequate uptake systems for DNA degrada-
Finkel & Kolter (2001) provided evidence that E. coli can tion products (Dubnau, 1999). Extracellular nucleases
grow with DNA as the sole source of carbon and energy; have also been described for Vibrio cholerae (Newland

ª 2012 Federation of European Microbiological Societies FEMS Microbiol Rev 37 (2013) 336–363
Published by Blackwell Publishing Ltd. All rights reserved
Regulation of natural competence in Gram-negative bacteria 353

et al., 1985; Focareta & Manning, 1987). Most notably, incoming ssDNA is protected against degradation in
the main nuclease in this organism, Dns, is oppositely naturally competent bacteria by competence-specific
regulated from the competence genes that are directly proteins such as DprA (Mortier-Barriere et al., 2007).
involved in DNA uptake (Blokesch & Schoolnik, 2008; This mechanism is also more consistent with a role of the
Lo Scrudato & Blokesch, 2012), and Dns is a major transforming DNA in DNA-repair processes or in the
inhibitor of natural transformation in V. cholerae because donation of new alleles/genes.
it degrades the transforming material around the cell
(Blokesch & Schoolnik, 2008). A role of this nuclease in
DNA for repair
the utilization of DNA as a nutrient source has been sug-
gested (Blokesch & Schoolnik, 2008) and was experimen- Arguments for the repair hypothesis are principally based
tally supported with respect to the utilization of DNA as on the following facts:
a phosphate source (Seper et al., 2011). Interestingly, the The ability to take up DNA in some Gram-negative,
crystal structure of a concentrative nucleoside transporter naturally transformable bacteria is highly biased towards
of V. cholerae (NupC) has recently been solved. This pro- genetic material from the same or closely related species.
tein uses a sodium-ion gradient for nucleoside transport Various strategies have evolved to this end (Fig. 2). For
across the inner membrane (Johnson et al., 2012) and example, N. gonorrhoeae and H. influenzae discriminate
may be involved in the uptake of nucleotides released by between self and foreign DNA through the recognition of
the extracellular nuclease Dns. DUS that are overrepresented in their own genomes
Another important point that undermines the DNA for (Danner et al., 1980; Fitzmaurice et al., 1984; Elkins
food hypothesis is the energy associated with DNA uptake et al., 1991). Vibrio cholerae, in contrast, does not dis-
itself. As mentioned earlier and discussed in detail else- criminate between self and foreign DNA at the level of
where (Chen & Dubnau, 2004; Allemand & Maier, 2009; the DNA uptake (Suckow et al., 2011). Because compe-
Burton & Dubnau, 2010; Allemand et al., 2012), the tence induction in this organism is tightly linked with an
DNA-uptake machinery is most likely a multiprotein accumulation of the species-specific autoinducer CAI-1
complex (Box 1). Although the composition and mode of (Suckow et al., 2011; Lo Scrudato & Blokesch, 2012), spe-
action of this complex has not fully been elucidated, the cies-specific DNA is highly likely to reach the cytosol. In
complex is assumed to resemble type IV pili (with the contrast, H. pylori does not display any preference for
exception of H. pylori, as discussed earlier). Such type IV species-specific DNA. This assumption is based on the
pilus-like structures (or shortened forms, also known as fact that competence is constitutive in this organism and
pseudopili; Pugsley, 1993; Chen & Dubnau, 2004) allow that DUS-dependent sorting does not occur at the level
for the transport of DNA across the peptidoglycan layer of the DNA-uptake machinery. However, recent data has
and/or the outer membrane of Gram-positive and Gram- indicated that a mechanism based on R-M systems might
negative bacteria, respectively. An inner membrane chan- control DNA uptake in H. pylori, as explained above
nel subsequently allows translocation of the DNA across (Aras et al., 2002; Humbert & Salama, 2008; Humbert
the inner membrane; this structure is probably conserved et al., 2011). This system may also ensure the species-
across all naturally transformable bacteria (Draskovic & specificity of transforming DNA by recognizing and
Dubnau, 2005; Stingl et al., 2010; Suckow et al., 2011). degrading foreign genetic material, and protecting the
Consistent with the resemblance of the components, the genome from foreign DNA (Fig. 2; Aras et al., 2002;
forces generated by type IV pilus retraction and DNA Humbert & Salama, 2008; Humbert et al., 2011). Another
uptake were in the same range for both systems and rep- source for species-specific DNA may be fratricide. More
resent some of the strongest linear motors characterized precisely, bacterial fratricide is associated with natural
to date (Merz et al., 2000; Maier et al., 2002, 2004). As competence of S. pneumoniae (Guiral et al., 2005; Havar-
recently reviewed by Berenike Maier and others (Maier, stein et al., 2006 and recent review by Claverys & Havar-
2005; Allemand & Maier, 2009; Allemand et al., 2012) stein, 2007) and probably also in H. pylori (Dorer et al.,
such ‘directed DNA translocation is often energetically 2010; Fig. 2). In this context, bacterial cells of the same
unfavourable and requires an active process that uses population are killed to provide transforming DNA.
energy, namely the action of molecular motors’ (Alle- Based on several examples of the intentional killing of
mand et al., 2012). Thus, the question arises as to bacterial siblings, Gilmore & Haas (2005) concluded that
whether the use of transforming DNA as an energy ‘the selective lysis of siblings by a subpopulation of bacte-
source would be able to supply enough energy to rial cells appears to be a highly evolved and complex pro-
compensate for the consuming uptake process and still cess’.
provide sufficient extra energy to be more cost-effective Based on these important points, the idea that natural
than the de novo synthesis of nucleotides. Furthermore, transformation serves as a mechanism of DNA repair

FEMS Microbiol Rev 37 (2013) 336–363 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
354 P. Seitz & M. Blokesch

seems sound. In this scenario, the uptake of DNA from growth arrest, as seen in B. subtilis), benefits arising from
closely related organisms would facilitate the maintenance these processes might have been overlooked in the past
of genomic integrity. Evidence from experiments on the (Engelmoer & Rozen, 2011). In this study, the authors
naturally competent Gram-positive bacterium B. subtilis investigated the natural competence of S. pneumoniae and
supports this hypothesis (Michod et al., 1988; Wojcie- confirmed that transformation is beneficial in a ‘DNA for
chowski et al., 1989), but recent findings on the two repair’ scenario upon treating cells with DNA-damaging
Gram-negative bacteria, H. pylori and L. pneumophila, agents (Engelmoer & Rozen, 2011), but they also provided
with nonsense mutations in their DNA-uptake systems, evidence that competence is beneficial in withstanding
did not support the repair hypothesis (Dorer et al., other kinds of stresses and that these benefits do not rely
2010; Charpentier et al., 2011). In these studies the on transformation (Engelmoer & Rozen, 2011). The
bacterial strains incapable of DNA uptake showed no authors concluded that their findings were in line with
increased sensitivity to genotoxic agents (Dorer et al., ‘Claverys’ hypothesis (Claverys et al., 2006) that compe-
2010; Charpentier et al., 2011). tence but not necessarily transformation may act as a
general process to relieve stress’ (Engelmoer & Rozen,
2011).
DNA for evolution
In summary, we conclude that there are many benefits
Finally, natural transformation might enable rapid evolu- of natural competence and transformation, and there
tion when diversity may be beneficial; these circumstances might be no single reason that transformation is main-
include such stresses as high population densities, DNA tained in bacteria. The fact that numerous bacteria are
damage, abundance or a lack of certain carbon sources known or predicted to be naturally transformable is a
and/or starvation. All of these conditions are known to strong indication of the importance of this mode of
induce natural competence in at least a subset of natu- HGT.
rally transformable bacteria, as described earlier and illus-
trated in Fig. 1. A recent study on the long-term in vitro
Concluding remarks
passage of H. pylori (c. 1000 generations) supported an
evolutionary advantage of natural transformation because
Regulation of the competence window may
competent bacteria increased their fitness faster than
have co-evolved as an ability to respond to
those unable to take up external DNA (Baltrus et al.,
environmental cues
2008). Another recent study investigated the occurrence
of multi-drug-resistant (MDR) strains based on recombi- Summarizing the information on competence induction
nation following DNA uptake as part of the natural trans- presented here, we conclude that natural competence
formation process of A. baylyi (Perron et al., 2012). The occurs constitutively in certain Gram-negative bacteria
authors demonstrated that in the presence of recombina- but is tightly regulated and transient in others. But still
tion, resistance genes were readily exchanged, and MDR the question of ‘who’s competent and when’, which was
strains were obtained within fewer generations (Perron initially asked by Solomon & Grossman (1996), cannot
et al., 2012). yet be fully answered. Interestingly, several recent studies
However, as noted by Perron et al. and as described in examine how natural competence and transformation are
depth in a recent review on this topic (MacLean et al., evolutionarily maintained over time (Johnsen et al., 2009;
2010), it is important that ‘evolution experiments offer a Maughan & Redfield, 2009). The question of why some
useful approach to uncover the factors determining the naturally competent bacteria are competent only in
evolution of resistance, but most experiments have stud- a brief, finely tuned time window has not yet been
ied clonal populations without any contribution of
recombination’. Furthermore, the ‘benefits of recombina-
tion are context-dependent’, and experimental setups are
crucial to the outcome of such experiments. Such context
dependency was also highlighted in a recent study by
Engelmoer & Rozen (2011). These authors emphasized
once more the biased setup of most experimental studies,
which only examine benefits dependent on the acquisition
of DNA as part of natural transformation. However,
because natural competence is a developmental programme Fig. 4. The direct correlation between the variation encountered in
and often induces other processes apart from DNA uptake an organism’s niche and the complexity of the regulatory network
(e.g. fratricide in S. pneumoniae or competence-dependent driving competence induction. For details, see the text.

ª 2012 Federation of European Microbiological Societies FEMS Microbiol Rev 37 (2013) 336–363
Published by Blackwell Publishing Ltd. All rights reserved
Regulation of natural competence in Gram-negative bacteria 355

conclusively addressed. An examination of the Acknowledgements


environments of naturally competent bacteria might yield
evidence as to why some of these organisms have tighter We apologize to those researchers whose work we could
competence windows than others (Fig. 4). For example, not include due to space limitations. This work was sup-
H. pylori, as a gastric pathogen, solely colonizes the epi- ported by Swiss National Science Foundation (SNSF)
thelium of the human stomach, and no other reservoir grant 31003A_127029.
has been reported. Helicobacter pylori does not need to
cope with sudden and/or dramatic changes in its environ- References
ment (Fig. 4). Instead, such parameters as pH or nutrient
availability are likely to fluctuate only within a narrow Aas FE, Wolfgang M, Frye S, Dunham S, Lovold C & Koomey M
range. However, during long-term infection, environmen- (2002) Competence for natural transformation in Neisseria
tal alterations might include the ageing of the host and gonorrhoeae: components of DNA binding and uptake linked
concomitant changes in immune status or a secondary to type IV pilus expression. Mol Microbiol 46: 749–760.
Achtman M, Azuma T, Berg DE, Ito Y, Morelli G, Pan ZJ,
infection with another H. pylori strain. In this context,
Suerbaum S, Thompson SA, van der Ende A & van Doorn LJ
natural transformation can lead to the rapid spread of
(1999) Recombination and clonal groupings within
beneficial mutations and thus provides a means for adap-
Helicobacter pylori from different geographical regions. Mol
tion. Because H. pylori is constitutively competent, it Microbiol 32: 459–470.
maintains high genetic plasticity during infection. There- Ahmed N, Dobrindt U, Hacker J & Hasnain SE (2008)
fore, the bacteria are constantly able to fine-tune interac- Genomic fluidity and pathogenic bacteria: applications in
tions with their human hosts, as suggested in a recent diagnostics, epidemiology and intervention. Nat Rev
review on the evolution and phenotypic diversification of Microbiol 6: 387–394.
H. pylori (Suerbaum & Josenhans, 2007). Alexander HE & Redman W (1953) Transformation of type
The dynamics might differ significantly in naturally specificity of meningococci; change in heritable type
transformable bacteria that frequently encounter dramatic induced by type-specific extracts containing
fluctuations in their environment (Fig. 4). A good exam- desoxyribonucleic acid. J Exp Med 97: 797–806.
ple is V. cholerae, an aquatic and naturally transformable Allemand JF & Maier B (2009) Bacterial translocation motors
bacterium. In its environmental niche, this organism can investigated by single molecule techniques. FEMS Microbiol
be found as free-living bacterium in a planktonic state or Rev 33: 593–610.
Allemand JF, Maier B & Smith DE (2012) Molecular motors
as part of a sessile biofilm community (Yildiz & Visick,
for DNA translocation in prokaryotes. Curr Opin Biotechnol
2009). Furthermore, V. cholerae is well known for its abil-
23: 503–509.
ity to interact with phyto- and zooplankton, of which the
Alvarez-Martinez CE & Christie PJ (2009) Biological diversity
mucilaginous sheath and the exoskeleton, respectively, of prokaryotic type IV secretion systems. Microbiol Mol Biol
can serve as carbon and energy sources (reviewed by Lipp Rev 73: 775–808.
et al., 2002). The abundance of such carbon sources, Ambur OH, Frye SA & Tonjum T (2007) New functional
however, can fluctuate significantly, and climate changes identity for the DNA uptake sequence in transformation
and human behaviour are thought to contribute signifi- and its presence in transcriptional terminators. J Bacteriol
cantly to plankton blooms and V. cholerae abundance 189: 2077–2085.
(Colwell, 1996; Lobitz et al., 2000; Lipp et al., 2002; Pru- Ambur OH, Davidsen T, Frye SA, Balasingham SV, Lagesen K,
zzo et al., 2008; Fig. 4). Furthermore, selection pressure Rognes T & Tonjum T (2009) Genome dynamics in major
from environmental cholera bacteriophages, which are bacterial pathogens. FEMS Microbiol Rev 33: 453–470.
also thought to play a significant role in cholera seasonal- Anderson MT & Seifert HS (2011) Opportunity and means:
ity and the emergence of new clones (Faruque et al., horizontal gene transfer from the human host to a bacterial
2005), as well as pressure from protozoan grazers (Matz pathogen. mBio 2: e00005–e00011.
Antonova ES & Hammer BK (2011) Quorum-sensing
et al., 2005; Erken et al., 2011), may vary in time and
autoinducer molecules produced by members of a
space. Thus, the adjustment between famine and feast
multispecies biofilm promote horizontal gene transfer to
and between growth and growth arrest including the Vibrio cholerae. FEMS Microbiol Lett 322: 68–76.
ability to enter the state of natural competence must be Aras RA, Small AJ, Ando T & Blaser MJ (2002) Helicobacter
fine-tuned (Fig. 4). Further studies will be required to pylori interstrain restriction-modification diversity prevents
address the potential disadvantages of constitutive compe- genome subversion by chromosomal DNA from competing
tence and transformation under changing environmental strains. Nucleic Acids Res 30: 5391–5397.
conditions; such weaknesses could explain why the com- Averhoff B (2009) Shuffling genes around in hot
petence window is adjusted according to the bacterium’s environments: the unique DNA transporter of Thermus
lifestyle. thermophilus. FEMS Microbiol Rev 33: 611–626.

FEMS Microbiol Rev 37 (2013) 336–363 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
356 P. Seitz & M. Blokesch

Averhoff B & Friedrich A (2003) Type IV pili-related natural Burton B & Dubnau D (2010) Membrane-associated DNA
transformation systems: DNA transport in mesophilic and transport machines. Cold Spring Harb Perspect Biol 2:
thermophilic bacteria. Arch Microbiol 180: 385–393. a000406.
Averhoff B & Müller V (2010) Exploring research frontiers in Cambray G, Guerout AM & Mazel D (2010) Integrons. Annu
microbiology: recent advances in halophilic and thermophilic Rev Genet 44: 141–166.
extremophiles. Res Microbiol 161: 506–514. Cameron AD & Redfield RJ (2006) Non-canonical CRP sites
Baharoglu Z, Krin E & Mazel D (2012) Connecting control competence regulons in Escherichia coli and many
environment and genome plasticity in the characterization other gamma-proteobacteria. Nucleic Acids Res 34: 6001–6014.
of transformation-induced SOS regulation and carbon Cameron AD & Redfield RJ (2008) CRP binding and
catabolite control of the Vibrio cholerae integron integrase. transcription activation at CRP-S sites. J Mol Biol 383:
J Bacteriol 194: 1659–1667. 313–323.
Baltrus DA & Guillemin K (2006) Multiple phases of Cameron AD, Volar M, Bannister LA & Redfield RJ (2008)
competence occur during the Helicobacter pylori growth RNA secondary structure regulates the translation of sxy
cycle. FEMS Microbiol Lett 255: 148–155. and competence development in Haemophilus influenzae.
Baltrus DA, Guillemin K & Phillips PC (2008) Natural Nucleic Acids Res 36: 10–20.
transformation increases the rate of adaptation in the Carlson CA, Pierson LS, Rosen JJ & Ingraham JL (1983)
human pathogen Helicobacter pylori. Evolution 62: 39–49. Pseudomonas stutzeri and related species undergo natural
Baumann P (1968) Isolation of Acinetobacter from soil and transformation. J Bacteriol 153: 93–99.
water. J Bacteriol 96: 39–42. Carr EL, Kampfer P, Patel BK, Gurtler V & Seviour RJ (2003)
Berndt C, Meier P & Wackernagel W (2003) DNA restriction Seven novel species of Acinetobacter isolated from activated
is a barrier to natural transformation in Pseudomonas sludge. Int J Syst Evol Microbiol 53: 953–963.
stutzeri JM300. Microbiology 149: 895–901. Catlin BW & Cunningham LS (1961) Transforming activities
Bertolla F, Van Gijsegem F, Nesme X & Simonet P and base contents of deoxyribonucleate preparations from
(1997) Conditions for natural transformation of various Neisseriae. J Gen Microbiol 26: 303–312.
Ralstonia solanacearum. Appl Environ Microbiol 63: Cazalet C, Rusniok C, Bruggemann H et al. (2004) Evidence in
4965–4968. the Legionella pneumophila genome for exploitation of host
Bertolla F, Frostesard A, Brito B, Nesme X & Simonet P cell functions and high genome plasticity. Nat Genet 36:
(1999) During infection of its host, the plant pathogen 1165–1173.
Ralstonia solanacearum naturally develops a state of Chandler MS (1992) The gene encoding cAMP receptor
competence and exchanges genetic material. Mol Plant protein is required for competence development in
Microbe Interact 12: 467–472. Haemophilus influenzae Rd. P Natl Acad Sci USA 89:
Bielaszewska M, Mellmann A, Zhang W, Kock R, Fruth A, 1626–1630.
Bauwens A, Peters G & Karch H (2011) Characterisation Charpentier X, Kay E, Schneider D & Shuman HA (2011)
of the Escherichia coli strain associated with an outbreak Antibiotics and UV radiation induce competence for natural
of haemolytic uraemic syndrome in Germany, 2011: transformation in Legionella pneumophila. J Bacteriol 193:
a microbiological study. Lancet Infect Dis 11: 1114–1121.
671–676. Chatterjee S, Almeida RP & Lindow S (2008) Living in two
Blokesch M (2012) Chitin colonization, chitin degradation and worlds: the plant and insect lifestyles of Xylella fastidiosa.
chitin-induced natural competence of Vibrio cholerae are Annu Rev Phytopathol 46: 243–271.
subject to catabolite repression. Environ Microbiol 14: Chen I & Dubnau D (2004) DNA uptake during bacterial
1898–1912. transformation. Nat Rev Microbiol 2: 241–249.
Blokesch M & Schoolnik GK (2007) Serogroup conversion of Chen X, Schauder S, Potier N, Van Dorsselaer A, Pelczer I,
Vibrio cholerae in aquatic reservoirs. PLoS Pathog 3: e81. Bassler BL & Hughson FM (2002) Structural identification
Blokesch M & Schoolnik GK (2008) The extracellular nuclease of a bacterial quorum-sensing signal containing boron.
Dns and its role in natural transformation of Vibrio Nature 415: 545–549.
cholerae. J Bacteriol 190: 7232–7240. Chen Y, Dai J, Morris JG Jr & Johnson JA (2010) Genetic
Boucher Y, Nesbo CL & Doolittle WF (2001) Microbial genomes: analysis of the capsule polysaccharide (K antigen) and
dealing with diversity. Curr Opin Microbiol 4: 285–289. exopolysaccharide genes in pandemic Vibrio
Boucher Y, Douady CJ, Papke RT, Walsh DA, Boudreau ME, parahaemolyticus O3:K6. BMC Microbiol 10: 274.
Nesbo CL, Case RJ & Doolittle WF (2003) Lateral gene Chin CS, Sorenson J, Harris JB et al. (2010) The origin of
transfer and the origins of prokaryotic groups. Annu Rev the Haitian cholera outbreak strain. N Engl J Med 364:
Genet 37: 283–328. 33–42.
Brumbley SM, Carney BF & Denny TP (1993) Phenotype Chun J, Grim CJ, Hasan NA et al. (2009) Comparative
conversion in Pseudomonas solanacearum due to genomics reveals mechanism for short-term and long-term
spontaneous inactivation of PhcA, a putative LysR clonal transitions in pandemic Vibrio cholerae. P Natl Acad
transcriptional regulator. J Bacteriol 175: 5477–5487. Sci USA 106: 15442–15447.

ª 2012 Federation of European Microbiological Societies FEMS Microbiol Rev 37 (2013) 336–363
Published by Blackwell Publishing Ltd. All rights reserved
Regulation of natural competence in Gram-negative bacteria 357

Claverys JP & Havarstein LS (2007) Cannibalism and DNA for natural transformation and is found more
fratricide: mechanisms and raisons d’etre. Nat Rev Microbiol often in disseminated infection isolates. Mol Microbiol 41:
5: 219–229. 263–277.
Claverys JP, Prudhomme M, Mortier-Barriere I & Martin B Dobrindt U, Hochhut B, Hentschel U & Hacker J (2004)
(2000) Adaptation to the environment: Streptococcus Genomic islands in pathogenic and environmental
pneumoniae, a paradigm for recombination-mediated microorganisms. Nat Rev Microbiol 2: 414–424.
genetic plasticity? Mol Microbiol 35: 251–259. Dobrindt U, Chowdary MG, Krumbholz G & Hacker J (2010)
Claverys JP, Prudhomme M & Martin B (2006) Induction of Genome dynamics and its impact on evolution of
competence regulons as a general response to stress in Escherichia coli. Med Microbiol Immunol 199: 145–154.
gram-positive bacteria. Annu Rev Microbiol 60: 451–475. Dorer MS, Fero J & Salama NR (2010) DNA damage triggers
Claverys JP, Martin B & Polard P (2009) The genetic genetic exchange in Helicobacter pylori. PLoS Pathog 6:
transformation machinery: composition, localization, and e1001026.
mechanism. FEMS Microbiol Rev 33: 643–656. Dorer MS, Sessler TH & Salama NR (2011) Recombination
Clough SJ, Lee KE, Schell MA & Denny TP (1997) A two- and DNA repair in Helicobacter pylori. Annu Rev Microbiol
component system in Ralstonia (Pseudomonas) solanacearum 65: 329–348.
modulates production of PhcA-regulated virulence factors in Dorocicz IR, Williams PM & Redfield RJ (1993) The
response to 3-hydroxypalmitic acid methyl ester. J Bacteriol Haemophilus influenzae adenylate cyclase gene: cloning,
179: 3639–3648. sequence, and essential role in competence. J Bacteriol 175:
Cohan FM & Koeppel AF (2008) The origins of ecological 7142–7149.
diversity in prokaryotes. Curr Biol 18: R1024–R1034. Draskovic I & Dubnau D (2005) Biogenesis of a putative
Colwell RR (1996) Global climate and infectious disease: the channel protein, ComEC, required for DNA uptake:
cholera paradigm. Science 274: 2025–2031. membrane topology, oligomerization and formation of
Coupat B, Chaumeille-Dole F, Fall S, Prior P, Simonet P, Nesme disulphide bonds. Mol Microbiol 55: 881–896.
X & Bertolla F (2008) Natural transformation in the Ralstonia Dubnau D (1999) DNA uptake in bacteria. Annu Rev
solanacearum species complex: number and size of DNA that Microbiol 53: 217–244.
can be transferred. FEMS Microbiol Ecol 66: 14–24. Duffin PM & Seifert HS (2010) DNA uptake sequence-
Coupat-Goutaland B, Bernillon D, Guidot A, Prior P, Nesme X mediated enhancement of transformation in Neisseria
& Bertolla F (2011) Ralstonia solanacearum virulence gonorrhoeae is strain dependent. J Bacteriol 192:
increased following large interstrain gene transfers by natural 4436–4444.
transformation. Mol Plant Microbe Interact 24: 497–505. Duffin PM & Seifert HS (2012) Genetic transformation of
Croucher NJ, Harris SR, Fraser C et al. (2011) Rapid Neisseria gonorrhoeae shows a strand preference. FEMS
pneumococcal evolution in response to clinical Microbiol Lett 334: 44–48.
interventions. Science 331: 430–434. Dziejman M, Balon E, Boyd D, Fraser CM, Heidelberg JF &
Cruze JA, Singer JT & Finnerty WR (1979) Conditions for Mekalanos JJ (2002) Comparative genomic analysis of
quantitative transformation in Acinetobacter calcoaceticus. Vibrio cholerae: genes that correlate with cholera
Curr Microbiol 3: 129–132. endemic and pandemic disease. P Natl Acad Sci USA 99:
Danner DB, Deich RA, Sisco KL & Smith HO (1980) An 1556–1561.
eleven-base-pair sequence determines the specificity of DNA Dziejman M, Serruto D, Tam VC et al. (2005) Genomic
uptake in Haemophilus transformation. Gene 11: 311–318. characterization of non-O1, non-O139 Vibrio cholerae
Davidsen T, Rodland EA, Lagesen K, Seeberg E, Rognes T & reveals genes for a type III secretion system. P Natl Acad Sci
Tonjum T (2004) Biased distribution of DNA uptake USA 102: 3465–3470.
sequences towards genome maintenance genes. Nucleic Acids Eisen JA (2000) Horizontal gene transfer among microbial
Res 32: 1050–1058. genomes: new insights from complete genome analysis. Curr
Davidsen T, Koomey M & Tonjum T (2007) Microbial Opin Genet Dev 10: 606–611.
genome dynamics in CNS pathogenesis. Neuroscience 145: Elkins C, Thomas CE, Seifert HS & Sparling PF (1991)
1375–1387. Species-specific uptake of DNA by gonococci is mediated by
Demaneche S, Kay E, Gourbiere F & Simonet P (2001) a 10-base-pair sequence. J Bacteriol 173: 3911–3913.
Natural transformation of Pseudomonas fluorescens and Engelmoer DJ & Rozen DE (2011) Competence increases
Agrobacterium tumefaciens in soil. Appl Environ Microbiol survival during stress in Streptococcus pneumoniae. Evolution
67: 2617–2621. 65: 3475–3485.
Deutscher J, Francke C & Postma PW (2006) How Erken M, Weitere M, Kjelleberg S & McDougald D (2011) In
phosphotransferase system-related protein phosphorylation situ grazing resistance of Vibrio cholerae in the marine
regulates carbohydrate metabolism in bacteria. Microbiol environment. FEMS Microbiol Ecol 76: 504–512.
Mol Biol Rev 70: 939–1031. Falush D, Kraft C, Taylor NS, Correa P, Fox JG, Achtman M
Dillard JP & Seifert HS (2001) A variable genetic island & Suerbaum S (2001) Recombination and mutation during
specific for Neisseria gonorrhoeae is involved in providing long-term gastric colonization by Helicobacter pylori:

FEMS Microbiol Rev 37 (2013) 336–363 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
358 P. Seitz & M. Blokesch

estimates of clock rates, recombination size, and minimal Gaasbeek EJ, Wagenaar JA, Guilhabert MR, van Putten JP,
age. P Natl Acad Sci USA 98: 15056–15061. Parker CT & van der Wal FJ (2010) Nucleases encoded
Falush D, Wirth T, Linz B et al. (2003) Traces of human by the integrated elements CJIE2 and CJIE4 inhibit
migrations in Helicobacter pylori populations. Science 299: natural transformation of Campylobacter jejuni. J Bacteriol
1582–1585. 192: 936–941.
Faruque SM, Naser IB, Islam MJ, Faruque AS, Ghosh AN, Genin S (2010) Molecular traits controlling host range and
Nair GB, Sack DA & Mekalanos JJ (2005) Seasonal adaptation to plants in Ralstonia solanacearum. New Phytol
epidemics of cholera inversely correlate with the prevalence 187: 920–928.
of environmental cholera phages. Proc Natl Acad Sci USA Genin S & Boucher C (2004) Lessons learned from the
102: 1702–1707. genome analysis of Ralstonia solanacearum. Annu Rev
Feil E, Zhou J, Maynard Smith J & Spratt BG (1996) A Phytopathol 42: 107–134.
comparison of the nucleotide sequences of the adk and recA Gilmore MS & Haas W (2005) The selective advantage of
genes of pathogenic and commensal Neisseria species: microbial fratricide. P Natl Acad Sci USA 102: 8401–
evidence for extensive interspecies recombination within 8402.
adk. J Mol Evol 43: 631–640. Gomez-Valero L, Rusniok C, Jarraud S, Vacherie B, Rouy Z,
Feil EJ, Maiden MCJ, Achtman M & Spratt BG (1999) The Barbe V, Medigue C, Etienne J & Buchrieser C (2011)
relative contributions of recombination and mutation to the Extensive recombination events and horizontal gene transfer
divergence of clones of Neisseria meningitidis. Mol Biol Evol shaped the Legionella pneumophila genomes. BMC Genomics
16: 1496–1502. 12: 536.
Finkel SE & Kolter R (2001) DNA as a nutrient: novel role for Goodman SD & Scocca JJ (1988) Identification and
bacterial competence gene homologs. J Bacteriol 183: 6288– arrangement of the DNA sequence recognized in specific
6293. transformation of Neisseria gonorrhoeae. P Natl Acad Sci
Fitzmaurice WP, Benjamin RC, Huang PC & Scocca JJ (1984) USA 85: 6982–6986.
Characterization of recognition sites on bacteriophage Goodman SD & Scocca JJ (1991) Factors influencing the
HP1c1 DNA which interact with the DNA uptake system specific interaction of Neisseria gonorrhoeae with
of Haemophilus influenzae Rd. Gene 31: 187 transforming DNA. J Bacteriol 173: 5921–5923.
–196. Grad YH, Lipsitch M, Feldgarden M et al. (2012) Genomic
Flavier AB, Clough SJ, Schell MA & Denny TP (1997) epidemiology of the Escherichia coli O104:H4 outbreaks in
Identification of 3-hydroxypalmitic acid methyl ester as a Europe, 2011. P Natl Acad Sci USA 109: 3065–3070.
novel autoregulator controlling virulence in Ralstonia Graupner S, Frey V, Hashemi R, Lorenz MG, Brandes G &
solanacearum. Mol Microbiol 26: 251–259. Wackernagel W (2000) Type IV pilus genes pilA and pilC of
Focareta T & Manning PA (1987) Extracellular proteins of Pseudomonas stutzeri are required for natural genetic
Vibrio cholerae: molecular cloning, nucleotide sequence and transformation, and pilA can be replaced by corresponding
characterization of the deoxyribonuclease (DNase) together genes from nontransformable species. J Bacteriol 182: 2184–
with its periplasmic localization in Escherichia coli K-12. 2190.
Gene 53: 31–40. Griffith F (1928) The significance of pneumococcal types. J
Frischer ME, Thurmond JM & Paul JH (1990) Natural Hyg 27: 113–159.
plasmid transformation in a high-frequency-of- Guiral S, Mitchell TJ, Martin B & Claverys JP (2005)
transformation marine Vibrio strain. Appl Environ Microbiol Competence-programmed predation of noncompetent cells
56: 3439–3444. in the human pathogen Streptococcus pneumoniae: genetic
Frischer ME, Thurmond JM & Paul JH (1993) Factors requirements. P Natl Acad Sci USA 102: 8710–8715.
affecting competence in a high-frequency of transformation Gulig PA, Tucker MS, Thiaville PC, Joseph JL & Brown RN
marine Vibrio. J Gen Microbiol 139: 753–761. (2009) USER friendly cloning coupled with chitin-based
Frischer ME, Williams HG, Bennison B, Drake GR, Balkwill natural transformation enables rapid mutagenesis of Vibrio
DL & Paul JH (1996) The naturally transformable marine vulnificus. Appl Environ Microbiol 75: 4936–4949.
bacterium WJT-1C formally identified as “Vibrio” is a Hacker J, Blum-Oehler G, Mühldorfer I & Tschäpe H (1997)
pseudomonad. Curr Microbiol 33: 287–291. Pathogenicity islands of virulent bacteria: structure,
Fullner KJ & Mekalanos JJ (1999) Genetic characterization of a function and impact on microbial evolution. Mol Microbiol
new type IV-A pilus gene cluster found in both classical 23: 1089–1097.
and El Tor biotypes of Vibrio cholerae. Infect Immun 67: Hacker J, Hentschel U & Dobrindt U (2003) Prokaryotic
1393–1404. chromosomes and disease. Science 301: 790–793.
Gaasbeek EJ, Wagenaar JA, Guilhabert MR, Wosten MM, van Hamilton HL & Dillard JP (2006) Natural transformation of
Putten JP, van der Graaf-van Bloois L, Parker CT & van der Neisseria gonorrhoeae: from DNA donation to homologous
Wal FJ (2009) A DNase encoded by integrated element recombination. Mol Microbiol 59: 376–385.
CJIE1 inhibits natural transformation of Campylobacter Hamilton HL, Dominguez NM, Schwartz KJ, Hackett KT &
jejuni. J Bacteriol 191: 2296–2306. Dillard JP (2005) Neisseria gonorrhoeae secretes

ª 2012 Federation of European Microbiological Societies FEMS Microbiol Rev 37 (2013) 336–363
Published by Blackwell Publishing Ltd. All rights reserved
Regulation of natural competence in Gram-negative bacteria 359

chromosomal DNA via a novel type IV secretion system. Johnsen PJ, Dubnau D & Levin BR (2009) Episodic selection
Mol Microbiol 55: 1704–1721. and the maintenance of competence and natural
Hanage WP, Fraser C & Spratt BG (2005) Fuzzy species transformation in Bacillus subtilis. Genetics 181: 1521–1533.
among recombinogenic bacteria. BMC Biol 3: 6. Johnson ZL, Cheong CG & Lee SY (2012) Crystal structure of
Havarstein LS, Martin B, Johnsborg O, Granadel C & Claverys a concentrative nucleoside transporter from Vibrio cholerae
JP (2006) New insights into the pneumococcal fratricide: at 2.4 A. Nature 483: 489–493.
relationship to clumping and identification of a novel Juni E (1978) Genetics and physiology of Acinetobacter. Annu
immunity factor. Mol Microbiol 59: 1297–1307. Rev Microbiol 32: 349–371.
Heidelberg JF, Eisen JA, Nelson WC et al. (2000) DNA Juni E & Janik A (1969) Transformation of Acinetobacter calco-
sequence of both chromosomes of the cholera pathogen aceticus (Bacterium anitratum). J Bacteriol 98: 281–288.
Vibrio cholerae. Nature 406: 477–483. Kang Y, Liu H, Genin S, Schell MA & Denny TP (2002)
Hendriksen RS, Price LB, Schupp JM et al. (2011) Population Ralstonia solanacearum requires type 4 pili to adhere to
genetics of Vibrio cholerae from Nepal in 2010: evidence on multiple surfaces and for natural transformation and
the origin of the Haitian outbreak. mBio 2: e00157–11. virulence. Mol Microbiol 46: 427–437.
Herriott RM, Meyer EM & Vogt M (1970) Defined nongrowth Karnholz A, Hoefler C, Odenbreit S, Fischer W, Hofreuter D
media for stage II development of competence in & Haas R (2006) Functional and topological
Haemophilus influenzae. J Bacteriol 101: 517–524. characterization of novel components of the comB DNA
Herzberg C, Friedrich A & Averhoff B (2000) comB, a novel transformation competence system in Helicobacter pylori.
competence gene required for natural transformation of J Bacteriol 188: 882–893.
Acinetobacter sp. BD413: identification, characterization, and Karudapuram S & Barcak GJ (1997) The Haemophilus
analysis of growth-phase-dependent regulation. Arch influenzae dprABC genes constitute a competence-inducible
Microbiol 173: 220–228. operon that requires the product of the tfoX (sxy) gene for
Higgins DA, Pomianek ME, Kraml CM, Taylor RK, transcriptional activation. J Bacteriol 179: 4815–4820.
Semmelhack MF & Bassler BL (2007) The major Vibrio Kersulyte D, Chalkauskas H & Berg DE (1999) Emergence of
cholerae autoinducer and its role in virulence factor recombinant strains of Helicobacter pylori during human
production. Nature 450: 883–886. infection. Mol Microbiol 31: 31–43.
Hobbs M & Mattick JS (1993) Common components in the Kessler C & Manta V (1990) Specificity of restriction
assembly of type 4 fimbriae, DNA transfer systems, endonucleases and DNA modification methyltransferases a
filamentous phage and protein-secretion apparatus: a review. Gene 92: 1–248.
general system for the formation of surface-associated Keyhani NO & Roseman S (1999) Physiological aspects of
protein complexes. Mol Microbiol 10: 233–243. chitin catabolism in marine bacteria. Biochim Biophys Acta
Hofreuter D, Odenbreit S, Henke G & Haas R (1998) Natural 1473: 108–122.
competence for DNA transformation in Helicobacter pylori: Killiny N, Prado SS & Almeida RP (2010) Chitin utilization by
identification and genetic characterization of the comB the insect-transmitted bacterium Xylella fastidiosa. Appl
locus. Mol Microbiol 28: 1027–1038. Environ Microbiol 76: 6134–6140.
Hofreuter D, Odenbreit S, Puls J, Schwan D & Haas R (2000) Koomey M (1998) Competence for natural transformation in
Genetic competence in Helicobacter pylori: mechanisms and Neisseria gonorrhoeae: a model system for studies of
biological implications. Res Microbiol 151: 487–491. horizontal gene transfer. APMIS Suppl 84: 56–61.
Humbert O & Salama NR (2008) The Helicobacter pylori Koonin EV & Wolf YI (2008) Genomics of bacteria and
HpyAXII restriction-modification system limits exogenous archaea: the emerging dynamic view of the prokaryotic
DNA uptake by targeting GTAC sites but shows asymmetric world. Nucleic Acids Res 36: 6688–6719.
conservation of the DNA methyltransferase and restriction Koonin EV, Makarova KS & Aravind L (2001) Horizontal gene
endonuclease components. Nucleic Acids Res 36: 6893–6906. transfer in prokaryotes: quantification and classification.
Humbert O, Dorer MS & Salama NR (2011) Characterization Annu Rev Microbiol 55: 709–742.
of Helicobacter pylori factors that control transformation Koyama Y, Hoshino T, Tomizuka N & Furukawa K (1986)
frequency and integration length during inter-strain DNA Genetic transformation of the extreme thermophile Thermus
recombination. Mol Microbiol 79: 387–401. thermophilus and of other Thermus spp. J Bacteriol 166:
Hunt DE, Gevers D, Vahora NM & Polz MF (2008) 338–340.
Conservation of the chitin utilization pathway in the Kroll JS, Wilks KE, Farrant JL & Langford PR (1998)
Vibrionaceae. Appl Environ Microbiol 74: 44–51. Natural genetic exchange between Haemophilus and
Israel DA, Lou AS & Blaser MJ (2000) Characteristics of Neisseria: intergeneric transfer of chromosomal genes
Helicobacter pylori natural transformation. FEMS Microbiol between major human pathogens. P Natl Acad Sci USA
Lett 186: 275–280. 95: 12381–12385.
Jeffrey WH, Paul JH & Stewart GJ (1990) Natural Kru ger NJ & Stingl K (2011) Two steps away from novelty–
transformation of a marine Vibrio species by plasmid DNA. principles of bacterial DNA uptake. Mol Microbiol 80: 860–
Microb Ecol 19: 259–268. 867.

FEMS Microbiol Rev 37 (2013) 336–363 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
360 P. Seitz & M. Blokesch

Kung SH & Almeida RP (2011) Natural competence and Lorenz MG, Reipschlager K & Wackernagel W (1992) Plasmid
recombination in the plant pathogen Xylella fastidiosa. Appl transformation of naturally competent Acinetobacter
Environ Microbiol 77: 5278–5284. calcoaceticus in non-sterile soil extract and groundwater.
Lalucat J, Bennasar A, Bosch R, Garcia-Valdes E & Palleroni Arch Microbiol 157: 355–360.
NJ (2006) Biology of Pseudomonas stutzeri. Microbiol Mol Macfadyen LP (2000) Regulation of competence development
Biol Rev 70: 510–547. in Haemophilus influenzae. J Theor Biol 207: 349–359.
Langenberg W, Rauws EAJ, Widjojokusumo A, Tytgat GNJ & MacFadyen LP, Chen D, Vo HC, Liao D, Sinotte R & Redfield
Zanen HC (1986) Identification of Campylobacter pyloridis RJ (2001) Competence development by Haemophilus
isolates by restriction endonuclease DNA analysis. J Clin influenzae is regulated by the availability of nucleic acid
Microbiol 24: 414–417. precursors. Mol Microbiol 40: 700–707.
Li X & Roseman S (2004) The chitinolytic cascade in Vibrios is MacLean RC, Hall AR, Perron GG & Buckling A (2010) The
regulated by chitin oligosaccharides and a two-component population genetics of antibiotic resistance: integrating
chitin catabolic sensor/kinase. P Natl Acad Sci USA 101: molecular mechanisms and treatment contexts. Nat Rev
627–631. Genet 11: 405–414.
Liang W, Pascual-Montano A, Silva AJ & Benitez JA (2007) Maiden MC (2008) Population genomics: diversity and
The cyclic AMP receptor protein modulates quorum virulence in the Neisseria. Curr Opin Microbiol 11: 467–
sensing, motility and multiple genes that affect intestinal 471.
colonization in Vibrio cholerae. Microbiology 153: 2964– Maier B (2005) Using laser tweezers to measure twitching
2975. motility in Neisseria. Curr Opin Microbiol 8: 344–349.
Liang W, Sultan SZ, Silva AJ & Benitez JA (2008) Cyclic Maier B, Potter L, So M, Long CD, Seifert HS & Sheetz MP
AMP post-transcriptionally regulates the biosynthesis of a (2002) Single pilus motor forces exceed 100 pN. P Natl
major bacterial autoinducer to modulate the cell density Acad Sci USA 99: 16012–16017.
required to activate quorum sensing. FEBS Lett 582: Maier B, Chen I, Dubnau D & Sheetz MP (2004) DNA
3744–3750. transport into Bacillus subtilis requires proton motive force
Linz B, Balloux F, Moodley Y et al. (2007) An African origin to generate large molecular forces. Nat Struct Mol Biol 11:
for the intimate association between humans and 643–649.
Helicobacter pylori. Nature 445: 915–918. Majewski SI & Goodwin CS (1988) Restriction endonuclease
Lipp EK, Huq A & Colwell RR (2002) Effects of global climate analysis of the genome of Campylobacter pylori with a rapid
on infectious disease: the cholera model. Clin Microbiol Rev extraction method: evidence for considerable genomic
15: 757–770. variation. J Infect Dis 157: 465–471.
Lo Scrudato M & Blokesch M (2012) The regulatory network Marsich E, Zuccato P, Rizzi S, Vetere A, Tonin E & Paoletti S
of natural competence and transformation of Vibrio (2002) Helicobacter pylori expresses an autolytic enzyme:
cholerae. PLoS Genet 8: e1002778. gene identification, cloning, and theoretical protein
Lobitz B, Beck L, Huq A, Wood B, Fuchs G, Faruque AS & structure. J Bacteriol 184: 6270–6279.
Colwell R (2000) Climate and infectious disease: use of Matz C, McDougald D, Moreno AM, Yung PY, Yildiz FH &
remote sensing for detection of Vibrio cholerae by indirect Kjelleberg S (2005) Biofilm formation and phenotypic
measurement. P Natl Acad Sci USA 97: 1438–1443. variation enhance predation-driven persistence of Vibrio
Lorenz MG & Sikorski J (2000) The potential for intraspecific cholerae. P Natl Acad Sci USA 102: 16819–16824.
horizontal gene exchange by natural genetic transformation: Maughan H & Redfield RJ (2009) Tracing the evolution of
sexual isolation among genomovars of Pseudomonas stutzeri. competence in Haemophilus influenzae. PLoS ONE 4:
Microbiology 146(Pt 12): 3081–3090. e5854.
Lorenz MG & Wackernagel W (1990) Natural genetic Meibom KL, Li XB, Nielsen AT, Wu CY, Roseman S &
transformation of Pseudomonas stutzeri by sand-adsorbed Schoolnik GK (2004) The Vibrio cholerae chitin utilization
DNA. Arch Microbiol 154: 380–385. program. P Natl Acad Sci USA 101: 2524–2529.
Lorenz MG & Wackernagel W (1991) High frequency of Meibom KL, Blokesch M, Dolganov NA, Wu C-Y & Schoolnik
natural genetic transformation of Pseudomonas stutzeri in GK (2005) Chitin induces natural competence in Vibrio
soil extract supplemented with a carbon/energy and cholerae. Science 310: 1824–1827.
phosphorus source. Appl Environ Microbiol 57: 1246– Meier P & Wackernagel W (2003) Mechanisms of homology-
1251. facilitated illegitimate recombination for foreign DNA
Lorenz MG & Wackernagel W (1994) Bacterial gene transfer acquisition in transformable Pseudomonas stutzeri. Mol
by natural genetic transformation in the environment. Microbiol 48: 1107–1118.
Microbiol Rev 58: 563–602. Meier P, Berndt C, Weger N & Wackernagel W (2002) Natural
Lorenz MG, Gerjets D & Wackernagel W (1991) Release of transformation of Pseudomonas stutzeri by single-stranded
transforming plasmid and chromosomal DNA from two DNA requires type IV pili, competence state and comA.
cultured soil bacteria. Arch Microbiol 156: 319–326. FEMS Microbiol Lett 207: 75–80.

ª 2012 Federation of European Microbiological Societies FEMS Microbiol Rev 37 (2013) 336–363
Published by Blackwell Publishing Ltd. All rights reserved
Regulation of natural competence in Gram-negative bacteria 361

Merz AJ, So M & Sheetz MP (2000) Pilus retraction powers Omelchenko MV, Wolf YI, Gaidamakova EK, Matrosova VY,
bacterial twitching motility. Nature 407: 98–102. Vasilenko A, Zhai M, Daly MJ, Koonin EV & Makarova KS
Michod RE, Wojciechowski MF & Hoelzer MA (1988) DNA (2005) Comparative genomics of Thermus thermophilus and
repair and the evolution of transformation in the bacterium Deinococcus radiodurans: divergent routes of adaptation to
Bacillus subtilis. Genetics 118: 31–39. thermophily and radiation resistance. BMC Evol Biol 5: 57.
Michod RE, Bernstein H & Nedelcu AM (2008) Adaptive Palchevskiy V & Finkel SE (2006) Escherichia coli competence
value of sex in microbial pathogens. Infect Genet Evol 8: gene homologs are essential for competitive fitness and the
267–285. use of DNA as a nutrient. J Bacteriol 188: 3902–3910.
Miller MC, Keymer DP, Avelar A, Boehm AB & Schoolnik GK Palmen R & Hellingwerf KJ (1997) Uptake and processing of
(2007) Detection and transformation of genome segments DNA by Acinetobacter calcoaceticus–a review. Gene 192:
that differ within a coastal population of Vibrio cholerae 179–190.
strains. Appl Environ Microbiol 73: 3695–3704. Palmen R, Vosman B, Buijsman P, Breek CK & Hellingwerf KJ
Milton DL (2006) Quorum sensing in Vibrios: complexity for (1993) Physiological characterization of natural
diversification. Int J Med Microbiol 296: 61–71. transformation in Acinetobacter calcoaceticus. J Gen Microbiol
Morelli G, Didelot X, Kusecek B, Schwarz S, Bahlawane C, 139: 295–305.
Falush D, Suerbaum S & Achtman M (2010) Palmen R, Buijsman P & Hellingwerf KJ (1994) Physiological
Microevolution of Helicobacter pylori during prolonged regulation of competence induction for natural
infection of single hosts and within families. PLoS Genet 6: transformation in Acinetobacter calcoaceticus. Arch Microbiol
e1001036. 162: 344–351.
Mortier-Barriere I, Velten M, Dupaigne P et al. (2007) A key Paul JH, Thurmond JM, Frischer ME & Cannon JP (1992)
presynaptic role in transformation for a widespread bacterial Intergeneric natural plasmid transformation between E. coli
protein: DprA conveys incoming ssDNA to RecA. Cell 130: and a marine Vibrio species. Mol Ecol 1: 37–46.
824–836. Peix A, Ramirez-Bahena MH & Velazquez E (2009) Historical
Munoz-Price LS & Weinstein RA (2008) Acinetobacter evolution and current status of the taxonomy of genus
infection. N Engl J Med 358: 1271–1281. Pseudomonas. Infect Genet Evol 9: 1132–1147.
Murphy TF & Brauer AL (2011) Expression of urease by Perron GG, Lee AE, Wang Y, Huang WE & Barraclough TG
Haemophilus influenzae during human respiratory tract (2012) Bacterial recombination promotes the evolution of
infection and role in survival in an acid environment. BMC multi-drug-resistance in functionally diverse populations.
Microbiol 11: 183. Proc Biol Sci 279: 1477–1484.
Mutreja A, Kim DW, Thomson NR et al. (2011) Evidence for Pollack-Berti A, Wollenberg MS & Ruby EG (2010) Natural
several waves of global transmission in the seventh cholera transformation of Vibrio fischeri requires tfoX and tfoY.
pandemic. Nature 477: 462–465. Environ Microbiol 12: 2302–2311.
Neiman J, Guo Y & Rowe-Magnus DA (2011) Chitin-induced Pontiroli A, Rizzi A, Simonet P, Daffonchio D, Vogel TM &
carbotype conversion in Vibrio vulnificus. Infect Immun 79: Monier JM (2009) Visual evidence of horizontal gene
3195–3203. transfer between plants and bacteria in the phytosphere of
Newland JW, Green BA, Foulds J & Holmes RK (1985) transplastomic tobacco. Appl Environ Microbiol 75: 3314–
Cloning of extracellular DNase and construction of a 3322.
DNase-negative strain of Vibrio cholerae. Infect Immun 47: Popa O & Dagan T (2011) Trends and barriers to lateral gene
691–696. transfer in prokaryotes. Curr Opin Microbiol 14: 615–623.
Ng WL & Bassler BL (2009) Bacterial quorum-sensing network Porstendörfer D, Gohl O, Mayer F & Averhoff B (2000)
architectures. Annu Rev Genet 43: 197–222. ComP, a pilin-like protein essential for natural competence
Ng WL, Perez LJ, Wei Y, Kraml C, Semmelhack MF & Bassler in Acinetobacter sp. Strain BD413: regulation, modification,
BL (2011) Signal production and detection specificity in and cellular localization. J Bacteriol 182: 3673–3680.
Vibrio CqsA/CqsS quorum-sensing systems. Mol Microbiol Prudhomme M, Attaiech L, Sanchez G, Martin B & Claverys
79: 1407–1417. JP (2006) Antibiotic stress induces genetic transformability
Nielsen KM, Bones AM & Van Elsas JD (1997) Induced in the human pathogen Streptococcus pneumoniae. Science
natural transformation of Acinetobacter calcoaceticus in soil 313: 89–92.
microcosms. Appl Environ Microbiol 63: 3972–3977. Pruzzo C, Vezzulli L & Colwell RR (2008) Global impact of
Ochman H, Lawrence JG & Groisman EA (2000) Lateral gene Vibrio cholerae interactions with chitin. Environ Microbiol
transfer and the nature of bacterial innovation. Nature 405: 10: 1400–1410.
299–304. Pugsley AP (1993) The complete general secretory pathway in
Odenbreit S, Puls J, Sedlmaier B, Gerland E, Fischer W & gram-negative bacteria. Microbiol Rev 57: 50–108.
Haas R (2000) Translocation of Helicobacter pylori CagA Rasko DA, Webster DR, Sahl JW et al. (2011) Origins of the
into gastric epithelial cells by type IV secretion. Science 287: E. coli strain causing an outbreak of hemolytic-uremic
1497–1500. syndrome in Germany. N Engl J Med 365: 709–717.

FEMS Microbiol Rev 37 (2013) 336–363 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
362 P. Seitz & M. Blokesch

Redfield RJ (1988) Evolution of bacterial transformation: is sex Stokes HW & Gillings MR (2011) Gene flow, mobile genetic
with dead cells ever better than no sex at all? Genetics 119: elements and the recruitment of antibiotic resistance genes
213–221. into Gram-negative pathogens. FEMS Microbiol Rev 35:
Redfield RJ (1991) sxy-1, a Haemophilus influenzae mutation 790–819.
causing greatly enhanced spontaneous competence. Stone BJ & Kwaik YA (1999) Natural competence for DNA
J Bacteriol 173: 5612–5618. transformation by Legionella pneumophila and its association
Redfield RJ (1993a) Evolution of natural transformation: with expression of type IV pili. J Bacteriol 181: 1395–1402.
testing the DNA repair hypothesis in Bacillus subtilis and Suckow G, Seitz P & Blokesch M (2011) Quorum sensing
Haemophilus influenzae. Genetics 133: 755–761. contributes to natural transformation of Vibrio cholerae in a
Redfield RJ (1993b) Genes for breakfast: the have-your-cake- species-specific manner. J Bacteriol 193: 4914–4924.
and-eat-it-too of bacterial transformation. J Hered 84: 400– Sudarsan N, Lee ER, Weinberg Z, Moy RH, Kim JN, Link KH
404. & Breaker RR (2008) Riboswitches in eubacteria sense the
Redfield RJ (2001) Do bacteria have sex? Nat Rev Genet 2: second messenger cyclic di-GMP. Science 321: 411–413.
634–639. Suerbaum S & Josenhans C (2007) Helicobacter pylori
Redfield RJ, Schrag MR & Dean AM (1997) The evolution of evolution and phenotypic diversification in a changing host.
bacterial transformation: sex with poor relations. Genetics Nat Rev Microbiol 5: 441–452.
146: 27–38. Suerbaum S & Michetti P (2002) Helicobacter pylori infection.
Redfield RJ, Cameron AD, Qian Q, Hinds J, Ali TR, Kroll JS N Engl J Med 347: 1175–1186.
& Langford PR (2005) A novel CRP-dependent regulon Suerbaum S, Smith JM, Bapumia K, Morelli G, Smith NH,
controls expression of competence genes in Haemophilus Kunstmann E, Dyrek I & Achtman M (1998) Free
influenzae. J Mol Biol 347: 735–747. recombination within Helicobacter pylori. Proc Natl Acad Sci
Salanoubat M, Genin S, Artiguenave F et al. (2002) Genome USA 95: 12619–12624.
sequence of the plant pathogen Ralstonia solanacearum. Tettelin H, Saunders NJ, Heidelberg J et al. (2000) Complete
Nature 415: 497–502. genome sequence of Neisseria meningitidis serogroup B
Seper A, Fengler VH, Roier S, Wolinski H, Kohlwein SD, strain MC58. Science 287: 1809–1815.
Bishop AL, Camilli A, Reidl J & Schild S (2011) Tettelin H, Masignani V, Cieslewicz MJ et al. (2005) Genome
Extracellular nucleases and extracellular DNA play analysis of multiple pathogenic isolates of Streptococcus
important roles in Vibrio cholerae biofilm formation. Mol agalactiae: implications for the microbial “pan-genome”.
Microbiol 82: 1015–1037. P Natl Acad Sci USA 102: 13950–13955.
Sikorski J, Graupner S, Lorenz MG & Wackernagel W (1998) Thomas CM & Nielsen KM (2005) Mechanisms of, and
Natural genetic transformation of Pseudomonas stutzeri in a barriers to, horizontal gene transfer between bacteria. Nat
non-sterile soil. Microbiology 144: 569–576. Rev Microbiol 3: 711–721.
Sikorski J, Teschner N & Wackernagel W (2002) Highly Tribble GD, Rigney TW, Dao DH, Wong CT, Kerr JE, Taylor BE,
different levels of natural transformation are associated with Pacha S & Kaplan HB (2012) Natural competence is a
genomic subgroups within a local population of major mechanism for horizontal DNA transfer in the oral
Pseudomonas stutzeri from soil. Appl Environ Microbiol 68: pathogen Porphyromonas gingivalis. mBio 3: e00231–11.
865–873. Udden SMN, Zahid MSH, Biswas K, Ahmad QS, Cravioto A,
Smith HO, Gwinn ML & Salzberg SL (1999) DNA uptake Nair GB, Mekalanos JJ & Faruque SM (2008) Acquisition of
signal sequences in naturally transformable bacteria. Res classical CTX prophage from Vibrio cholerae O141 by El Tor
Microbiol 150: 603–616. strains aided by lytic phages and chitin-induced
Solomon JM & Grossman AD (1996) Who’s competent and competence. Proc Natl Acad Sci USA 105: 11951–11956.
when: regulation of natural genetic competence in bacteria. Viscidi RP & Demma JC (2003) Genetic diversity of
Trends Genet 12: 150–155. Neisseria gonorrhoeae housekeeping genes. J Clin Microbiol
Sparling PF (1966) Genetic transformation of Neisseria 41: 197–204.
gonorrhoeae to streptomycin resistance. J Bacteriol 92: 1364– Vos M (2009) Why do bacteria engage in homologous
1371. recombination? Trends Microbiol 17: 226–232.
Spirig T, Tiaden A, Kiefer P, Buchrieser C, Vorholt JA & Hilbi Warskow AL & Juni E (1972) Nutritional requirements of
H (2008) The Legionella autoinducer synthase LqsA Acinetobacter strains isolated from soil, water, and sewage.
produces an alpha-hydroxyketone signaling molecule. J Biol J Bacteriol 112: 1014–1016.
Chem 283: 18113–18123. Wei Y, Perez LJ, Ng WL, Semmelhack MF & Bassler BL (2011)
Stewart GJ, Carlson CA & Ingraham JL (1983) Evidence for an Mechanism of Vibrio cholerae autoinducer-1 biosynthesis.
active role of donor cells in natural transformation of ACS Chem Biol 6: 356–365.
Pseudomonas stutzeri. J Bacteriol 156: 30–35. Weinberg Z, Barrick JE, Yao Z et al. (2007) Identification of
Stingl K, Muller S, Scheidgen-Kleyboldt G, Clausen M & Maier 22 candidate structured RNAs in bacteria using the
B (2010) Composite system mediates two-step DNA uptake CMfinder comparative genomics pipeline. Nucleic Acids Res
into Helicobacter pylori. P Natl Acad Sci USA 107: 1184–1189. 35: 4809–4819.

ª 2012 Federation of European Microbiological Societies FEMS Microbiol Rev 37 (2013) 336–363
Published by Blackwell Publishing Ltd. All rights reserved
Regulation of natural competence in Gram-negative bacteria 363

Whitehead NA, Barnard AM, Slater H, Simpson NJ & Yamamoto S, Morita M, Izumiya H & Watanabe H (2010)
Salmond GP (2001) Quorum-sensing in Gram-negative Chitin disaccharide (GlcNAc)2 induces natural competence
bacteria. FEMS Microbiol Rev 25: 365–404. in Vibrio cholerae through transcriptional and translational
Wiedenbeck J & Cohan FM (2011) Origins of bacterial activation of a positive regulatory gene tfoXVC. Gene 457:
diversity through horizontal genetic transfer and 42–49.
adaptation to new ecological niches. FEMS Microbiol Rev Yamamoto S, Izumiya H, Mitobe J, Morita M, Arakawa E,
35: 957–976. Ohnishi M & Watanabe H (2011) Identification of a chitin-
Wiesner RS, Hendrixson DR & DiRita VJ (2003) Natural induced small rna that regulates translation of the tfoX gene,
transformation of Campylobacter jejuni requires components encoding a positive regulator of natural competence in
of a type II secretion system. J Bacteriol 185: 5408–5418. Vibrio cholerae. J Bacteriol 193: 1953–1965.
Williams PM, Bannister LA & Redfield RJ (1994) The Yildiz FH & Visick KL (2009) Vibrio biofilms: so much
Haemophilus influenzae sxy-1 mutation is in a newly the same yet so different. Trends Microbiol 17: 109–
identified gene essential for competence. J Bacteriol 176: 118.
6789–6794. Young DM, Parke D & Ornston LN (2005) Opportunities for
Wise EM Jr, Alexander SP & Powers M (1973) Adenosine genetic investigation afforded by Acinetobacter baylyi, a
3′:5′-cyclic monophosphate as a regulator of bacterial nutritionally versatile bacterial species that is highly
transformation. P Natl Acad Sci USA 70: 471–474. competent for natural transformation. Annu Rev Microbiol
Wojciechowski MF, Hoelzer MA & Michod RE (1989) 59: 519–551.
DNA repair and the evolution of transformation in Young KT, Davis LM & Dirita VJ (2007) Campylobacter jejuni:
Bacillus subtilis. II. Role of inducible repair. Genetics 121: molecular biology and pathogenesis. Nat Rev Microbiol 5:
411–422. 665–679.
Woodhams KL, Benet ZL, Blonsky SE, Hackett KT & Dillard Zulty JJ & Barcak GJ (1995) Identification of a DNA
JP (2012) Prevalence and detailed mapping of the transformation gene required for com101A+ expression and
gonococcal genetic island in Neisseria meningitidis. J supertransformer phenotype in Haemophilus influenzae.
Bacteriol 194: 2275–2285. P Natl Acad Sci USA 92: 3616–3620.
Xavier KB & Bassler BL (2003) LuxS quorum sensing: more
than just a numbers game. Curr Opin Microbiol 6: 191–197.

FEMS Microbiol Rev 37 (2013) 336–363 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
JOURNAL OF BACTERIOLOGY, Nov. 2001, p. 6288–6293 Vol. 183, No. 21
0021-9193/01/$04.00⫹0 DOI: 10.1128/JB.183.21.6288–6293.2001
Copyright © 2001, American Society for Microbiology. All Rights Reserved.

DNA as a Nutrient: Novel Role for Bacterial


Competence Gene Homologs
STEVEN E. FINKEL1* AND ROBERTO KOLTER2
Department of Biological Sciences, University of Southern California, Los Angeles,
California 90089-1340,1 and Department of Microbiology and Molecular Genetics,
Harvard Medical School, Boston, Massachusetts 021152
Received 5 April 2001/Accepted 7 August 2001

The uptake and stable maintenance of extracellular DNA, genetic transformation, is universally recognized
as a major force in microbial evolution. We show here that extracellular DNA, both homospecific and
heterospecific, can also serve as the sole source of carbon and energy supporting microbial growth. Mutants

Downloaded from http://jb.asm.org/ on August 12, 2014 by guest


unable to consume DNA suffer a significant loss of fitness during stationary-phase competition. In Escherichia
coli, the use of DNA as a nutrient depends on homologs of proteins involved in natural genetic competence and
transformation in Haemophilus influenzae and Neisseria gonorrhoeae. Homologs of these E. coli genes are present
in many members of the ␥ subclass of Proteobacteria, suggesting that the mechanisms for consumption of DNA
may have been widely conserved during evolution.

Horizontal gene transfer in bacteria can occur between or- during extended stationary-phase incubation. The mutated
ganisms of the same or different species via one of three mech- gene showed high homology with a putative competence gene
anisms: conjugation, transduction, or transformation (7). The in H. influenzae. Since for some naturally transformable bac-
last mechanism relies on the cell being able to take up and teria, genetic competence is induced during starvation, and
stably maintain extracellular DNA. Many bacteria are “natu- since no such natural induction of competence under standard
rally competent” for genetic transformation and, at least under laboratory conditions has yet to be described for E. coli, we
some environmental conditions, can take up and integrate ex- chose to address whether the competitive disadvantage of this
tracellular DNA. The mechanisms of DNA uptake in several mutant was due to an inability of that strain to compete for a
naturally competent gram-negative and gram-positive bacteria nutrient resource, namely, extracellular DNA.
have been extensively studied and reviewed (8, 9, 12, 14, 27, 29,
31). The process of transformation in naturally competent bac- MATERIALS AND METHODS
teria involves several steps. First, double-stranded DNA Transposon insertion mutagenesis and initial screen for stationary phase-
(dsDNA) is bound to the surface of the cell and enters a specific competition-defective mutants. All experiments were performed using
compartment where it becomes resistant to exogenous nucle- strains derived from E. coli K-12 strain ZK126 (W3110 ⌬lacU169 tna-2) (33).
ase. Next, one strand of the DNA enters the cytoplasm while ZK1142 is a nalidixic acid-resistant (Nalr) derivative of ZK126. Transposon
insertion mutagenesis of ZK126 using ␭NK1324 was performed as previously
the other strand is degraded (9). Some bacteria, such as Hae-
described (16), resulting in a pool of mutant cells carrying a mini-Tn10d-Camr
mophilus influenzae and Neisseria gonorrhoeae, preferentially insert conferring resistance to chloramphenicol. Using this transposon, a screen
take up homospecific DNA. Specificity of DNA uptake in these for “stationary-phase-specific competition-defective” mutants was performed:
organisms is determined by the presence of “uptake signal mutant candidates were identified after coculture at 37°C of individual transpo-
sequences,” which are overrepresented conserved sequences son insertion mutant strains with wild-type ZK1142 (Nalr) cells in 200 ␮l of
Luria-Bertani (LB) broth in 96-well microtiter plates. Both the Nalr allele (16)
found throughout the genome (28). Finally, after a recombi- and the presence of the chloramphenicol resistance (Camr) marker (S. Finkel
nation event, the new DNA is integrated into the chromosome. and R. Kolter, unpublished data) are neutral in the absence of drug selection.
Natural competence and transformation have not been ob- Transposon insertions which resulted in the loss of mutant cells, as determined
served to occur in many bacterial species, including Escherichia by detecting Nalr cells and few or no Camr cells, after 5 days of competition were
then rescreened in 5-ml batch cultures (see below). Mutant candidates were
coli. It has been proposed that natural competence, in addition
reconstructed by bacteriophage P1 transduction (19) and rescreened. The ZK126
to playing a role in genetic recombination, might serve to allow hofQ::Tn10d-Camr mutant was kindly provided by L. Pratt and R. Kolter (un-
the use of extracellular DNA for a nutritional purpose (22, 24, published data).
29, 30). That is, the uptake of DNA into the cell may have two Batch culture competition assays. E. coli wild-type (ZK1142 Nalr) and mutant
non-mutually exclusive functions: to provide DNA for genetic (Camr) strains were each grown overnight in LB broth (reaching a density of
⬃5 ⫻ 109 CFU/ml.) Cultures were then inoculated 1:1,000,000 (vol/vol) into
transformation and to provide nutrients. While studying mech- fresh LB broth, either in coculture or alone. Cell titers were determined by serial
anisms of survival of E. coli during long-term starvation, we dilution on LB agar plates supplemented with nalidixic acid (20 ␮g/ml) or
identified a transposon insertion mutant that demonstrated an chloramphenicol (30 ␮g/ml) as appropriate. The limit of detection of this titra-
inability to survive when competing with its wild-type parent tion method is ⬍100 CFU/ml.
DNA sequencing of transposon insertion sites. The DNA sequence of the
region flanking the transposon insertion was obtained using an arbitrary PCR-
based technique (4). The primers specific to the mini-Tn10d-Camr element were
* Corresponding author. Mailing address: Department of Biological primer 1L (CTGCCTCCCAGAGCCTG) and primer OUT 1L (CAGGCTCTC
Sciences, Program in Molecular Biology, SHS 172, University of CCCGTGGAGG).
Southern California, Los Angeles, CA 90089-1340. Phone: (213) 821- Preparation of conditioned medium. Filter-sterilized conditioned medium was
1498. Fax: (213) 740-8631. E-mail: sfinkel@usc.edu. prepared as follows. LB cultures (50 ml) were inoculated 1:1,000 (vol/vol) with

6288
VOL. 183, 2001 DNA AS A NUTRIENT 6289

cells from a fresh overnight culture of ZK126 and incubated for 5 days in 250-ml
Erlenmeyer flasks at 37°C with vigorous aeration. After 5 days, cells were pel-
leted and the supernatant was removed and filtered through a 0.2-␮m NYL filter
unit (Nalgene). It was essential that filters be rinsed with at least 100 ml of sterile
distilled water prior to use to ensure removal of trace contaminants on the filter
which are metabolizable by E. coli (data not shown). Supernatants treated with
DNase I were incubated at 37°C with 10 ␮g of DNase I (Sigma Chemical Co., St.
Louis, Mo.) per ml for 20 min prior to inoculation. Cultures were then inocu-
lated, and titers were determined after overnight incubation.
Preparation of minimal medium supplemented with purified DNA. M63 min-
imal medium (1⫻) was prepared as described (19) and supplemented with 1 mM
MgSO4 and 1 ␮g of vitamin B1 per ml. E. coli chromosomal DNA was prepared
as described previously (2). Isolated DNA was sonicated to an average length of
300 to 500 bp and extensively extracted with phenol, phenol-chloroform, chlo-
roform, and ethyl ether. DNA was then precipitated and reprecipitated with
ethanol and resuspended in sterile distilled water immediately before use. It was
essential to use freshly precipitated DNA, most likely because DNA stored for
long periods of time contained easily metabolized nucleotides or other break-
down products from the dsDNA. For the experiments in Fig. 3, chromosomal

Downloaded from http://jb.asm.org/ on August 12, 2014 by guest


DNA was added at a concentration of ⬃6 ␮g/ml. Cultures were inoculated and
titers were determined as described above.
DNA sequence analysis. DNA sequence similarity searches using the basic
BLAST, tBLASTn (1), and Microbial Genomes BLAST algorithms and open
reading frame searches using the ORF Finder program were performed at the
National Center for Biotechnology Information website (http://www.ncbi.nlm.ni-
h.gov/). Protein sequence alignments using the ClustalW 1.8, MAP, and PIMA
algorithms were performed at the Baylor College of Medicine Molecular Biology
website (http://www.hgsc.bcm.tmc.edu/SearchLauncher/).

RESULTS

Identification of the yhiR mutant. We performed a genetic


screen for stationary-phase-specific competition-defective mu-
tants; the analysis of one of those mutants is presented here.
The transposon insertion mutant exhibits no fitness loss during
the exponential phase of growth or upon entry into stationary
phase when cocultured with the wild-type parent. However,
after 2 days of coincubation in stationary phase, the mutant is
outcompeted by wild-type cells and is completely lost from the FIG. 1. Survival patterns of yhiR and the wild-type (WT) parental
culture after 10 to 12 days (Fig. 1A). Yet, when cultured strain in the presence or absence of competition. LB cultures were
incubated for 12 days. (A) Cells grown in coculture; (B) cells grown
separately, the mutant and the wild-type strains show identical separately. The asterisk indicates no detectable counts (limit of detec-
patterns of exponential-phase growth and survival during long- tion of ⬍100 CFU/ml).
term stationary phase incubation (Fig. 1B).
Analysis of the DNA sequence flanking the insertion muta-
tion showed that the transposon inserted into yhiR, a gene of to ⬃5 ⫻ 107 CFU/ml after ⬃3 days (the death phase), and level
previously unassigned function, located at 78.5 min on the E. off at that density for many weeks (10, 33). We hypothesized
coli chromosome. Sequence comparisons identified this gene that during the death phase, DNA might be released from
as a homolog of the comJ (orfJ) gene of H. influenzae; the dead cells into the medium and serve as a nutrient for the
predicted amino acid sequences of ComJ and YhiR are ⬃66% minority of cells that are still alive. (It has already been shown
identical and ⬃85% similar. comJ, located at chromosomal that amino acids are released for catabolic use, presumably
nucleotide position 463327, was first identified as a gene adja- from dead cells, during this time [34].) To determine whether
cent to the H. influenzae competence locus, a cluster of genes DNA was available as a nutrient source and whether the yhiR
that mediate natural competence of this organism (32). mutant is defective in the utilization of released DNA, we
Though direct evidence that comJ plays a role in competence prepared conditioned media by filter sterilization of 5-day-old
is lacking, a deletion mutation removing part of comJ and an cultures of wild-type E. coli. When inoculated at ⬃5 ⫻ 103
adjacent gene causes a transformation-deficient phenotype in CFU/ml, the wild-type cultures reach a density of ⬃8 ⫻ 106
H. influenzae (5). Since the yhiR gene appears to be present in CFU/ml after overnight incubation (Fig. 2). In contrast, yhiR
a single gene operon and transduction of the yhiR::Tn10d- mutant cells reach a density of only ⬃6 ⫻ 105 CFU/ml, less
Camr mutation into fresh wild-type strains confers the compe- than 1/10 that of the wild type. Yet if the conditioned medium
tition-defective phenotype, we can assume that the effect of the is pretreated with DNase I for 20 min and then inoculated with
transposon insertion is due directly to the loss of YhiR activity. either wild-type or yhiR mutant cells, the growth yields are
Conditioned medium experiments indicate the use of DNA identical to that of the wild type growing without added DNase
as a nutrient. During long-term incubation in a rich medium I. These results suggest that DNA is present in the conditioned
(LB broth), viable cell counts of E. coli ZK126 reach a plateau medium and is metabolized by wild-type E. coli but not by the
of ⬃5 ⫻ 109 CFU per ml at the end of exponential phase, drop yhiR mutant.
6290 FINKEL AND KOLTER J. BACTERIOL.

Downloaded from http://jb.asm.org/ on August 12, 2014 by guest


FIG. 2. Growth yields for overnight cultures of wild-type (WT) or
yhiR cells incubated in conditioned medium. Culture medium was
prepared from 5-day-old LB cultures without or with DNase I treat-
ment prior to inoculation. Results are the averages of four experi- FIG. 3. Growth of E. coli utilizing DNA as the sole carbon source.
ments. Cells were grown in M63 minimal medium in the presence or absence
of purified E. coli chromosomal DNA as the sole carbon source.
Results are averages of three experiments. WT, wild type.
Use of defined minimal media supplemented with DNA.
While it is clear that prior to DNase I treatment the condi-
tioned medium contains a nutrient that only the wild-type cells Identification of other com gene homologs in E. coli. The
can utilize, we chose to directly address the question of experiments presented here demonstrate that E. coli can grow
whether this nutrient was DNA. Minimal medium which con- using DNA as a source of carbon and energy and that this
tained only inorganic salts, vitamin B1, and purified E. coli ability depends on a homolog of a gene in the competence
chromosomal DNA as the sole source of carbon and energy locus of another organism, H. influenzae. This prompted us to
was prepared. This medium was inoculated with wild-type or determine whether other H. influenzae competence gene ho-
yhiR mutant cells at ⬃5 ⫻ 103 CFU/ml and incubated for 4 mologs are present in the E. coli genome. In fact, E. coli has
days. As shown in Fig. 3, the growth yields of wild-type cells are homologs to eight H. influenzae genes believed to be involved
more than 50-fold greater than those of the yhiR mutant; in competence and transformation (Table 1). The H. influenzae
wild-type cells reach final densities of 2 ⫻ 105 to 5 ⫻ 105 com (11) locus includes seven genes (comABCDEFG) in a
CFU/ml. This directly demonstrates that E. coli has the ability putative operon plus comJ, which is transcribed divergently
to take up and utilize exogenous DNA as a carbon source. and separated from the cluster by the ponA gene, encoding
Similar results were obtained using heterologous DNAs, in- penicillin-binding protein 1a (Fig. 4). The eight E. coli com
cluding sonicated salmon sperm DNA or synthetic double- gene homologs show various degrees of amino acid sequence
stranded oligonucleotides (data not shown). However, when similarity to the H. influenzae com cluster gene products, rang-
minimal medium containing added DNA is first pretreated ing from 12 to 74% identity (Table 1). In E. coli, these genes
with DNase I, both wild-type and mutant cells grow similarly,
reaching final cell densities of ⬃2 ⫻ 109 CFU/ml (data not
shown). No growth of either wild-type or mutant strains when TABLE 1. Degree of similarity and location of E. coli and
cells were incubated in M63 minimal medium plus DNase I H. influenzae competence genes
alone, with no added DNA or other source of carbon, was H. influenzae E. coli % Identity
observed (data not shown). A similar result is observed when Gene Position (nt) Gene Position (min) (similarity)
wild-type and yhiR mutant strains are grown in minimal me-
dium with 100 mM concentrations of deoxynucleoside triphos- comA 460543 yrfD 75.8 19.8 (33.2)
comB 459748 yrfC 75.8 21.6 (59.2)
phates as the sole carbon source; both strains grow to the same comC 459245 yrfB 75.8 24.2 (49.7)
density (data not shown). Together, these results indicate that comD 458727 yrfA 75.8 11.8 (44.2)
yhiR mutants cannot catabolize dsDNA but can consume DNA comE 458304 hofQ 75.8 36.9 (68.9)
breakdown products. This defect in dsDNA consumption comF 456958 yhgH 76.4 39.5 (69.5)
causes the yhiR mutant to have a competitive disadvantage comG/orfG 456193 yhgI 76.4 73.7 (91.9)
comJ/orfJ 463327 yhiR 78.5 65.8 (85.1)
during coculture with its wild-type parent.
VOL. 183, 2001 DNA AS A NUTRIENT 6291

Downloaded from http://jb.asm.org/ on August 12, 2014 by guest


FIG. 4. Genetic maps of the H. influenzae com locus genes and their homologs. Dashed lines indicate the limits of gene clusters between H.
influenzae and E. coli. Arrows indicate direction of transcription, and arrow length is proportional to gene length (with the exception of ponA/mrcA
homologs, which are not to scale). Genes that are not contiguous are indicated by breaks between genes or clusters. Annotated sequence
information from the completed genomes of H. influenzae and E. coli was obtained from The Institute for Genomic Research (www.tigr.org) and
the University of Wisconsin (www.genetics.wisc.edu) websites, respectively. Except for the published pil locus sequences (6, 18, 25), unannotated
sequence data were obtained as follows: for S. enterica serovar Typhi, www.sanger.ac.uk/projects/S.typhi; for V. cholerae and S. putrefaciens,
www.tigr.org; for N. gonorrhoeae, dna1.chem.ou.edu/gono.html; for P. aeruginosa, www.pseudomonas.com; for K. pneumoniae, genome.wustl.edu/
gsc/Projects/bacterial/klebsiella/klebsiella.shtml.

are found in three loci: five genes (yrfDCBA and hofQ) in an tential advantage of taking up extracellular DNA solely for
apparent operon located at 75.8 min, two genes (yhgHI) in a nutritional purposes, particularly when that DNA is heterolo-
putative bicistronic operon at 76.4 min, and yhiR at 78.5 min gous and less likely to recombine onto the chromosome.
transcribed monocistronically. Whereas there is a chance to lose an essential function or
The identification in E. coli of homologs of the H. influenzae acquire a deleterious allele when taking in extracellular DNA
competence apparatus encouraged us to determine if another as genetic material, using DNA solely as a nutrient source
com gene homolog is essential for growth on DNA as the sole might pose little threat to the cell. Since so many organisms
carbon source. An insertional mutant with a mutation in the E. have developed mechanisms for natural competence and ge-
coli hofQ gene, a homolog of H. influenzae comE, was tested netic transformation, it seems that over evolutionary time, the
for its ability to compete with its wild-type parent and to utilize benefits of maintaining a system for horizontal genetic transfer
DNA as a sole carbon source. hofQ is located in a different outweigh the costs. However, having an additional, and not
operon from yhiR, over 2.5 min away. Under all conditions mutually exclusive, system for nutrient uptake could also pro-
tested it behaved identically to the yhiR mutant, showing a
stationary-phase competition defect during coculture with the
wild type (Fig. 5) and an inability to utilize extracellular DNA
as the sole source of carbon or energy (data not shown). Im-
portantly, both yhiR and hofQ mutants can be artificially in-
duced to competence, by treatment with calcium chloride or by
electroporation (3, 13, 15), as efficiently as the wild type (data
not shown). This indicates that the mechanism of DNA uptake
when DNA is used as a nutrient is distinct from the uptake
mechanism used during induction of artificial competence.

DISCUSSION

We have presented direct evidence supporting a model (9,


22, 24, 30) in which the DNA uptake function of a competence
system can be used for nutrient acquisition rather than (or in
addition to) obtaining and processing DNA for genetic trans-
formation. While the potential evolutionary fitness advantages FIG. 5. Survival patterns of the hofQ mutant and the wild-type
conferred by the acquisition of a beneficial gene by horizontal parental strain (WT) during competition in stationary phase. LB cul-
transfer are obvious (23), also as significant might be the po- tures were incubated for 12 days.
6292 FINKEL AND KOLTER J. BACTERIOL.

vide great benefit. It has been noted that because in naturally expression was observed in cells grown in LB medium during
transformable bacteria, such as B. subtilis, Streptococcus pneu- exponential or early stationary phase, as determined by lacZ
moniae, H. influenzae, and N. gonorrhoeae, only a single strand fusions or reverse transcription-PCR techniques. However, the
enters the cytoplasm during transformation (with the other fact that we observe a phenotype under conditions of compe-
strand being degraded), this mechanism lacks efficiency as a tition in stationary-phase LB cultures and of outgrowth in
nutrient acquisition system (9). That is, if the mechanism for minimal media supplemented with DNA suggests that these
DNA uptake, when the DNA is being used as a source of genes are, in fact, expressed. These differences may be due to
nutrients rather than genetic information, is the same for “non- several factors, including the time points during stationary
competent” bacteria as for naturally transformable species, phase when cells were harvested and the possibility that extra-
then these bacteria would only be able to take up “half” of the cellular DNA may act as an inducer of yrfD/hofM operon gene
DNA as food. While this may be more of an issue for gram- expression.
positive organisms, which do not have an outer membrane, we Bacteria inhabit a wide variety of niches, and within many of
feel that this may be less of a problem for gram-negative these environments extracellular DNA may be available. Esti-
organisms, particularly because current models of natural mates of extracellular DNA concentrations in various marine
transformation for these bacteria suggest that DNA degrada- and aquatic environments range from 0.2 to 44 ␮g/liter (re-
tion of the single strand which does not enter the cytoplasm viewed in reference 17). DNA has been shown to be quite

Downloaded from http://jb.asm.org/ on August 12, 2014 by guest


may take place in the periplasm (9), thus allowing the retention stable when complexed with various clays and soil minerals,
and possible transport of the resulting nucleotides into the binding at concentrations in the microgram to milligram range
cytoplasm for catabolic use. It has also been proposed that per gram of material (17). Extracellular DNA concentrations
bacteria may produce an extracellular nuclease which digests within the mammalian host, including the gastrointestinal tract
dsDNA for catabolic purposes. However, we do not believe and the mucosa in normal human lung, are estimated at hun-
that our data support this model in E. coli, since we would dreds of micrograms per milliliter, reaching as much as 4
expect the products of such a nuclease to help support the mg/ml in the lungs of cystic fibrosis patients (20). With an
growth of not only wild-type strains but also the yhiR or hofQ abundance of DNA available in the environment and primarily
mutants when grown in coculture with the wild-type. of heterologous origin, it is unlikely that much of the DNA
It is interesting that the organization of these genes has been would be suitable for incorporation into the bacterial chromo-
conserved in many organisms. For example, the relative posi- some by homologous recombination, hence reducing its poten-
tion of the E. coli ponA homolog, mrcA, adjacent to the com- tial as a source of genetic diversity. Therefore, it is quite rea-
ABCDE homologs, is similar to that of H. influenzae ponA (Fig. sonable that E. coli and other organisms would take advantage
4). In fact, this organization, with the ponA homolog tran- of this rich nutrient source. Put simply, DNA is “good eating.”
scribed divergently from a five-gene cluster containing ho- It remains to be determined if the system in place for the
mologs of competence genes, is conserved in a wide variety of consumption of DNA arose first and then evolved into a sys-
gram-negative bacteria, including Salmonella enterica serovar tem for genetic transformation, or vice versa. Also remaining
Typhi, Vibrio cholerae, Shewanella putrefaciens, N. gonorrhoeae, to be demonstrated is the possibility that E. coli itself is capable
Klebsiella pneumoniae, and Pseudomonas aeruginosa (Fig. 4). of natural genetic competence and transformation and that we
What is particularly striking about these homologs is that the simply have not found the appropriate environmental condi-
genetic organization of the cluster is highly conserved but tions to observe such a phenomenon in the laboratory. It is
levels of sequence conservation and gene sizes are quite vari- possible that once the DNA is taken up, it can undergo two
able. This suggests that these genes were present in a common fates: recombination-replication, to yield stable transforma-
ancestor and that this genetic organization has been conserved tion, or digestion, to be used as a nutrient. Since the genes used
over evolutionary time, but the functions encoded by these for this process, regardless of the final fate, appear to be
genes may have diverged. As observed by A. Pugsley, homologs homologs of competence genes, we propose to name the E. coli
of comE are part of a superfamily with roles in cellular pro- genes com genes after the designation of the H. influenzae
cesses involving the movement of macromolecules across genes. The ability of the com genes to mediate utilization of
membranes (21), including pilus biogenesis, protein secretion, DNA as a nutrient appears to play a significant role in deter-
competence, and twitching motility; this prompted D. Dubnau, mining the relative fitness of organisms competing for very
in a review of mechanisms of DNA uptake in bacteria, to term limited nutrient resources in natural environments.
them PSTC proteins (9). For example, members of this family
play roles in competence in H. influenzae and N. gonorrhoeae, ACKNOWLEDGMENTS
type IV pilus biogenesis in N. gonorrhoeae and P. aeruginosa, We thank members of the Kolter lab and Leah Macfadyen for
and twitching motility in P. aeruginosa (6, 18, 21, 25). It is helpful discussions; Erik Zinser, George O’Toole, Patrick Stragier,
interesting that P. aeruginosa is not known to be naturally Michael Farrell, and Miriam Susskind for comments on the manu-
competent. script; and Vyacheslav Palchevskiy for technical assistance.
This work was supported by grants from the NIH and NSF to R.K.,
Several investigators have noted that the E. coli yrfDBCA/ a postdoctoral fellowship from the Helen Hay Whitney Foundation to
hofQ genes are homologs of genes involved in the production S.E.F., and a grant from the USC/Norris Comprehensive Cancer Cen-
of type IV pili in pseudomonads (18, 25, 26). In an effort to ter to S.E.F.
address the lack of expression of type IV pili in K-12 E. coli,
REFERENCES
Sauvonnet and coworkers (26) recently measured mRNA lev-
1. Altschul, S. F., et al. 1997. Gapped BLAST and PSI-BLAST: a new gener-
els expressed from the putative hofQ-containing operon (they ation of protein database search programs. Nucleic Acids Res. 25:3389–3402.
refer to the five-gene cluster as hofMNOPQ). No significant 2. Ausubel, F. M., et al. (ed.). 1987. Current protocols in molecular biology.
VOL. 183, 2001 DNA AS A NUTRIENT 6293

John Wiley and Sons, New York, N.Y. Laboratory Press, Cold Spring Harbor, N.Y.
3. Baur, B., K. Hanselmann, W. Schlimme, and B. Jenni. 1996. Genetic trans- 20. Potter, J., S. Spector, L. Matthews, and J. Lemm. 1963. Studies on pulmo-
formation in freshwater: Escherichia coli is able to develop natural compe- nary secretions. 3. The nucleic acids in whole pulmonary secretions from
tence. Appl. Environ. Microbiol. 62:3673–3678. patients with cystic fibrosis, bronchiectasis, and laryngectomy. Am. Rev.
4. Caetano-Annoles, G. 1993. Amplifying DNA with arbitrary oligonucleotide Respir. Dis. 99:909–916.
primers. PCR Methods Appl. 3:85–92. 21. Pugsley, A. P. 1993. The complete general secretory pathway in gram-neg-
5. Dougherty, B. A., and H. O. Smith. 1999. Identification of Haemophilus ative bacteria. Microbiol. Rev. 57:50–108.
influenzae Rd transformation genes using cassette mutagenesis. Microbiol- 22. Redfield, R. 1993. Genes for breakfast: the have-your-cake-and-eat-it-too of
ogy 145:401–409. bacterial transformation. J. Hered. 84:400–404.
6. Drake, S. L., and M. Koomey. 1995. The product of the pilQ gene is essential 23. Redfield, R. J. 1988. Evolution of bacterial transformation: is sex with dead
for the biogenesis of type IV pili in Neisseria gonorrhoeae. Mol. Microbiol. cells ever better than no sex at all? Genetics 119:213–221.
18:975–986. 24. Redfield, R. J., M. R. Schrag, and A. M. Dean. 1997. The evolution of
7. Dreiseikelmann, B. 1994. Translocation of DNA across bacterial mem- bacterial transformation: sex with poor relations. Genetics 146:27–38.
branes. Microbiol. Rev. 58:293–316. 25. Ropp, P. A., and R. A. P. Nicholas. 1997. Cloning and characterization of the
8. Dubnau, D. 1991. Genetic competence in Bacillus subtilis. Microbiol. Rev. ponA gene encoding penicillin-binding protein 1 from Neisseria gonorrhoeae
55:395–424. and Neisseria meningitidis. J. Bacteriol. 179:2783–2787.
9. Dubnau, D. 1999. DNA uptake in bacteria. Annu. Rev. Microbiol. 53:217– 26. Sauvonnet, N., P. Gounon, and A. P. Pugsley. 2000. PpdD type IV pilin of
244. Escherichia coli K-12 can be assembled into pili in Pseudomonas aeruginosa.
10. Finkel, S. E., E. Zinser, and R. Kolter. 2000. Long-term survival and evolu- J. Bacteriol. 182:848–854.
tion in stationary phase, p. 231–238. In G. Storz and R. Hengge-Aronis (ed.), 27. Smith, H. O., D. B. Danner, and R. A. Deich. 1981. Genetic transformation.
Bacterial stress responses. ASM Press, Washington, D.C. Annu. Rev. Biochem. 50:41–68.

Downloaded from http://jb.asm.org/ on August 12, 2014 by guest


11. Fleischmann, R. D., et al. 1995. Whole-genome random sequencing and 28. Smith, H. O., J.-F. Tomb, B. A. Dougherty, R. D. Fleischmann, and J. C.
assembly of Haemophilus influenzae Rd. Science 269:496–512. Venter. 1995. Frequency and distribution of DNA uptake signal sequences in
12. Goodgal, S. H. 1982. DNA uptake in Haemophilus transformation. Annu. the Haemophilus influenzae Rd genome. Science 269:538–540.
Rev. Genet. 16:169–192. 29. Solomon, J. M., and A. D. Grossman. 1996. Who’s competent and when:
13. Huang, R., and R. Reusch. 1995. Genetic competence in Escherichia coli regulation of natural genetic competence in bacteria. Trends Genet. 12:150–
requires poly-beta-hydroxybutyrate/calcium polyphosphate membrane com- 155.
plexes and certain divalent cations. J. Bacteriol. 177:486–489. 30. Stewart, G. J., and C. A. Carlson. 1986. The biology of natural transforma-
14. Kahn, M. E., and H. O. Smith. 1984. Transformation in Haemophilus: a tion. Annu. Rev. Microbiol. 40:211–235.
problem in membrane biology. J. Membr. Biol. 81:89–103. 31. Syvanen, M., and C. I. Kado. 1998. Horizontal gene transfer. Chapman &
15. Kimoto, H., and A. Taketo. 1997. Initial stage of DNA-electrotransfer into E. Hall, London, United Kingdom.
coli cells. J. Biochem. 122:237–242. 32. Tomb, J.-F., H. el-Hajj, and H. O. Smith. 1991. Nucleotide sequence of a
16. Kleckner, N., J. Bender, and S. Gottesman. 1991. Uses of transposons with cluster of genes involved in the transformation of Haemophilus influenzae
emphasis on Tn10. Methods Enzymol. 204:139–180. Rd. Gene 104:1–10.
17. Lorenz, M. G., and W. Wackernagel. 1994. Bacterial gene transfer by natural 33. Zambrano, M. M., D. A. Siegele, M. Almirón, A. Tormo, and R. Kolter. 1993.
genetic transformation in the environment. Microbiol. Rev. 58:563–602. Microbial competition: Escherichia coli mutants that take over stationary
18. Martin, R., A. A. Watson, T. F. McCaul, and J. S. Mattick. 1995. Charac- phase cultures. Science 259:1757–1760.
terization of a five-gene cluster required for the biogenesis of type 4 fimbriae 34. Zinser, E. R., and R. Kolter. 1999. Mutations enhancing amino acid catab-
in Pseudomonas aeruginosa. Mol. Microbiol. 16:497–508. olism confer a growth advantage in stationary phase. J. Bacteriol. 181:5800–
19. Miller, J. H. 1992. A short course in bacterial genetics. Cold Spring Harbor 5807.
Blackwell Science, LtdOxford, UKMMIMolecular Microbiology0950-382X© 2005 The Authors; Journal compilation © 2005 Blackwell Publishing Ltd? 2005592376385Review ArticleNatural transformation of Neisseria gonorrhoeaeH. L. Hamilton
and J. P. Dillard

Molecular Microbiology (2006) 59(2), 376–385 doi:10.1111/j.1365-2958.2005.04964.x


First published online 9 November 2005

MicroReview

Natural transformation of Neisseria gonorrhoeae: from


DNA donation to homologous recombination

Holly L. Hamilton and Joseph P. Dillard* identification of additional transformation genes and
Department of Medical Microbiology and Immunology, provides insight into previous investigations of gono-
University of Wisconsin-Madison Medical School, coccal transformation. Here we review these recent
Madison, WI 53706, USA. developments and address the implications of natural
transformation in the evolution and pathogenesis
N. gonorrhoeae.
Summary

Gonococci undergo frequent and efficient natural


Overview
transformation. Transformation occurs so often that
the population structure is panmictic, with only one Bacteria use three major mechanisms for genetic
long-lived clone having been identified. This high exchange: conjugation, transduction and transformation.
degree of genetic exchange is likely necessary to Neisseria gonorrhoeae is one of at least 44 species of
generate antigenic diversity and allow the persistence bacteria that are naturally competent for genetic transfor-
of gonococcal infection within the human population. mation (Lorenz and Wackernagel, 1994; Sparling, 1966).
In addition to spreading different alleles of genes Phages able to infect N. gonorrhoeae are not known
for surface markers and allowing avoidance of the (Goldberg et al., 1978) and, although conjugative plas-
immune response, transformation facilitates the mids are present in some gonococcal isolates, trans-
spread of antibiotic resistance markers, a continuing formation is the only mechanism for mobilization of
problem for treatment of gonococcal infections. gonococcal chromosomal loci (Sox et al., 1978; Koomey,
Transforming DNA is donated by neighbouring 1998).
gonococci by two different mechanisms: autolysis or Natural transformation is the ability of bacteria to take
type IV secretion. All types of DNA are bound non- up and incorporate macromolecular DNA efficiently. It is
specifically to the cell surface. However, for DNA hypothesized that natural transformation evolved as a
uptake, Neisseria gonorrhoeae recognizes only DNA mechanism for using DNA as a food source (Finkel and
containing a 10-base sequence (GCCGTCTGAA) Kolter, 2001) or to aid in the repair of damaged chromo-
present frequently in the chromosome of neisserial somes (Solomon and Grossman, 1996). For gonococci, it
species. Type IV pilus components and several pilus- is clear that transformation has been harnessed as a
associated proteins are necessary for gonococcal powerful mechanism for generating genetic diversity,
DNA uptake. Incoming DNA is subject to restriction, spreading advantageous alleles and mediating some
making establishment of replicating plasmids difficult forms of antigenic variation (Hobbs et al., 1994; Fudyk
but not greatly affecting chromosomal transforma- et al., 1999; Snyder et al., 2004).
tion. Processing and integration of transforming DNA Neisseria spp. are unusual in that they do not regulate
into the chromosome involves enzymes required for competence like many other naturally competent bacteria,
homologous recombination. Recent research on DNA including Streptococcus pneumoniae, Bacillus subtilis and
donation mechanisms and extensive work on type IV Haemophilus influenzae; rather, the Neisseriae are
pilus biogenesis and recombination proteins have competent for natural transformation during all phases
greatly improved our understanding of natural trans- of growth (Biswas et al., 1977; Goodgal, 1982). The
formation in N. gonorrhoeae. The completion of the competence of N. gonorrhoeae is not dependent on any
gonococcal genome sequence has facilitated the soluble competence factor (Biswas et al., 1977). Like
H. influenzae, N. gonorrhoeae only takes up DNA that
contains a genus-specific uptake sequence. In N. gonor-
Accepted 17 October, 2005. *For correspondence. E-mail rhoeae, the DNA uptake sequence (DUS) is a 10-base
jpdillard@wisc.edu; Tel. (+1) 608 265 2837; Fax (+1) 608 262 8418. sequence (GCCGTCTGAA) that appears very frequently

© 2005 The Authors


Journal compilation © 2005 Blackwell Publishing Ltd
Natural transformation of Neisseria gonorrhoeae 377
in its own chromosome and that of other Neisseria spp. for subsequent transformation by autolysis. However,
The 2.15 Mb genome sequence of strain FA1090 contains N. gonorrhoeae can also donate DNA for transformation
1965 copies of the DUS (Davidsen et al., 2004), thus by type IV secretion.
averaging one DUS for every 1096 bp. The DUS some-
times occurs in inverted repeats between genes forming
Type IV secretion
possible transcriptional terminators (Goodman and
Scocca, 1988). Transformation of N. gonorrhoeae with Type IV secretion systems (T4SS) are one of more
DUS-bearing supercoiled or linear DNA is equally effi- than five multicomponent secretion systems utilized by
cient. It was reported that the transformation efficiency of Gram-negative bacteria to secrete macromolecular sub-
single-stranded DNA (ssDNA) is comparable to that of strates across the cell envelope. T4SS are noted for
double-stranded DNA (dsDNA) (Stein, 1991). However, their ability to secrete both protein and DNA substrates,
this idea is still controversial (see Remaining questions although not all systems secrete both. The presence of
below). a T4SS encoded on the gonococcal genetic island
This review will focus on the studies that have eluci- (GGI) has recently been described. The GGI is present
dated the four major steps in gonococcal transformation: in approximately 80% of N. gonorrhoeae strains and
donation of DNA, binding and uptake of transforming appears to have been horizontally acquired (Dillard and
DNA, processing, and integration into the gonococcal Seifert, 2001). Many of the T4SS homologues encoded
chromosome. It should be noted that many of the in the GGI have been shown to be necessary for the
mechanisms involved in N. gonorrhoeae transformation secretion of gonococcal chromosomal DNA into the
are the same in Neisseria meningitidis transformation. extracellular milieu during growth (Hamilton et al., 2001;
Therefore, where appropriate, insights derived from stud- 2005). The DNA secreted by donors is taken up by
ies of N. meningitidis will also be described and significant recipient bacteria when donors and recipients are
differences will be mentioned. cultured together (Dillard and Seifert, 2001; Hamilton
et al., 2001).
The DNA has been detected in culture medium by use
of a fluorescent DNA-binding dye, as well as by co-culture
Step 1. DNA donation
transformation. The addition of DNase I to the culture
Most descriptions of natural transformation begin with supernatants eliminates the increased fluorescence seen
DNA uptake. The DNA is simply presumed to be in the in the wild-type culture medium as compared with that
environment, and the donor is thought to have no role from the T4SS mutants (Hamilton et al., 2001; 2005), con-
other than having made the supreme sacrifice. However, firming that DNA is the material being detected. Also
the donor may be an important player. What set of events DNase I reduces transfer of chromosomal markers 250-
has resulted in the presence of naked DNA in proximity fold, indicating that DNA is present in the medium and is
to the competent recipient? In the case of N. gonorrhoeae, not transferred by conjugation (Dillard and Seifert, 2001).
the bacteria only live inside human beings and only take Theoretically, the DNA could be released by autolysis of
up DNA containing the genus-specific DUS; therefore, the a portion of the population. However, all measures of
donated DNA must be from siblings of the infecting strain autolysis have shown no differences between the wild-
or from a co-infecting gonococcal strain. In rare instances type strain and the T4SS mutants. Neither the release of
the DNA might come from another species, including a cytoplasmic protein (Dillard and Seifert, 2001) nor the
other Neisseria or H. influenzae (Kroll et al., 1998). Trans- release of radiolabelled RNA (our unpublished observa-
fer from other Neisseria might be expected, as all Neis- tion) was different between wild type and mutants during
seria species use the same DUS. In fact, transfer from growth. Furthermore, cell death was not reduced in the
commensal Neisseria spp. appears to be responsible for T4SS mutants as compared with wild type when analysed
the creation of penA alleles that confer penicillin resis- by live/dead staining and fluorescent microscopy or
tance in N. gonorrhoeae (Spratt et al., 1992). Similarly, fluorometry (Hamilton et al., 2005). Thus, the T4SS
argF of N. meningitidis has regions that appear to have releases DNA into the medium by a method different from
been acquired from commensal Neisseria (Zhou and autolysis.
Spratt, 1992). Transfer from H. influenzae would not be as Little is known about the secreted DNA; how it is pro-
likely as Haemophilus spp. use a different DUS. However, cessed, if secretion is linked to chromosome replication,
the H. influenzae chromosome was found to contain four or if secreted DNA is single- or double-stranded. However,
copies of the Neisseria DUS, and one of these is thought it is easy to imagine type IV secretion as a mechanism of
to have facilitated the transfer of the bio locus from DNA donation for lateral gene transfer within a population
Haemophilus to N. meningitidis and N. gonorrhoeae (Kroll of gonococci without sacrificing vertical transmission or
et al., 1998). Most species are thought to donate DNA risking lysis and death of the entire population.

© 2005 The Authors


Journal compilation © 2005 Blackwell Publishing Ltd, Molecular Microbiology, 59, 376–385
378 H. L. Hamilton and J. P. Dillard
Autolysis slipped-strand mispairing, similar to the well-characterized
mechanism of opacity protein variation in gonococci
Autolysis is a curious phenomenon. As many microbiolo- (Stern et al., 1986; Belland et al., 1996; Snyder et al.,
gists study survival strategies, the idea that a bacterium 2001a). The type III restriction-modification system meth-
would act to promote its own death and lysis seems inher- ylase is required for sequence recognition. Therefore, the
ently wrong. In spite of our feelings on the subject, switching on of the methylase with the restriction subunit
N. gonorrhoeae autolyses readily; in vitro, autolysis nor- already produced could result in destruction of the chro-
mally occurs during stationary phase or during non-growth mosome (Bourniquel and Bickle, 2002). A second type III
conditions, such as exhaustion of nutrients, non-optimal methylase (NG0641) carries 37 copies of a 4 bp repeat
temperature, pH or osmolarity (Hebeler and Young, 1975). and would similarly be expected to phase-vary (Snyder
The major autolysin in vitro is thought to be an N- et al., 2001a). Phase-variable expression of these lethal
acetylmuramyl-L-alanine amidase (Hebeler and Young, activities could allow for random death of a portion of the
1976). An enzyme with this specificity is also the major population.
autolysin in S. pneumoniae, and recent evidence sug- Most gonococci also contain a toxin-antitoxin gene pair
gests that the pneumococcal enzyme acts in the lysis of on the chromosome (J.P. Dillard and H.S. Seifert, unpub-
a portion of the population to provide donor DNA for lished). Although the gonococcal toxin–antitoxin (TA) sys-
transformation (Steinmoen et al., 2002). Two amidase tem has not been characterized, related protein-based
homologues are encoded in the gonococcal chromosome killer systems cause cell death or cell cycle arrest. Inter-
sequence (GenBank Accession No. AE004969). One of estingly, the TA systems appear to respond to the growth
these is similar to Escherichia coli AmpD and is predicted state of the cell (Gerdes et al., 2005). As gonococci are
to act in the breakdown of peptidoglycan monomers in the known to autolyse under non-growth conditions, the TA
cytoplasm, but not in breakdown of the cell wall. The system would be one explanation for the linkage of autol-
second is similar to E. coli AmiC and would be predicted ysis to growth conditions and could explain the massive
to act on macromolecular peptidoglycan. The AmiC homo- death and lysis occurring in cultures following the exhaus-
logue is likely responsible for the peptidoglycan hydrolysis tion of nutrients (Morse and Bartenstein, 1974).
activity identified by Hebeler and Young (1976) and might
play an important part in gonococcal autolysis. Membrane
Step 2. DNA binding and uptake
instability also contributes to autolysis (Cacciapuoti et al.,
1978) and a recent report describes mutations in a The process of DNA binding and uptake by the ever-
phospholipase that result in decreased autolysis in competent gonococcus has been intensely studied and
N. meningitidis and N. gonorrhoeae (Bos et al., 2005). recently reviewed (Chen and Dubnau, 2004). A major
DNA released through gonococcal autolysis can con- remaining question is which, if any, protein acts as a
tribute to DNA donated for natural transformation (Nor- receptor for transforming DNA. Because of its strong
lander et al., 1979). For the 20% of gonococcal strains preference for DUS-containing transforming DNA,
that lack the T4SS and for all N. meningitidis strains, autol- N. gonorrhoeae must distinguish between self DNA
ysis is likely the only mechanism of DNA donation. A (carrying the DUS) and non-self DNA (lacking the DUS).
number of questions arise when considering donation of Thus, DNA binding events are classified into two types:
DNA by cell death. Is cell lysis mediated by host defences, non-specific and specific. Once specific binding of DUS-
or do the bacteria commit suicide by autolysis? How can containing DNA has occurred, uptake across the outer
autolysis be controlled such that one sibling dies while membrane, the murein layer and the inner membrane
another does not? Is autolysis part of a programmed cell follow.
death event that processes DNA to make it better for Nearly all naturally transformable bacteria, both Gram-
transport to, or incorporation into, another cell? Which is positive and Gram-negative, utilize type IV pili (Tfp) or a
better for transformation, DNA released by autolysis or by type IV pilus-like apparatus for the process of DNA uptake.
secretion? Both from an immunological standpoint and for Although the Gram-positive species do not have type IV
understanding DNA donation, the process of autolysis pili, the DNA uptake proteins show significant similarity to
warrants further investigation. proteins required for assembly of Tfp or the related type
One possible mechanism for initiating a programmed II secretion systems (Chen and Dubnau, 2004). Very early
cell death event was described for N. gonorrhoeae. in the studies of gonococcal competence, a correlation
Gonococci were found to carry a phase-variable type III was established between the expression of type IV pili and
restriction-modification enzyme, designated NgoX. The competence. Piliated gonococci were found to be more
coding sequence of the methylase subunit carried 8–12 transformable than non-piliated bacteria (Sparling, 1966).
pentanucleotide repeats near the 5′ end of the gene, At high DNA concentrations, the transformation frequency
allowing the sequence to slip in and out of frame by can exceed 20% (Gunn and Stein, 1996), whereas non-

© 2005 The Authors


Journal compilation © 2005 Blackwell Publishing Ltd, Molecular Microbiology, 59, 376–385
Natural transformation of Neisseria gonorrhoeae 379
piliated variants yield transformation frequencies near mapped to pilT (Wolfgang et al., 1998). PilT is necessary
1 × 10−7 (Sparling, 1966). Both non-specific and specific for twitching motility, which is believed to occur via retrac-
DNA binding has been measured (Dougherty et al., tion of gonococcal Tfp. PilT, whose homologue has been
1979). Only strains expressing retractable Tfp bound DNA shown to be an NTPase (Herdendorf et al., 2002; Aukema
non-specifically, suggesting the hypothesis that electro- et al., 2005), is not necessary for specific binding of DNA
static interactions between DNA and positively charged but is rather involved in uptake of DNA (Aas et al., 2002a;
patches on Tfp might lead to non-specific binding (Aas Herdendorf et al., 2002). Uptake of DNA across the outer
et al., 2002a). It is unclear whether the pilus or a different membrane, i.e. the transfer to a DNase-resistant state,
pilus-like apparatus mediates uptake of DUS-carrying has been demonstrated to be distinct from DNA binding
DNA. Mutations in genes involved in pilus biogenesis [pilE (Aas et al., 2002a), and components involved in compe-
(pilin subunit), pilF (putative NTP-binding protein), pilD tence have been linked to each step. ComE was identified
(prepilin peptidase), pilG (putative inner membrane as the gonococcal orthologue of the B. subtilis compe-
protein) and pilQ (outer membrane secretin protein)] tence protein, ComEA. There are four copies of comE in
reduce natural transformation to undetectable or nearly the gonococcal chromosome and sequential mutation of
undetectable levels, and reduce binding of DUS- comE1–4 additively diminished transformation efficiency
containing DNA to the level of non-specific DNA (Drake (Chen and Gotschlich, 2001). ComE binds DNA non-spe-
and Koomey, 1995; Freitag et al., 1995; Boyle-Vavra and cifically and its presence is necessary for efficient DNA
Seifert, 1996; Aas et al., 2002a). PilC, which is localized uptake (Chen and Gotschlich, 2001). As ComE is likely
to the pilus tip or to the outer membrane, has also been not localized to the gonococcal surface, it has been pro-
implicated in competence (Jonsson et al., 1991; Rudel posed that ComE binds DNA within the periplasm after
et al., 1995). specific DNA uptake has occurred (Chen and Gotschlich,
Even small amounts or altered forms of pilin are suffi- 2001; Aas et al., 2002a).
cient for gonococcal transformation. S- and L-pilin forms Two pilin-like proteins, ComP and PilV, have been
result from recombination events in the pilE expression shown to affect the level of specific DNA binding (Aas
locus; S-pilin variants display intermediate piliation and et al., 2002b). Mutants lacking one or the other of these
secrete a soluble form of the pilin monomer, while L-pilin proteins still produce pili. However, mutants defective in
variants are the result of a tandem duplication in pilE that comP and pilV are greatly reduced in piliation. Increased
generates an overlong pilin that is not assembled into pili. levels of ComP result in high transformation frequencies,
Gibbs et al. (1989) demonstrated that S-pilin variants whereas increased PilV decreases transformation.
display wild-type transformation frequencies, while the Although no regulation of competence has been observed
competence of gonococcal L-pilin variants is reduced in wild-type Neisseria, control of the relative levels of
approximately 35-fold. S-pilin variants make some nor- these two proteins could be used to modulate compe-
mally processed pilin (Long et al., 1998), and it is likely tence (Aas et al., 2002b). Mutants defective in pilC1 and
that L-pilin variants make a small amount as well, thereby pilC2, or mutants lacking one of five newly discovered
fulfilling the need for pilin, if not pili, for transformation. The pilin-like proteins PilH–L were not transformable or were
fact that PilE and the pilus assembly proteins are neces- significantly reduced in transformation competence and
sary for natural transformation suggested that Tfp mediate were found not to localize ComP to the pili (Winther-
DNA transformation directly; however, this hypothesis has Larsen et al., 2005).
not been proven, leaving open the possibility that a pilus- Mutations in the additional genes comA, comL, dca and
like apparatus might be enough to mediate natural trans- tpc also diminish natural transformation. ComL, compe-
formation. In fact, Long et al. (2003) demonstrated that tence lipoprotein, is associated with the murein layer and
gonococcal pilin variants that elaborate virtually no is thought to be involved either directly or indirectly in
observable pili but that still express pilin are competent for puncturing the peptidoglycan layer for incoming DNA
natural transformation and that this transformation is entry into the gonococcus (Facius et al., 1996; Fusseneg-
dependent on the pilus-related proteins PilQ and PilT. This ger et al., 1996a). In addition to competence defects,
finding generates support for the hypothesis that full, mutants in tpc display a reduction in total murein hydro-
extended pili are not necessary for natural transformation lase activity and have cell separation defects; they grow
and that a pilus-like apparatus is sufficient. in tetrapacs rather than as diplococci (Fussenegger et al.,
Two proteins known to be involved in gonococcal com- 1996b). The involvement of ComL and Tpc might indicate
petence do not have a demonstrated role in Tfp biogene- the need for localized openings in the gonococcal pepti-
sis: PilT and ComE (Biswas et al., 1989; Chen and doglycan for DNA uptake to occur, or may be due to
Gotschlich, 2001; Aas et al., 2002a,b). The dud-1 mutant pleiotropic effects on cell wall metabolism. Dca is encoded
was one of the first reported transformation-deficient in the division and cell wall gene cluster, but was not found
mutants (Biswas et al., 1989) and the mutation was later to have any effect on peptidoglycan metabolism. It is

© 2005 The Authors


Journal compilation © 2005 Blackwell Publishing Ltd, Molecular Microbiology, 59, 376–385
380 H. L. Hamilton and J. P. Dillard

Donation Binding and Processing and Homologous


Uptake Recombination

ComP OM
Autolysis
V
C OM
Q
ComE
ComL

Tpc
PG N? PG

D
IM
T F G
ATP

ADP ComA IM

Type IV secretion

A
C A
A
RM A X
B D

RM

Fig. 1. Gonococcal transformation occurs in four steps: DNA donation, binding and uptake of DNA, processing and homologous recombination.
Type IV secretion of DNA and autolysis of gonococci serve as two mechanisms for the donation of DNA for natural transformation. Binding and
uptake requires many pilus-related proteins (single letters indicate Pil gene products) as well as ComP, ComL, ComE, ComA and Tpc. During
uptake, plasmid DNA is processed into linear double-stranded DNA (dsDNA), and at least some of this incoming dsDNA is converted to single-
stranded DNA (ssDNA). Once in the cytoplasm, dsDNA might be processed by restriction-modification enzymes (RM) as well as by RecBCD
nuclease (B, C, D). ssDNA is bound by cytoplasmic RecA (A), which mediates homologous recombination into the gonococcal chromosome.
Small black boxes indicate DNA uptake sequences, which are necessary for efficient uptake. OM, outer membrane; PG, peptidoglycan layer; IM,
inner membrane.

predicted to be an inner membrane protein, and it is DNA and aids in transformation, while Tpc and ComL may
required for competence in N. gonorrhoeae but not allow the DNA to cross the peptidoglycan layer. PilT is
N. meningitidis (Snyder et al., 2001b). ComA is predicted necessary for DNA uptake, theoretically by retracting the
to be an inner membrane protein involved in transport of Tfp or the subunits of a pilus-like apparatus. ComA might
DNA into the cytosol (Facius and Meyer, 1993). This idea help the DNA cross the inner membrane into the cytosol.
is supported by studies of the related protein ComEC in Once within the cytoplasm, this DNA might be processed
B. subtilis. ComEC is a polytopic membrane protein by gonococcal enzymes.
required for DNA uptake and is thought to form a channel
for DNA transport (Draskovic and Dubnau, 2005).
Step 3. DNA processing
Taken together, these data suggest a model for gono-
coccal competence (Fig. 1). Many components involved The fate of transforming DNA in N. gonorrhoeae is depen-
in Tfp biogenesis are necessary for competence, and dent on a number of factors, including its size and nature.
DNA is bound by an as yet unidentified component that Transformation of N. gonorrhoeae with plasmid DNA is
is likely associated with the pilus or a pilus-like apparatus. 1000-fold less efficient than with chromosomally derived
ComP and PilV influence specific binding of DUS-contain- loci (Eisenstein et al., 1977). Sox et al. (1979) demon-
ing DNA. Once across the outer membrane, ComE binds strated that transformation of N. gonorrhoeae with a
© 2005 The Authors
Journal compilation © 2005 Blackwell Publishing Ltd, Molecular Microbiology, 59, 376–385
Natural transformation of Neisseria gonorrhoeae 381
native 7.1 kb, penicillinase-producing plasmid often gen- tively. These genes are separated by 11 kb in strain
erated larger or smaller versions of the plasmid in gono- FA1090. Similarly, linkage was found with str-7 (rpsL) and
coccal transformants. E. coli-propagated versions of the spc-3 (rpsE), which are 13 kb apart. So, 11 kb and 13 kb
same gonococcal plasmids did not transform recipient fragments can be imported. However, no linkage was
gonococci. These experiments provided evidence that found between rif-1 and spc-3, indicating that the ∼25 kb
plasmid DNA is subject to restriction by the gonococcus fragment carrying both of these markers is rarely or never
during transformation (Sox et al., 1979). N. gonorrhoeae imported.
encodes on the order of 16 methyltransferases, many of
which have corresponding endonucleases that, together,
Step 4. Homologous recombination into the
generate an impressive restriction barrier for transforming
gonococcal chromosome
plasmids (Stein et al., 1995). The capacity for plasmid
transformation can be improved by inactivating several of Homologues of enzymes involved in homologous recom-
these restriction-modification systems (Gunn and Stein, bination in E. coli have been identified in N. gonorrhoeae,
1996). Large plasmids are processed into linear dsDNA and a number of these proteins are required for gonococ-
pieces during transformation, before any restriction of cal transformation. Gonococcal genes encoding RecA, B,
DNA that occurs inside the gonococcus (Biswas et al., C, D, O, Q and X are present in the gonococcal chromo-
1986). some, suggesting the existence of both RecBCD and
The restriction barrier to plasmid DNA transformation in RecF pathways for homologous recombination, both of
N. gonorrhoeae is not evident during transformation with which are dependent on RecA (Koomey and Falkow,
chromosomal loci. Although initial studies of gonococcal 1987; Mehr and Seifert, 1998; Stohl and Seifert, 2001).
transformation suggested that transforming DNA enters RecA is necessary to maintain newly acquired chromo-
the cell as double-stranded molecules (Biswas and Spar- somal markers via homologous recombination into the
ling, 1981), more recent studies suggest that at least genome, and RecA-deficient gonococci are not transform-
some ssDNA is formed during transformation and that this able (Koomey and Falkow, 1987).
conversion occurs primarily in the periplasm (Chaussee Mehr and Seifert demonstrated that the RecBCD path-
and Hill, 1998). While the enzyme(s) responsible for this way of homologous recombination, but not the RecF path-
activity have not been identified, a similar mechanism of way, is necessary for efficient gonococcal transformation.
the degradation of one DNA strand during transformation However, gonococci are only 40-fold reduced in transfor-
exists in Gram-positive bacteria (reviewed in Dubnau, mation efficiency in a RecBCD-pathway mutant as well as
1999). The lack of an observable restriction barrier during a RecBCD-/RecF-pathway mutant, suggesting the possi-
transformation of chromosomal loci argues that this DNA, bility of a third, as yet undescribed pathway for homolo-
at least once it is in the cytoplasm, is single-stranded and gous recombination during natural transformation (Mehr
therefore resistant to endonucleases. Incoming ssDNA and Seifert, 1998). Additionally, the homologous recombi-
would not be restricted, and the integrated heteroduplex nation activity that remains in these mutants might reflect
would be methylated on the resident strand and therefore a difference in the nature of the transforming DNA; single-
not restricted. As plasmid transformation results from the stranded transforming DNA might bypass RecBCD and
re-assembly of an intact plasmid by the annealing of bind directly to RecA to mediate recombination (Mehr and
overlapping imported strands, both strands would be Seifert, 1998). Finally, gonococci lacking RecX, which is
unmodified and therefore susceptible to restriction. A presumed to regulate RecA activity, are fivefold reduced
similar restriction barrier is observed for plasmids intro- in DNA transformation (Stohl and Seifert, 2001; Stohl
duced into N. gonorrhoeae by conjugation (Butler and et al., 2003).
Gotschlich, 1991).
The size of the imported DNA has not been extensively
Advantages and implications
studied. Experiments with plasmids (mentioned above)
show that it is easy to recreate a 7 kb plasmid following It is notable that many naturally transformable species are
transformation, but impossible to recreate a 42 kb plas- human pathogens. This might be an artefact of our
mid. This result might indicate that the 42 kb plasmid was human-centric view of the world or the current state of
cut at multiple sites before import, and suggests that funding. However, it is clear that transformation impacts
imported fragments are less than 42 kb. Linkage studies N. gonorrhoeae pathogenesis and consequently, its
performed using chromosomal markers conferring anti- evolutionary survival. The genome sequence of
biotic resistance can now be re-analysed for size using N. gonorrhoeae strain FA1090 reveals the presence of at
the chromosome sequence data. Sarubbi et al. (1974) least six genetic islands, presumably acquired via trans-
showed linkage of antibiotic resistance markers rif-1 and formation from different bacterial species, which may
str-7, mutations known to occur in rpoB and rpsL respec- encode any number of fitness advantages to the organism
© 2005 The Authors
Journal compilation © 2005 Blackwell Publishing Ltd, Molecular Microbiology, 59, 376–385
382 H. L. Hamilton and J. P. Dillard
(GenBank Accession No. AE004969). The GGI, which is Are there effects on recipient gonococci apart from
present in 80% of gonococcal strains (but not FA1090), acquisition of genes? N. meningitidis shows increased
has the characteristics of a horizontally acquired mobile phase variation in response to transforming DNA from
genetic element that might be transferred between gono- heterologous Neisseria species or N. meningitidis strains,
coccal strains by natural transformation (Hamilton et al., due to titrating DNA repair proteins (Alexander et al.,
2005). In addition to the acquisition of large genetic 2004). A similar phenomenon may also occur in
islands, gonococcal transformation is known to generate N. gonorrhoeae. In addition, it might be advantageous for
hybrid porin alleles and is likely to also generate other gonococci to use the recognition of incoming DNA as the
advantageous alleles which affect gonococcal pathogen- indication of the presence of significant numbers of other
esis (Hobbs et al., 1994; Cooke et al., 1998). gonococci, i.e. DNA could act as a quorum-sensing mol-
ecule. Presumably genes might be turned on in response
Remaining questions to transforming DNA, much like genes are regulated in the
response to detection of other well-known quorum-
Although the gonococcus is an excellent model for the
sensing molecules.
study of natural transformation, many questions still
Could the methylation state of incoming DNA aid in
remain. How is DNA donation controlled? Is cell lysis a
distinguishing self gonococcal DNA from non-self gono-
stochastic or a regulated process? What are the identities
coccal DNA? The many restriction-modification systems
of the autolysins? Both a phospholipase and a peptidogly-
of N. gonorrhoeae might differentially methylate genomic
can hydrolase appear to be involved in this process, and
DNA. Taking up DNA of non-self rather than self DNA (i.e.
mechanisms of autolysis were characterized decades ago
DNA from a different gonococcal strain) could be more
(Hebeler and Young, 1976; Cacciapuoti et al., 1978).
advantageous for the purposes of allelic diversification
However, no one has yet been able to identify a nonauto-
and fitness.
lytic mutant. Does type IV secretion of DNA occur in all
Does N. gonorrhoeae possess a competence nuclease
cells or only a portion of the population, and how is it
to degrade one incoming strand during transformation
regulated?
with dsDNA? It is interesting that most models of trans-
What binds the DUS on DNA during transformation? As
formation in both Gram-positive and Gram-negative bac-
mentioned previously, the receptor for species-specific
teria depict a competence nuclease that degrades one
DNA for natural transformation has not yet been identified.
strand of the double-stranded transforming DNA, but only
The prime candidates for this function would appear to be
one competence nuclease has yet been identified, EndA
PilE, ComP, or another protein that is controlled by the
of S. pneumoniae (reviewed in Dubnau, 1999). DNA deg-
presence of ComP. However, no DNA binding to PilE or
radation studies like those performed by Provvedi et al.
ComP could be demonstrated (Mathis and Scocca, 1984;
(2001) in B. subtilis might aid in elucidating the fate of
Aas et al., 2002a), and a ComP-dependent DNA-binding
incoming DNA in N. gonorrhoeae. Along a similar line
protein has not yet been found.
what is the identity of the specific enzymes that linearize
Are gonococci as efficiently transformed with ssDNA as
circular DNA molecules during transformation with plas-
with dsDNA? Stein has demonstrated that ssDNA gener-
mid DNA?
ated by M13 phage transformed gonococci at similar
Natural transformation appears to be an ever-present
levels as double-stranded donor DNA (Stein, 1991); how-
and essential mechanism for the acquisition and
ever, some researchers adhere to the belief that only
exchange of genetic material in the gonococcus. This
dsDNA transforms. ssDNA was also reported to transform
process has undoubtedly contributed to the success of
H. influenzae with an efficiency on the order of ∼50% that
N. gonorrhoeae as a human pathogen. N. gonorrhoeae
of dsDNA (Postel and Goodgal, 1966), although this result
remains an ideal organism for the study of natural trans-
has also been challenged (Mulder and Doty, 1968).
formation and will be a crucial tool for understanding bac-
Recently a third Gram-negative bacterial species,
terial competence for many years to come.
Pseudomonas stutzeri, was shown to be transformed by
ssDNA (Meier et al., 2002). Additionally, N. meningitidis
Acknowledgements
PilQ, which forms the outer membrane ‘pore’ of the pilus
apparatus and is required for natural transformation, has We acknowledge the Gonococcal Genome Sequencing
been recently found to bind ssDNA better than dsDNA Project supported by USPHS/NIH Grant #AI38399, and
(Frye et al., 2004). These facts argue that ssDNA should B.A. Roe, L. Song, S.P. Lin, X. Yuan, S. Clifton, T. Ducey,
be a good substrate for transformation of N. gonorrhoeae. L. Lewis and D.W. Dyer at the University of Oklahoma. We
Additionally, DNA secreted by the gonococcal T4SS, would like to acknowledge the support from our laboratory
which is known to transform recipient bacteria, is presum- through NIH Grant AI47958 to J.P.D. and traineeship on
ably single-stranded (Hamilton et al., 2001). the NIH T32 Grant AI055397 to H.L.H. We apologize to

© 2005 The Authors


Journal compilation © 2005 Blackwell Publishing Ltd, Molecular Microbiology, 59, 376–385
Natural transformation of Neisseria gonorrhoeae 383
those whose work contributed to the fields discussed in Chen, I., and Dubnau, D. (2004) DNA uptake during bacterial
this review but were not specifically mentioned. transformation. Nat Rev Microbiol 2: 241–249.
Chen, I., and Gotschlich, E.C. (2001) ComE, a competence
protein from Neisseria gonorrhoeae with DNA-binding
References activity. J Bacteriol 183: 3160–3168.
Cooke, S.J., Jolley, K., Ison, C.A., Young, H., and Heckels,
Aas, F.E., Wolfgang, M., Frye, S., Dunham, S., Lovold, C., J.E. (1998) Naturally occurring isolates of Neisseria gon-
and Koomey, M. (2002a) Competence for natural transfor- orrhoeae, which display anomalous serovar properties,
mation in Neisseria gonorrhoeae: components of DNA express PIA/PIB hybrid porins, deletions in PIB or novel
binding and uptake linked to type IV pilus expression. Mol PIA molecules. FEMS Microbiol Lett 162: 75–82.
Microbiol 46: 749–760. Davidsen, T., Rodland, E.A., Lagesen, K., Seeberg, E., Tor-
Aas, F.E., Lovold, C., and Koomey, J.M. (2002b) An inhibitor bjorn, R., and Tonjum, T. (2004) Biased distribution of DNA
of DNA binding and uptake events dictates the proficiency uptake sequences towards genome maintenance genes.
of genetic transformation in Neisseria gonorrhoeae: mech- Nucleic Acids Res 32: 1050–1058.
anism of action and links to type IV pilus expression. Mol Dillard, J.P., and Seifert, H.S. (2001) A variable genetic island
Microbiol 46: 1441–1450. specific for Neisseria gonorrhoeae is involved in providing
Alexander, H.L., Richardson, A.R., and Stojiljkovic, I. (2004) DNA for natural transformation and is found more often in
Natural transformation and phase variation modulation in disseminated infection isolates. Mol Microbiol 41: 263–
Neisseria meningitidis. Mol Microbiol 52: 771–783. 278.
Aukema, K.G., Kron, E.M., Herdendorf, T.J., and Forest, K.T. Dougherty, T.J., Asmus, A., and Tomasz, A. (1979) Specific-
(2005) Functional dissection of a conserved motif within ity of DNA uptake in genetic transformation of gonococci.
the pilus protein PilT. J Bacteriol 187: 611–618. Biochem Biophys Res Commun 86: 97–104.
Belland, R.J., Morrison, S.G., and Hogan, D. (1996) A phase- Drake, S.L., and Koomey, M. (1995) The product of the pilQ
variable type III restriction-modification system in Neisseria gene is essential for the biogenesis of type IV pili in Neis-
gonorrhoeae. In Abstracts of the Tenth International Patho- seria gonorrhoeae. Mol Microbiol 18: 975–986.
genic Neisseria Conference. Zollinger, W.D., Frasch, C.E., Draskovic, I., and Dubnau, D. (2005) Biogenesis of a putative
and Deal, C.D. (eds). http://neisseria.org/ipnc. channel protein, ComEC, required for DNA uptake: mem-
Biswas, G.D., and Sparling, P.F. (1981) Entry of double- brane topology, oligomerization and formation of disulphide
stranded deoxyribonucleic acid during transformation of bonds. Mol Microbiol 55: 881–896.
Neisseria gonorrhoeae. J Bacteriol 145: 638–640. Dubnau, D. (1999) DNA uptake in bacteria. Annu Rev Micro-
Biswas, G.D., Sox, T., Blackman, E., and Sparling, P.F. biol 53: 217–244.
(1977) Factors affecting genetic transformation of Neis- Eisenstein, B.I., Sox, T., Biswas, G., Blackman, E., and Spar-
seria gonorrhoeae. J Bacteriol 129: 983–992. ling, P.F. (1977) Conjugal transfer of the gonococcal pen-
Biswas, G.D., Burnstein, K.L., and Sparling, P.F. (1986) Lin- icillinase plasmid. Science 195: 998–1000.
earization of donor DNA during plasmid transformation in Facius, D., and Meyer, T.F. (1993) A novel determinant
Neisseria gonorrhoeae. J Bacteriol 168: 756–761. (comA) essential for natural transformation competence in
Biswas, G.D., Lacks, S.A., and Sparling, P.F. (1989) Trans- Neisseria gonorrhoeae and the effect of a comA mutant on
formation-deficient mutants of piliated Neisseria gonor- pilin variation. Mol Microbiol 10: 699–712.
rhoeae. J Bacteriol 171: 657–664. Facius, D., Fussenegger, M., and Meyer, T.F. (1996)
Bos, M.P., Tefsen, B., Voet, P., Weynants, V., Putten, Sequential action of factors involved in natural competence
J.P.M., and Tommassen, J. (2005) Function of neisserial for transformation of Neisseria gonorrhoeae. FEMS Micro-
outer membrane phospholipase A in autolysis and biol Lett 137: 159–164.
assessment of its vaccine potential. Infect Immun 73: Finkel, S.E., and Kolter, R. (2001) DNA as a nutrient: novel
2222–2231. role for bacterial competence gene homologs. J Bacteriol
Bourniquel, A.A., and Bickle, T.A. (2002) Complex restriction 183: 6288–6293.
enzymes: NTP-driven molecular motors. Biochimie 84: Freitag, N.E., Seifert, H.S., and Koomey, M. (1995) Charac-
1047–1059. terization of the pilF-pilD pilus-assembly locus of Neisseria
Boyle-Vavra, S., and Seifert, H.S. (1996) Uptake-sequence- gonorrhoeae. Mol Microbiol 16: 575–586.
independent DNA transformation exists in Neisseria gon- Frye, S., Assalkhou, R., Balasingham, S., Tuven, H.K.,
orrhoeae. Microbiology 142: 2839–2845. Benam, A.V., Homberset, H., and Tonjum, T. (2004) The
Butler, C.A., and Gotschlich, E.C. (1991) High-frequency meningococcal secretin PilQ: actions and interactions. In
mobilization of broad-host-range plasmids into Neisseria 14th International Pathogenic Neisseria Conference.
gonorrhoeae requires methylation in the donor. J Bacteriol Apicella, M.A., and Seifert, H.S. (eds). http://neisseria.org/
173: 5793–5799. ipnc.
Cacciapuoti, A.F., Wegener, W.S., and Morse, S.A. (1978) Fudyk, T.C., Maclean, I.W., Simonsen, J.N., Njagi, E.N.,
Cell envelope of Neisseria gonorrhoeae: phospholipase Kimani, J., Brunham, R.C., and Plummer, F.A. (1999)
activity and its relationship to autolysis. Infect Immun 20: Genetic diversity and mosaicism at the por locus of Neis-
418–420. seria gonorrhoeae. J Bacteriol 181: 5591–5599.
Chaussee, M.S., and Hill, S.A. (1998) Formation of single- Fussenegger, M., Facius, D., Meier, J., and Meyer, T.F.
stranded DNA during transformation of Neisseria gonor- (1996a) A novel peptidoglycan-linked lipoprotein
rhoeae. J Bacteriol 180: 5117–5122. (ComL) that functions in natural transformation com-

© 2005 The Authors


Journal compilation © 2005 Blackwell Publishing Ltd, Molecular Microbiology, 59, 376–385
384 H. L. Hamilton and J. P. Dillard
petence of Neisseria gonorrhoeae. Mol Microbiol 19: Kroll, J.S., Wilks, K.E., Farrant, J.L., and Langford, P.R.
1095–1105. (1998) Natural genetic exchange between Haemophilus
Fussenegger, M., Kahrs, A.F., Facius, D., and Meyer, T.F. and Neisseria: intergeneric transfer of chromosomal genes
(1996b) Tetrapac (tpc), a novel genotype of Neisseria gon- between major human pathogens. Proc Natl Acad Sci USA
orrhoeae affecting epithelial cell invasion, natural transfor- 95: 12381–12385.
mation competence and cell separation. Mol Microbiol 19: Long, C.D., Madraswala, R.N., and Seifert, H.S. (1998) Com-
1357–1372. parisons between colony phase variation of Neisseria gon-
Gerdes, K., Christensen, S.K., and Lonbner-Olsen, A. (2005) orrhoeae FA1090 and pilus, pilin, and S-pilin expression.
Prokaryotic toxin–antitoxin stress response loci. Nat Rev Infect Immun 66: 1918–1927.
Microbiol 3: 371–382. Long, C.D., Tobiason, D.M., Lazio, M.P., Kline, K.A., and
Gibbs, C.P., Reimann, B.Y., Schultz, E., Kaufmann, A., Haas, Seifert, H.S. (2003) Low-level pilin expression allows for
R., and Meyer, T.F. (1989) Reassortment of pilin genes in substantial DNA transformation competence in Neisseria
Neisseria gonorrhoeae occurs by two distinct mechanisms. gonorrhoeae. Infect Immun 71: 6279–6291.
Nature 338: 651–652. Lorenz, M.G., and Wackernagel, W. (1994) Bacterial gene
Goldberg, I.D., Steinberg, V.I., Siddiqui, A., Hart, E.J., and transfer by natural genetic transformation in the environ-
Schaper, D. (1978) Attempts to isolate bacteriophage ment. Microbiol Rev 58: 563–602.
specific for Neisseria gonorrhoeae. In Immunobiology of Mathis, L.S., and Scocca, J.J. (1984) On the role of pili in
Neisseria gonorrhoeae. Brooks, G.F., Gotschlich, E.C., transformation of Neisseria gonorrhoeae. J Gen Microbiol
Holmes, K.K., Sawyer, W.D. and Young, F.E. (eds). Wash- 130: 3165–3173.
ington, DC: American Society for Microbiology, pp. 55–59. Mehr, I.J., and Seifert, H.S. (1998) Differential roles of homol-
Goodgal, S.H. (1982) DNA uptake in Haemophilus transfor- ogous recombination pathways in Neisseria gonorrhoeae
mation. Annu Rev Genet 16: 169–192. pilin antigenic variation, DNA transformation and DNA
Goodman, S.D., and Scocca, J.J. (1988) Identification and repair. Mol Microbiol 30: 697–710.
arrangement of the DNA sequence recognized in specific Meier, P., Berndt, C., Weger, N., and Wackernagel, W.
transformation of Neisseria gonorrhoeae. Proc Natl Acad (2002) Natural transformation of Pseudomonas stutzeri by
Sci USA 85: 6982–6986. single-stranded DNA requires type IV pili, competence
Gunn, J.S., and Stein, D.C. (1996) Use of a nonselective state, and comA. FEMS Microbiol Lett 207: 75–80.
transformation technique to construct a multiply restriction/ Morse, S.A., and Bartenstein, L. (1974) Factors affecting
modification-deficient mutant of Neisseria gonorrhoeae. autolysis of Neisseria gonorrhoeae. Proc Soc Exp Biol Med
Mol Gen Genet 251: 509–517. 145: 1418–1421.
Hamilton, H.L., Schwartz, K.J., and Dillard, J.P. (2001) Inser- Mulder, C., and Doty, P. (1968) Residual activity of denatured
tion-duplication mutagenesis of Neisseria: use in charac- transforming DNA of Haemophilus influenzae: a naturally
terization of DNA transfer genes in the gonococcal genetic occurring cross-linked DNA. J Mol Biol 32: 423–435.
island. J Bacteriol 183: 4718–4726. Norlander, L., Davies, J., and Normark, S. (1979) Genetic
Hamilton, H.L., Dominguez, N.M., Schwartz, K.J., Hackett, exchange mechanisms in Neisseria gonorrhoeae. J Bac-
K.T., and Dillard, J.P. (2005) Neisseria gonorrhoeae teriol 138: 756–761.
secretes chromosomal DNA via a novel type IV secretion Postel, E.H., and Goodgal, S.H. (1966) Uptake of ‘single-
system. Mol Microbiol 55: 1704–1721. stranded’ DNA in Haemophilus influenzae and its ability to
Hebeler, B.H., and Young, F.E. (1975) Autolysis of Neisseria transform. J Mol Biol 16: 317–327.
gonorrhoeae. J Bacteriol 122: 385–392. Provvedi, R., Chen, I., and Dubnau, D. (2001) NucA is
Hebeler, B.H., and Young, F.E. (1976) Mechanism of autol- required for DNA cleavage during transformation of Bacil-
ysis of Neisseria gonorrhoeae. J Bacteriol 126: 1186– lus subtilis. Mol Microbiol 40: 634–644.
1193. Rudel, T., Facius, D., Barten, R., Scheuerpflug, I., Nonnen-
Herdendorf, T.J., McCaslin, D.R., and Forest, K.T. (2002) macher, E., and Meyer, T.F. (1995) Role of pili and the
Aquifex aeolicus PilT, homologue of a surface motility pro- phase-variable PilC protein in natural competence for
tein, is a thermostable oligomeric NTPase. J Bacteriol 126: transformation of Neisseria gonorrhoeae. Proc Natl Acad
6465–6471. Sci USA 92: 7986–7990.
Hobbs, M.M., Seiler, A., Achtman, M., and Cannon, J.G. Sarubbi, F.A., Jr, Blackman, E., and Sparling, P.F. (1974)
(1994) Microevolution within a clonal population of patho- Genetic mapping of linked antibiotic resistance loci in Neis-
genic bacteria: recombination, gene duplication and hori- seria gonorrhoeae. J Bacteriol 120: 1284–1292.
zontal genetic exchange in the opa gene family of Snyder, L.A.S., Butcher, S.A., and Saunders, N.J. (2001a)
Neisseria meningitidis. Mol Microbiol 12: 171–180. Comparative whole-genome analyses reveal over 100
Jonsson, A.B., Nyberg, G., and Normark, S. (1991) Phase putative phase-variable genes in the pathogenic Neisseria
variation of gonococcal pili by frameshift mutation in pilC, spp. Microbiology 147: 2321–2332.
a novel gene for pilus assembly. EMBO J 10: 477–488. Snyder, L.A.S., Saunders, N.J., and Shafer, W.M. (2001b) A
Koomey, M. (1998) Competence for natural transformation in putatively phase variable gene (dca) required for natural
Neisseria gonorrhoeae: a model system for studies of hor- transformation in Neisseria gonorrhoeae but not Neisseria
izontal gene transfer. APMIS Suppl 84: 56–61. meningitidis is located within the division cell wall (dcw)
Koomey, J.M., and Falkow, S. (1987) Cloning of the recA gene cluster. J Bacteriol 183: 1233–1241.
gene of Neisseria gonorrhoeae and construction of gono- Snyder, L.A.S., Davies, J.K., and Saunders, N.J. (2004)
coccal recA mutants. J Bacteriol 169: 790–795. Microarray genomotyping of key experimental strains of

© 2005 The Authors


Journal compilation © 2005 Blackwell Publishing Ltd, Molecular Microbiology, 59, 376–385
Natural transformation of Neisseria gonorrhoeae 385
Neisseria gonorrhoeae reveals gene complement diversity niae triggers lysis and DNA release from a subfraction of
and five new neisserial genes associated with Minimal the cell population. Proc Natl Acad Sci USA 99: 7681–
Mobile Elements. BMC Genomics 5: 23. 7686.
Solomon, J.M., and Grossman, A.D. (1996) Who’s competent Stern, A., Brown, M., Nickel, P., and Meyer, T.F. (1986)
and when: regulation of natural genetic competence in Opacity genes in Neisseria gonorrhoeae: control of phase
bacteria. Trends Genet 12: 150–155. and antigenic variation. Cell 47: 61–71.
Sox, T.E., Mohammed, W., Blackman, E., Biswas, G., and Stohl, E.A., and Seifert, H.S. (2001) The recX gene potenti-
Sparling, P.F. (1978) Conjugative plasmids in Neisseria ates homologous recombination in Neisseria gonorrhoeae.
gonorrhoeae. J Bacteriol 134: 278–286. Mol Microbiol 40: 1301–1310.
Sox, T.E., Mohammed, W., and Sparling, P.F. (1979) Trans- Stohl, E.A., Brockman, J.P., Burke, K.L., Morimatsu, K.,
formation-derived Neisseria gonorrhoeae plasmids with Kawalczykowski, S.C., and Seifert, H.S. (2003) Escheri-
altered structure and function. J Bacteriol 138: 510–518. chia coli RecX inhibits RecA recombinase and coprotease
Sparling, P.F. (1966) Genetic transformation of Neisseria activities in vitro and in vivo. J Biol Chem 278: 2278–
gonorrhoeae to streptomycin resistance. J Bacteriol 92: 2285.
1364–1371. Winther-Larsen, H.C., Wolfgang, M., Dunham, S., van
Spratt, B.G., Bowler, L.D., Zhang, Q.Y., Zhou, J., and Smith, Putten, J.P.M., Dorward, D., Lovold, C., et al. (2005) A
J.M. (1992) Role of interspecies transfer of chromosomal conserved set of pilin-like molecules controls type IV pilus
genes in the evolution of penicillin resistance in pathogenic dynamics and organelle-associated functions in Neisseria
and commensal Neisseria species. J Mol Evol 34: 115– gonorrhoeae. Mol Microbiol 56: 903–917.
125. Wolfgang, M., Lauer, P., Park, H.S., Brossay, L., Hebert, J.,
Stein, D.C. (1991) Transformation of Neisseria gonorrhoeae: and Koomey, M. (1998) PilT mutations lead to simulta-
physical requirements of the transforming DNA. Can J neous defects in competence for natural transformation
Microbiol 37: 345–349. and twitching motility in piliated Neisseria gonorrhoeae.
Stein, D.C., Gunn, J.S., Radlinska, M., and Piekarowicz, A. Mol Microbiol 29: 321–330.
(1995) Restriction and modification systems of Neisseria Zhou, J., and Spratt, B.G. (1992) Sequence diversity within
gonorrhoeae. Gene 157: 19–22. argF, fbp and recA genes of natural isolates of Neisseria
Steinmoen, H., Knutsen, E., and Havarstein, L.S. (2002) meningitidis: interspecies recombination within the argF
Induction of natural competence in Streptococcus pneumo- gene. Mol Microbiol 6: 2135–2146.

© 2005 The Authors


Journal compilation © 2005 Blackwell Publishing Ltd, Molecular Microbiology, 59, 376–385
REPORTS
States following Cretaceous thickening of the References and Notes 19. C. N. Garzione, J. Quade, P. G. DeCelles, N. B. English,
lithosphere (26). These stable isotope data alone 1. P. Molnar, P. England, Nature 346, 29 (1990). Earth Planet. Sci. Lett. 183, 215 (2000).
2. C. Beaumont, R. A. Jamieson, M. H. Nguyen, B. Lee, 20. D. B. Rowley, R. T. Pierrehumbert, B. S. Currie,
do not preclude Pliocene surface uplift for the Nature 414, 738 (2001). Earth Planet. Sci. Lett. 188, 253 (2001).
northern Sierra Nevada due to mantle de- 3. J. R. Unruh, Geol. Soc. Am. Bull. 103, 1395 (1991). 21. A. Mulch, C. Teyssier, M. A. Cosca, O. Vanderhaeghe,
lamination as has been proposed for the south- 4. J. Wakabayashi, T. L. Sawyer, J. Geol. 109, 539 (2001). T. W. Vennemann, Geology 32, 525 (2004).
ern Sierra Nevada (6–8). In fact, our estimates 5. C. H. Jones, G. L. Farmer, J. Unruh, Geol. Soc. Am. Bull. 22. M. A. Poage, C. P. Chamberlain, Am. J. Sci. 301, 1 (2001).
116, 1408 (2004). 23. N. L. Ingraham, B. E. Taylor, Water Resources Res. 27, 77
of post-Eocene uplift are within the range of 6. G. Zandt et al., Nature 431, 41 (2004). (1991).
elevation change predictable for mantle delam- 7. M. N. Ducea, J. Saleeby, Contrib. Mineral. Petrol. 133, 24. IAEA, http://isohis.iaea.org/userupdate/GNIP2001yearly.xls
ination and may actually constrain the magni- 169 (1998). (2001).
tude of mantle control on Pliocene surface uplift 8. J. Saleeby, M. N. Ducea, D. Clemens-Knott, Tectonics 22, 25. C. D. Henry, M. E. Perkins, Geology 29, 719 (2001).
doi:10.1029/2002TC001374 (2003). 26. P. G. DeCelles, Am. J. Sci. 304, 105 (2004).
in the northern Sierra Nevada (Fig. 4). However, 27. Spatial Climate Analysis Service, Oregon State University,
9. W. R. Dickinson, R. V. Ingersoll, S. A. Graham, Geol. Soc.
they are incompatible with a model of an Am. Bull. 90, 1458 (1979). www.ocs.oregonstate.edu/prism, created April 2005.
Eocene low-elevation landscape that experi- 10. B. Wernicke et al., Science 271, 190 (1996). 28. We acknowledge C. H. Jones and two anonymous
enced substantial (1.5 to 2.0 km) Pliocene 11. M. A. House, B. P. Wernicke, K. A. Farley, Nature 396, 66 reviewers for thorough and constructive reviews. This work
(1998). was supported by NSF grant EAR-0309011 to C.P.C.
surface uplift. Calling upon a Pliocene age for
12. M. A. Poage, C. P. Chamberlain, Tectonics 21, 1601 (2002).
major surface uplift in the northern Sierra 13. M. K. Clark, G. Maheo, J. Saleeby, K. A. Farley, GSA Today Supporting Online Material
Nevada would either require post-Eocene level- 15, 4 (2005). www.sciencemag.org/cgi/content/full/313/5783/87/DC1
Materials and Methods
ing of the mountains with subsequent renewed 14. G. M. Stock, R. S. Anderson, R. C. Finkel, Earth Surf.
Figs. S1 and S2
surface uplift or a tectonic and mechanical Process. Landforms 30, 985 (2005).
15. E. E. Small, R. S. Anderson, Science 270, 277 (1995). Table S1
decoupling of the northern and southern Sierra 16. W. Lindgren, U.S. Geol. Surv. Prof. Pap. 73 (1911). References and Notes
Nevada, both of which are unrecognized in the 17. W. H. Yeend, U.S. Geol. Surv. Prof. Pap. 722 (1974). 8 February 2006; accepted 22 May 2006
geologic record. 18. Available as supporting material on Science Online. 10.1126/science.1125986

Antibiotic Stress Induces Genetic in turn activates the expression of the so-called
early com genes (4), including comAB,
comCDE, and comX. The latter encodes an
Transformability in the Human alternative sigma factor sX (12), which most
probably recognizes a sequence (TACGAATA,
Pathogen Streptococcus pneumoniae hereafter called Pcin) conserved in the putative
promoter regions of the late com genes (4, 9).
The early control of competence induction is
Marc Prudhomme,* Laetitia Attaiech,* Guillaume Sanchez, not yet fully understood. It was first suggested
Bernard Martin, Jean-Pierre Claverys† that competence induction relies simply on passive
CSP accumulation, but we favor an alternative
Natural transformation is a widespread mechanism for genetic exchange in bacteria. model in which CSP production could be
Aminoglycoside and fluoroquinolone antibiotics, as well as mitomycin C, a DNA-damaging temporarily increased in response to changes
agent, induced transformation in Streptococcus pneumoniae. This induction required an intact in environmental conditions (4, 13). We further
competence regulatory cascade. Furthermore, mitomycin C induction of recA was strictly dependent propose that competence in S. pneumoniae is
on the development of competence. In response to antibiotic stress, S. pneumoniae, which lacks a general stress response, playing a role simi-
an SOS-like system, exhibited genetic transformation. The design of antibiotherapy should take into lar to that of the SOS response in Escherichia
consideration this potential of a major human pathogen to increase its rate of genetic exchange coli (4).
in response to antibiotics. We tested this hypothesis by investigating
the effect of mitomycin C, a DNA-damaging
acterial transformation, originally discov- In S. pneumoniae, competence for genetic agent known to induce the SOS response, on

B ered in the human pathogen S. pneumoniae


(1), relies on a process that is inherent
to the species, is independent of extrachromo-
transformation is a transient physiological state
allowing efficient DNA uptake (4) and a pre-
viously unrecognized capacity to kill non-
the com regulon. To monitor competence in-
duction (14), we used transcriptional fusions
with the luc gene from the firefly (Photinus
somal elements, and can be considered the only competent cells (5, 6), a phenomenon referred pyralis) encoding luciferase, the activity of
programmed mechanism for generalized genetic to as pneumococcal fratricide (6) or sobrinicide which can be monitored directly in living S.
exchange in bacteria. It allows the uptake and (7). The development of competence requires pneumoniae cells (14). We first used a fusion of
integration of exogenous DNA in the recipient transcriptional activation of the com regulon, luc with ssbB, a representative of the late com
genome and is considered to be a form of para- which comprises 105 to 124 genes (8, 9), in- genes (8, 9), which encodes a single-stranded
sexuality (2). Transformation is believed to con- cluding recA (10). The RecA protein plays a DNA–binding protein (15). Luciferase activity
tribute to the genetic plasticity of S. pneumoniae key role in transformation by catalyzing homol- was monitored during growth with various con-
and to play a central role in the adaptation of this ogous recombination between the internalized centrations of mitomycin C. Expression of the
pathogen to host defenses (3). We sought to DNA and the recipient genome. The com regulon reporter was stimulated by exposure to 25 to 60
establish whether transformation is induced in is induced when a competence-stimulating ng mlj1 of mitomycin C (Fig. 1A and fig.
response to antibiotic stress. peptide (CSP), encoded by comC and exported S1A), indicating that ssbB was induced when
through a dedicated secretion apparatus ComAB, cells were grown in the presence of the DNA-
Laboratoire de Microbiologie et Génétique Moléculaires, accumulates in the medium and stimulates its damaging agent. No induction of the same
UMR 5100 CNRS-Université Paul Sabatier, 118 route de
Narbonne, 31062 Toulouse Cedex 9, France. receptor, the membrane-bound histidine kinase fusion was detected in a comA mutant
*These authors contributed equally to this work.
ComD (4). It is assumed that ComD then auto- background Ethat is, in a strain unable to export
†To whom correspondence should be addressed. E-mail: phosphorylates and transphosphorylates its CSP and therefore to develop spontaneous
claverys@ibcg.biotoul.fr cognate response regulator ComE (11), which competence (Fig. 1B)^, demonstrating that an

www.sciencemag.org SCIENCE VOL 313 7 JULY 2006 89


REPORTS

A wild type B comA - D E


0.12 12 0.9
15 1.2

RLU/OD (x10 )
-3
RLU/OD (x10 )

0.09 9
-1

CSP MC 0.6

OD492

OD492
10 MC MC 0.8

OD492
0.06 6
0.3
5 0.4 0.03 3

0 0 0
0 100 200 300 0 100 200 300 0 50 100 150 200 0 50 100 150 200
min at 37°C min at 37°C
C
Pcin Pa Fig. 1. Mitomycin C (MC) induction of ssbB and recA. (A and B) Luciferase
cinA recA luc cat 'recA dinF
activity expressed in relative luminescence units (RLU)/optical density (OD)
(triangles) and OD492 (squares) of cultures in CþY medium of (A) strain
R895 (ssbB::luc) and (B) strain R1313 (ssbB::luc, comA–) without and with
mitomycin C (40 ng mlj1). Curves of luciferase activity [with (red triangles) and without (gray triangles) mitomycin C] and OD [with (black squares)
and without (gray squares) mitomycin C] represent compilations of data from 19 and 32 replicate cultures, respectively in (A) and (B). Standard
deviations are indicated for luciferase activities only. (C) Structure of the recA operon with the recA::luc fusion generated by the integration of plasmid
pR432. Plasmid sequences are not drawn to scale. Pcin and Pa are indicated by the small branched arrows. (D and E) Luciferase activity (triangles) and
OD492 (squares) of cultures in CþY medium of strain R1624 (recA::luc, DcomC). In (D), solid blue triangles and solid black squares indicate the
presence of CSP (100 ng mlj1), whereas open gray symbols indicate the absence of CSP. In (E), symbols indicate the following concentrations of
mitomycin C: solid gray symbols (0 ng mlj1), red triangles and open gray squares (25 ng mlj1), open black symbols (100 ng mlj1), and solid black
symbols (400 ng mlj1). Arrows indicate the addition of CSP or mitomycin C after 70 min of incubation.

intact competence regulatory cascade was Fig. 2. Antibiotics induce wild type comA -
required for induction by mitomycin C. These the competence regulon. (A
data established that a DNA-damaging agent to F) Luciferase activity (tri- A D 1.0
induced the com regulon of S. pneumoniae. angles) and OD492 (squares) 30
Mitomycin C is known to trigger the SOS of cultures of strain R895
(ssbB::luc, wild type) [(A) to 20
response in E. coli. DNA damage caused by Nf Nf 0.5
mitomycin C blocks the replication fork, gen- (C)], strain R1313 (ssbB::luc,
10
erating a single-stranded DNA region to which comA–) [(D) and (F)], and
RecA binds to form a nucleoprotein filament strain R1047 (E) (ssbB::luc, 0 0
comA–) [(D) to (F)] in CþY
(RecA*) (16). The coprotease activity of B E
medium with (red and black 15 1.0
RecA* then catalyzes the self-cleavage of the
RLU/OD (x10 )

symbols) and without (gray


-1

SOS repressor LexA (17), which leads to in- symbols) antibiotics. Antibi- Kn Kn
10

OD492
duction of the SOS genes, including recA. In S. otics were added after 70
pneumoniae, the recA gene has been shown to 0.5
min of incubation (arrows) in
be expressed from two promoters: Pcin, which the following concentrations: 5
generates a 5.7-kb-long transcript in competent 11 mg mlj1 of norfloxacin
cells, and Pa, a sA promoter that directs the (Nf) [(A) and (D)], 31.25 mg 0 0
synthesis of a 4.3-kb-long transcript (18) (Fig. mlj1 of kanamycin (Kn) [(B)
1C). The competence-specific induction of and (E)], and 12.5 mg mlj1 10 C F 1.0
Sm Sm
recA (that is, expression from Pcin) accounts of streptomycin (Sm) [(C) and
for 95% of the transformation (10). To establish (F)]. Curves of luciferase ac-
whether mitomycin C could induce recA ex- tivity (with standard devia- 0.5
5
pression, a recA::luc fusion was constructed tions) and OD represent
(Fig. 1C) and validated by measuring its in- compilations of data from 8
duction with CSP (Fig. 1D). Mitomycin C was cultures [(A) and (D)], 15 cul-
found to activate this fusion with kinetics sim- tures [(C) and (F)], or 16 cul- 0 0
ilar to that of ssbB (fig. S2). The recA::luc tures [(B) and (E)] of strains 0 100 200 300 0 100 200 300 400
construct was introduced in a strain unable to with the respective antibi-
otics. See Fig. 1 legend for min at 37°C
develop spontaneous competence because of
details.
the deletion of the entire comC coding region.
The DNA-damaging agent did not induce luc
expression in this genetic background (Fig. 1E), mechanism (7), instead uses the competence mechanisms probably reflects the need for a
demonstrating that the induction of recA by regulatory cascade to coordinate a response to slow accumulation of inducing lesions in both
mitomycin C occurs only from Pcin and is mitomycin C. Induction of the com regulon in species.
therefore strictly dependent on the ability of S. pneumoniae and of the SOS response in E. Several antibiotics are known to induce
this compound to induce competence. This coli occurs only after prolonged incubation, at the SOS response in SOS-proficient bacteria
observation supports the hypothesis that S. È2.5 hours (Fig. 1A) and 2 hours, respectively (20, 21). To investigate the parallels between
pneumoniae, which lacks an SOS-like induction (19). The similar delay of unrelated regulatory competence induction and the SOS response,

90 7 JULY 2006 VOL 313 SCIENCE www.sciencemag.org


REPORTS

Fig. 3. Induction of genetic transformation by streptomycin. (A and B) streptomycin. CSP (100 ng mlj1) was added (right arrow) after 182 min
Luciferase activity (A) and growth [(B), OD492 and colony-forming units (solid black triangles). (D) Genetic transformation in aliquots from cultures in
(CFUs) per milliliter] of cultures of strain R895 with 0 (open squares), (C) taken after 195 min of incubation and mixed with chromosomal DNA
6.25 (open triangles), 12.5 (solid gray triangles), or 25 (solid black carrying a marker conferring resistance to streptomycin (14). The monitoring
squares) mg mlj1 of streptomycin (Sm). Streptomycin was added after 70 of transformation with a marker conferring resistance to streptomycin is
min of incubation [arrow in (A)]. See Fig. 1 legend for details. Cell survival unrelated to the use of streptomycin to induce competence. The number of
was monitored by plating aliquots [dotted lines in (B)]. (C) Luciferase activity streptomycin-resistant chromosomal transformants obtained corresponds to
of strain R895 grown without (squares) and with (triangles) 6.25 mg mlj1 of transformation frequencies of 0.65% (Sm) and 0.57% (SmþCSP).

we measured luciferase synthesis from the expression (Fig. 3C) or the yield of transfor- suggesting that this nucleotide could act as a
ssbB::luc fusion during growth using a wide mants (Fig. 3D), demonstrating full competence signal (4).
range of concentrations of various antibiotics induction by the antibiotic. Chromosomal trans- Whatever the underlying mechanism(s) may
(table S2). Among the protein synthesis inhib- formants were also obtained in parallel cultures be, the induction of the com regulon by various
itors that were tested, kanamycin and strepto- treated with mitomycin C (60 ng mlj1) and antibiotics supports the hypothesis that com-
mycin triggered competence (Fig. 2, B and C, norfloxacin (10 mg mlj1) (fig. S4). petence is a general stress response of S.
and fig. S1A), but erythromycin and tetracy- The induction by mitomycin C and fluo- pneumoniae (4, 7) and is consistent with the
cline did not (fig. S3, A and B). The fluo- roquinolones of the SOS response in E. coli and proposal that CSP is not an effector of quorum
roquinolones (norfloxacin, levofloxacin, and of competence in S. pneumoniae suggests that sensing (7) but is an alarmone that conveys a
moxifloxacin, the latter two of which are used SOS and competence play similar roles in both stress signal (4). As a coordinator of compe-
for the treatment of respiratory tract infections), species. However, the parallel is only partial, tence, CSP enhances the efficiency of transfor-
which target type II topoisomerases, DNA gyrase, because competence was not induced by anti- mation as a rescue process in two ways: by
and topoisomerase IV (22), were found to biotics that disrupt cell wall integrity (such as increasing the number of potential transfor-
induce the ssbB::luc fusion (Fig. 2A and fig. b-lactams), which is contrary to the SOS sys- mants (3) and by triggering fratricide (5) through
S1, B and C). No induction was detected with tem in E. coli (21). Aminoglycosides that in- allolysis (defined as lysis in trans) and the
the DNA gyrase inhibitor novobiocin (fig. duce the com regulon (such as kanamycin and release of DNA from CSP-nonresponsive pneu-
S3B), the RNA polymerase inhibitor rifampi- streptomycin) did not trigger an SOS response mococcal cells. Whenever these cells differ
cin (fig. S3B), the glycopeptide antibiotic in E. coli but instead induced heat-shock pro- from the competent population (for example,
vancomycin (fig. S3B), or with the b-lactams tein expression (23), suggesting that similar during cocolonization), allolysis provides a
ampicillin (fig. S3A) and the third-generation stress signals are processed differently in the source of genetically diverse DNA. Through
cephalosporin, cefotaxime (fig. S3A). No cor- two species. Mitomycin C and fluoroquinolones the coupling of fratricide and transformation,
relation could be made between the intensity of may generate a common signal—chromosome CSP could thus play a crucial role in generating
growth inhibition and the induction of compe- replication arrest—owing to the formation of genetic diversity under stress conditions for a
tence (table S2). Similarly to mitomycin C, the interstrand cross-links (mitomycin C) or the pres- species that seems unable to rely on inducible
induction of ssbB::luc by aminoglycosides and ence of covalently bound topoisomerases (fluo- mutagenic repair (such as the SOS response)
norfloxacin required an intact competence roquinolones) (24). The situation with ribosome (7). Consequently, the high incidence of asymp-
regulatory cascade, because no induction could inhibitors is more complex, because some such tomatic carriage of this pathogen is a major
be detected in a comA mutant (Fig. 2, D to F). as kanamycin and streptomycin act as com- concern, because inappropriate antibiotic treat-
Induction of the com regulon normally al- petence inducers (Fig. 2, B and C), whereas ments could accelerate the occurrence of addi-
lows competent cells to take up and integrate others such as erythromycin and tetracycline do tional resistant clones and promote the evolution
exogenous DNA. To test whether antibiotic- not (fig. S3, A and B). A parallel can be made of virulence.
induced competence resulted in a bona fide with the situation in E. coli: Kanamycin and
References and Notes
transformation, transforming DNA was added streptomycin, which leave the ribosomal A site 1. F. Griffith, J. Hyg. (London) 27, 113 (1928).
to a culture treated with streptomycin. For the empty, induce a heat-shock–like response, 2. J. Maynard Smith, C. G. Dowson, B. G. Spratt, Nature
transformation assay, we selected an intermedi- whereas erythromycin and tetracycline, which 349, 29 (1991).
ate concentration of streptomycin (625 ng mlj1), either fill the A site with aminoacyl–transfer 3. J. P. Claverys, M. Prudhomme, I. Mortier-Barrière,
B. Martin, Mol. Microbiol. 35, 251 (2000).
which did not cause severe killing (Fig. 3B). RNA (tRNA) or block it, trigger a cold-shock– 4. J. P. Claverys, L. S. Håvarstein, Front. Biosci. 7, 1798
Chromosomal transformants were readily like response (23). The level of ppGpp, which (2002).
obtained in the streptomycin-treated culture, is produced by ribosomes that are stalled by a 5. S. Guiral, T. J. Mitchell, B. Martin, J. P. Claverys, Proc.
whereas no transformants were present in the lack of charged tRNAs, decreases upon the Natl. Acad. Sci. U.S.A. 102, 8710 (2005).
6. L. S. Håvarstein, B. Martin, O. Johnsborg, C. Granadel,
control culture (Fig. 3, C and D). The addition addition of erythromycin or tetracycline (or a J. P. Claverys, Mol. Microbiol. 59, 1297 (2006).
of CSP (100 ng mlj1) to the streptomycin- reduction in growth temperature) and increases 7. J. P. Claverys, M. Prudhomme, B. Martin, Annu. Rev.
treated culture did not further increase ssbB::luc as the growth temperature is increased (23, 25), Microbiol. (2006).

www.sciencemag.org SCIENCE VOL 313 7 JULY 2006 91


REPORTS
8. A. Dagkessamanskaia et al., Mol. Microbiol. 51, 1071 16. S. C. Kowalczykowski, Annu. Rev. Biophys. Biophys. Chem. 25. M. Cashel, D. R. Gentry, V. J. Hernandez, D. Vinella, in
(2004). 20, 539 (1991). Escherichia coli and Salmonella typhimurium: Cellular
9. S. Peterson et al., Mol. Microbiol. 51, 1051 (2004). 17. J. W. Little, Proc. Natl. Acad. Sci. U.S.A. 81, 1375 and Molecular Biology, F. C. Neidhardt, Ed. (American
10. I. Mortier-Barrière, A. de Saizieu, J. P. Claverys, B. Martin, (1984). Society for Microbiology Press, Washington, DC, 1996),
Mol. Microbiol. 27, 159 (1998). 18. B. Martin, P. Garcı́a, M. P. Castanié, J. P. Claverys, Mol. pp. 1458–1496.
11. E. V. Pestova, L. S. Håvarstein, D. A. Morrison, Mol. Microbiol. 15, 367 (1995). 26. We thank D. Lane for critical reading of the manuscript.
Microbiol. 21, 853 (1996). 19. J. W. Beaber, B. Hochhut, M. K. Waldor, Nature 427, 72 This work was supported in part by European Union grant
12. P. Luo, D. A. Morrison, J. Bacteriol. 185, 349 (2003). (2004). QLK2-CT-2000-00543.
13. B. Martin, M. Prudhomme, G. Alloing, C. Granadel, 20. I. Phillips, E. Culebras, F. Moreno, F. Baquero,
Supporting Online Material
J. P. Claverys, Mol. Microbiol. 38, 867 (2000). J. Antimicrob. Chemother. 20, 631 (1987).
www.sciencemag.org/cgi/content/full/313/5783/89/DC1
14. To monitor antibiotic-induced competence, cells 21. C. Miller et al., Science 305, 1629 (2004).
Materials and Methods
were incubated in CþY medium with an initial pH 22. C. Levine, H. Hiasa, K. J. Marians, Biochim. Biophys. Acta
Figs. S1 to S4
value adjusted so that spontaneous competence 1400, 29 (1998).
Tables S1 and S2
induction remained a rare event (see supporting 23. R. A. VanBogelen, F. C. Neidhardt, Proc. Natl. Acad. Sci.
References
online material). U.S.A. 87, 5589 (1990).
15. D. E. Grove, S. Willcox, J. D. Griffith, F. R. Bryant, J. Biol. 24. K. Drlica, X. Zhao, Microbiol. Mol. Biol. Rev. 61, 377 27 March 2006; accepted 1 June 2006
Chem. 280, 11067 (2005). (1997). 10.1126/science.1127912

Presymptomatic Detection In order to evaluate the application of PMCA


for the detection of prions in blood during the
presymptomatic phase, 46 hamsters were inocu-
of Prions in Blood lated intraperitoneally with 10% brain homoge-
nate of the 263K scrapie strain, and 38 control
Paula Saá,1,2 Joaquı́n Castilla,1 Claudio Soto1* animals were injected with phosphate-buffered
saline (PBS). At different times during the
Prions are thought to be the proteinaceous infectious agents responsible for transmissible incubation period, groups of animals were killed,
spongiform encephalopathies (TSEs). PrPSc, the main component of the infectious agent, is also blood was collected, and the buffy coat fraction
the only validated surrogate marker for the disease, and its sensitive detection is critical for was separated (13). Samples of the buffy coat
minimizing the spread of the disease. We detected PrPSc biochemically in the blood of hamsters were resuspended directly on healthy hamster
infected with scrapie during most of the presymptomatic phase of the disease. At early stages of brain homogenate and subjected to 144 PMCA
the incubation period, PrPSc detected in blood was likely to be from the peripheral replication of cycles. Three different aliquots were tested from
prions, whereas at the symptomatic phase, PrPSc in blood was more likely to have leaked from the each sample. To refresh the substrate, after a
brain. The ability to detect prions biochemically in the blood of infected but not clinically sick round of PMCA cycling, samples were diluted
animals offers a great promise for the noninvasive early diagnosis of TSEs. 10-fold into normal brain homogenate, followed
by another round of 144 PMCA cycles. This
rion diseases, also called transmissible from the onset of infection to the appearance of procedure was repeated seven times, because

P spongiform encephalopathies (TSEs), are


a group of fatal and infectious neurode-
generative diseases, including Creutzfeldt-Jakob
clinical symptoms (8–10).
PrPSc is not only the main component of the
infectious agent and the most likely cause of
according to our results, this enables the detection
of 20 to 50 molecules of monomeric hamster PrP,
which seems to correspond to a single unit of
disease (CJD) in humans and bovine spongiform the disease, but it is also the only validated infectious oligomeric PrPSc (16).
encephalopathy (BSE), scrapie, and chronic surrogate marker for TSEs (9). However, PrPSc The first group of hamsters was killed 2
wasting disease (CWD) in animals. Prions are concentration is high enough for routine bio- weeks after intraperitoneal inoculation. None of
composed mainly or exclusively of the misfolded chemical detection only in the brain and some the five infected or control animals showed any
prion protein (PrPSc) (1), which replicates in the lymphoid tissues at a time close to the symp- detectable quantity of PrPSc in their blood (Fig.
body, transforming the normal prion protein tomatic stage of the disease (9). The develop- 1 and Table 1). Thus, the PrPSc present in the
(PrPC) into more of the misfolded isoform. ment of highly sensitive presymptomatic assays inoculum disappeared to undetectable levels
Although prion diseases are rare in humans, for the biochemical detection of PrPSc is critical during the first few days after inoculation. PrPSc
the established link between a new variant form for minimizing the spread of the disease (9). was, however, readily detectable in blood 1
of CJD (vCJD) and BSE (2–4) has raised con- One important aim in prion diagnosis is the week later (20 days after inoculation) in 50% of
cern about a potential epidemic in the human noninvasive and presymptomatic biochemical the animals infected but in none of the controls
population. Over the past few years, BSE has detection of PrPSc in biological fluids, particu- (Fig. 1 and Table 1). The highest percentage of
become a substantial health problem affecting larly using blood, a fluid known to contain in- positive animals during the presymptomatic
many countries (5), and it seems now apparent fectivity even before the onset of clinical signs phase was observed 40 days after intraperi-
that vCJD can be iatrogenically transmitted from (6, 11, 12). toneal inoculation, in which the sensitivity of
human to human by blood transfusion (6, 7). PrPSc has been detected in the blood of sick PrPSc detection was 60%. After 60 days, the
Exacerbating this state of affairs is the lack of a animals by means of the protein misfolding cy- detection of PrPSc in blood became harder. In-
reliable test to identify individuals incubating clic amplification (PMCA) technology (13). deed, only one out of five animals scored pos-
the disease during the long and silent period PMCA produces accelerated prion replication, itive at 70 days, whereas none of the five
which dramatically amplifies the quantity of infected hamsters had detectable PrPSc in their
1
George and Cynthia Mitchell Center for Alzheimer’s PrPSc present in a sample (14, 15). In a cyclical blood 80 days after inoculation (Table 1). At
Disease Research, Departments of Neurology, Neuroscience process, large quantities of PrPC are converted the symptomatic stage, which in this experi-
and Cell Biology, and Biochemistry and Molecular Biology, into the misfolded form triggered by the pres- ment was at 114.2 T 5.6 days, 80% of animals
University of Texas Medical Branch, 301 University ence of minute and otherwise undetectable had PrPSc in their blood (Fig. 1). We never
Boulevard, Galveston, TX 77555–0646, USA. 2Centro de
Biologı́a Molecular, Universidad Autónoma de Madrid, amounts of PrPSc. The method is highly specific detected a false positive result in any of the 38
Madrid, Spain. for the detection of PrPSc and leads to a several- control samples analyzed (Table 1).
*To whom correspondence should be addressed. E-mail: million-fold increase in sensitivity as compared The distribution of PrPSc detection at dif-
clsoto@utmb.edu to that of standard Western blot assays (13). ferent times of the incubation period showed

92 7 JULY 2006 VOL 313 SCIENCE www.sciencemag.org


Downloaded from mbio.asm.org on May 23, 2016 - Published by mbio.asm.org
RESEARCH ARTICLE

Epidemic Klebsiella pneumoniae ST258 Is a Hybrid Strain


Liang Chen,a Barun Mathema,a,b Johann D. D. Pitout,c,d,e Frank R. DeLeo,f Barry N. Kreiswirtha
Public Health Research Institute Center, New Jersey Medical School, Rutgers University, Newark, New Jersey, USAa; Department of Epidemiology, Mailman School of
Public Health, Columbia University, New York, New York, USAb; Division of Microbiology, Calgary Laboratory Services, Calgary, Alberta, Canadac; Departments of Pathology
and Laboratory Medicine,d Department of Microbiology, Immunology, and Infectious Diseases,e University of Calgary, Calgary, Alberta, Canada; Laboratory of Human
Bacterial Pathogenesis, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USAf

ABSTRACT Carbapenem-resistant Enterobacteriaceae (CRE), especially Klebsiella pneumoniae carbapenemase (KPC)-producing


K. pneumoniae, pose an urgent threat in health facilities in the United States and worldwide. K. pneumoniae isolates classified as
sequence type 258 (ST258) by multilocus sequence typing are largely responsible for the global spread of KPC. A recent compara-
tive genome study revealed that ST258 K. pneumoniae strains are two distinct genetic clades; however, the molecular origin of
ST258 largely remains unknown, and our understanding of the evolution of the two genetic clades is incomplete. Here we com-
pared the genetic structures and single-nucleotide polymorphism (SNP) distributions in the core genomes of strains from two
ST258 clades and other STs (ST11, ST442, and ST42). We identified an ~1.1-Mbp region on ST258 genomes that is homogeneous
to that of ST442, while the rest of the ST258 genome resembles that of ST11. Our results suggest ST258 is a hybrid clone— 80% of
the genome originated from ST11-like strains and 20% from ST442-like strains. Meanwhile, we sequenced an ST42 strain that
carries the same K-antigen-encoding capsule polysaccharide biosynthesis gene (cps) region as ST258 clade I strains. Comparison
of the cps-harboring regions between the ST42 and ST258 strains (clades I and II) suggests the ST258 clade I strains evolved from
a clade II strain as a result of cps region replacement. Our findings unravel the molecular evolution history of ST258 strains, an
important first step toward the development of diagnostic, therapeutic, and vaccine strategies to combat infections caused by
multidrug-resistant K. pneumoniae.
IMPORTANCE Recombination events and replacement of chromosomal regions have been documented in various bacteria, and
these events have given rise to successful pathogenic clones. Here we used comparative genomic analyses to discover that the
ST258 K. pneumoniae genome is a hybrid— 80% of the chromosome is homologous to ST11 strains, while the remaining 20% is
homologous to that of ST442. Meanwhile, a recent study indicated that ST258 strains can be segregated into two ST258 clades,
with distinct capsule polysaccharide gene (cps) regions. Our analysis suggests ST258 clade I strains evolved from clade II through
homologous recombination of cps region. Horizontal transfer of the cps region appears to be a key element driving the molecular
diversification in K. pneumoniae strains. These findings not only extend our understanding of the molecular evolution of ST258
but are an important step toward the development of effective control and treatment strategies for multidrug-resistant K. pneu-
moniae.

Received 19 May 2014 Accepted 22 May 2014 Published 24 June 2014


Citation Chen L, Mathema B, Pitout JDD, DeLeo FR, Kreiswirth BN. 2014. Epidemic Klebsiella pneumoniae ST258 is a hybrid strain. mBio 5(3):e01355-14. doi:10.1128/mBio.01355-
14.
Editor George Jacoby, Lahey Hospital and Medical Center
Copyright © 2014 Chen et al. This is an open-access article distributed under the terms of the Creative Commons Attribution-Noncommercial-ShareAlike 3.0 Unported license,
which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original author and source are credited.
Address correspondence to Barry N. Kreiswirth, kreiswba@njms.rutgers.edu.
This article is a direct contribution from a member of the American Academy of Microbiology.

K lebsiella pneumoniae carbapenemase (KPC) has emerged as a se-


rious clinical challenge in health care facilities in the United States
and worldwide (1). The blaKPC-harboring plasmid encoding the car-
ical isolates collected between 2002 and 2012 from geographically
diverse sources. Phylogenetic analysis of the core genome of these
isolates revealed that ST258 K. pneumoniae strains are comprised
bapenemase has been found in numerous K. pneumoniae sequence of two distinct genetic clades (ST258 clades I and II), largely due to
types (STs)/clones as well as in other Gram-negative species; how- an ~215-kb region of divergence (RD) that includes genes in-
ever, the vast majority of the global KPC-producing K. pneumoniae volved in capsular polysaccharide (CPS) biosynthesis (7). Further
isolates are associated with a single multilocus sequence type— genotyping analysis with 2 cps-associated genes, wzi and wzy, in
ST258 (2–4). K. pneumoniae ST258 emerged as a notable clinical ST258 and other unrelated K. pneumoniae strains identified the
problem in the middle 2000s in the United States and remains the ST258 clade I genotype in genetically distinct ST42 strains (7).
main ST in the United States and elsewhere (3–6). Interestingly, a GenBank BLAST search using nucleotides encom-
Recently, two KPC-harboring K. pneumoniae ST258 clinical passing the ST258 clade II cps region indicated this region is highly
isolates were sequenced to closure (7). These genomes were used similar to that of a Brazilian ST442 strain, Kp13, which harbors
as references for a comparative genome analysis of 83 ST258 clin- cpsKp13 (8, 9).

May/June 2014 Volume 5 Issue 3 e01355-14 ®

mbio.asm.org 1
Downloaded from mbio.asm.org on May 23, 2016 - Published by mbio.asm.org
Chen et al.

TABLE 1 Features of completely sequenced ST258, ST11, and ST442 genomes


Result for strain:
Parameter a NJST258_1 NJST258_2 HS11286 JM45 ATCC BAA-2146 Kp13
ST no. 258 258 11 11 11 442
Yr 2010 2010 2011 2010 2010 2009
Country United States United States China China United States Brazil
bla genes blaKPC-3, blaOXA-9, blaKPC-3, blaSHV-11
blaKPC-2, blaKPC-2, blaNDM-1, blaCTX-M-15, blaKPC-2, blaCTX-M-2,
blaTEM-1 blaCTX-M-14, blaCTX-M-24, blaCMY-6, blaOXA-1, blaOXA-9, blaTEM-1,
blaTEM-1 blaVEB-3 blaTEM-1, blaSHV-11 blaSHV-12, blaSHV-110
Size (bp) 5,263,329 5,293,301 5,333,942 5,273,813 5,435,369 5,307,003
G⫹C content (%) 57.4 57.5 57.5 57.5 57.3 57.5
No. of CDS 5,475 5,434 5,316 4,872 5,315 5,189
rRNA (n) 25 25 25 25 25 24
16S 8 8 8 8 8 8
23S 8 8 8 8 8 8
5S 9 9 9 9 9 8
tRNA (n) 77 86 87 83 85 86
Plasmids (n) 5 3 6 2 4 6
Prophages (n) 8 7 7 7 8 2
ICEs (n) 2 2 2 1 3 1
IS elements (n) 22 19 23 14 23 31
IS family (n) IS1294 (3), IS1400 (1), IS1294 (2), IS1400 (1), IS1 (1), IS5 (1), IS1400 (1), IS5 (1), IS1 (1), IS1222 (1), IS1 (7), IS10 (1),
IS5 (7), IS5075 (1), IS5 (7), IS903 (2), IS903 (7), ISEc22 ISEcp1 (1), ISKpn1 IS1400 (1), IS2 (2), IS1222 (1), IS4321
IS903 (2), ISKpn1 ISKpn1 (5), (1), ISEcp1 (6), (6), ISKpn18 (6) IS26 (2), IS6100 (1), (2), IS3 (1), IS5 (3),
(5), ISKpn18 (2), ISKpn18 (2) ISKpn1 (6), IS66 (3), ISEc36 (2), IS903 (7), ISEc11
ISSm1 (1) ISKpn18 (1) ISEcp1 (2), ISKpn1 (2), ISEcp1 (1),
(5), ISKpn18 (2), ISKpn1 (5),
ISKpn21 (1) ISKpn21 (1)
a ICE, integrated conjugative element; IS, insertion sequence; CDS, coding sequences.

The emergence and global spread of ST258 and recent reports genomes have 7 of 8 prophages in common, and all harbor
that this clone has diversified as a result of recombination and ICEKp258.1 (see Fig. S1). Sequence comparison among the tonB
replacement of the cps region raise the question of its evolutionary alleles shows tonB79 (in ST258) differs from tonB4 (in ST11) by
history. One speculation is that ST11 (allelic profile 3-3-1-1-1-1- four single-nucleotide polymorphisms (SNPs) and differs from
4), a highly predominant multidrug-resistant clone in Asia and tonB14 (in ST442) by a single SNP. Of note, the three ST11 strains
South America (10–12), and a single-locus variant of ST258 (al- (HS11286, JM45, and ATCC BAA-2146) harbor three different cps
lelic profile 3-3-1-1-1-1-79) gave rise to the ST258 clone through operons, a finding similar to the distinguishing cps genotypes in
the acquisition of the tonB79 allele (13). ST258 clade I and II strains and which supports the observation
To better understand the phylogeny of the ST11 and ST258 that cps switching provides K. pneumoniae the plasticity to change
lineages, we compared the genomes of three ST11 strains its antigenic nature (Fig. 1B).
(HS11286, JM45, and ATCC BAA-2146), three ST258 strains Comparative genome and SNP distribution analyses of the
(NJST258_1, NJST258_2, and Kp1787 [a representative ST258 core chromosome region, as depicted in Fig. 1B, uncovered a
clade I strain present in our collection]), and an ST42 strain, number of surprising findings given the MLST results for ST11
Kp1832. The comparative analysis of these genomes indicates that and ST258. Except for differences in the tonB allele and the region
large and repeated chromosomal exchanges in K. pneumoniae encoding the capsular polysaccharide biosynthetic machinery, the
have occurred between ST11 and ST258, with a significant role for 6 genomes of ST11 and ST258 have a high degree of identity (re-
ST442 in the recent molecular evolution of epidemic ST258 gions of the same color in Fig. 1B). However, further analysis of
strains. the RD and flanking nucleotides revealed that the differences be-
tween ST11 and ST258 were expansive, covering an ~1.1-Mbp
RESULTS contiguous region corresponding to nucleotide positions
Large ~1.1-Mbp recombination region in ST258. To elucidate 1,660,631 to 2,723,681 in strain NJST258_1 (Fig. 1). Significantly,
the phylogenetic relationship among ST258, ST11, and ST442 the ~1.1-Mbp region identified in ST258 clade I and II strains has
strains, we first compared the genome sequences of six closed identical chromosomal nucleotide boundaries (Fig. 2).
Klebsiella pneumoniae strains (Table 1). The size of the chromo- Analysis of SNPs in the genomes of ST258 strains (NJST258_1,
somes was on average ~5.3 Mbp, but the number of mobile ge- NJST258_2, and Kp1787) and ST11 strains (HS11286, JM45, and
netic elements (MGEs), including plasmids, prophages, inte- ATCC BAA-2146) indicated that these strains differ by an average
grated conjugative elements (ICEs), and insertion sequences (IS), of 9,647 SNPs, and 98.1% (9,460 SNPs) of these polymorphisms
varied (Table 1; see Fig. S1 in the supplemental material). Consis- are concentrated in the contiguous ~1.1-Mbp region (identified
tent with multilocus sequence typing (MLST) (Fig. 1A), which above), which represents 20% of the genome (Fig. 3). By compar-
indicates ST11 and ST258 differ by a single locus (the tonB allele ison, the genomes of ST258 strains and ST442 strain Kp13 differed
distinguishes the two sequence types), the 3 ST11 and 2 ST258 by 21,095 SNPs, consistent with their genetically distinct MLST

2 ®

mbio.asm.org May/June 2014 Volume 5 Issue 3 e01355-14


Downloaded from mbio.asm.org on May 23, 2016 - Published by mbio.asm.org
Klebsiella pneumoniae ST258 Is a Hybrid Strain

A
~1.1 MB

NJST258_1
MLST mdh infB tonB gapA phoE pgi rpoB

ST11 1 3 4 3 1 1 3
ST258 1 3 79 3 1 1 3
ST442 2 20 14 10 9 1 11
ST42 1 6 15 2 8 3 1
B
100
Kp13
(ST442) 0
cps 100
HS11286
(ST11) 0
cps 100

JM45
(ST11) 0
cps 100

ATCC BAA-2146
0
(ST11) cps
100

NJST258_1/_2
(ST258 clade II) 0
cps ICE Kp258.2
100
Kp1787
(ST258 clade I) 0
cps
100
Kp1832
(ST42) 0
cps
FIG 1 (A) MLST allele locations on NJST258_1 genome. The light green arrow denotes the genome of NJST258_1, and the light blue region shows the ~1.1-Mbp
putative recombination region between the ST11 and ST442 genomes. The chromosomal positions of the seven MLST housekeeping genes (gapA, infB, mdh, pgi,
phoE, rpoB, and tonB) are illustrated beneath the genome arrow of NJST258_1, and the corresponding allele numbers for ST11, ST258, ST442, and ST42 are listed
below the gene names. (B) Core genome SNP distributions in the ST11, ST258 (clades I and II), ST442, and ST42 strains. The number of SNPs (y axis) per 1,000 nt
is plotted according to the position on the NJST258_1 genome (x axis). Different homogeneous regions (⬎98% identity based on SNP comparisons) are color
coded. Specifically, the ~52-kb cps-containing regions in Kp1787 (ST258 clade I) and Kp1832 (ST42), which are nearly identical in these strains, are shaded in
green. ICEKp258.2 and cps are illustrated by small vertical bars, and the same cps regions are shown in the same color.

profiles (Fig. 1B and SNP matrix in Fig. 3). Most significantly, the cps replacement in ST258 strains. In our previous study, we
SNP mapping revealed contiguous ~1.1-Mbp regions that were identified seven ST42 strains that harbored cps genetic markers
nearly identical in ST258 clade II (NJST258_1 and NJST258_2) (wzy and wzi) that are identical to those in ST258 clade I strains
and ST442 (Kp13) strains, differing by only 206 SNPs (1.0%). As (7), and we hypothesized that this unrelated sequence type (ST42)
depicted in Fig. 1B, the comparative genomic organization and was the donor for the cps region in ST258 clade I strains. As a first
SNP results provide additional support to the idea that the ST258 step toward testing this hypothesis, we used Illumina Miseq to
clade II strain is a hybrid strain containing 80% (~4.2 Mbp) of the sequence the DNA in the cps region of ST42 and ST258 clade I
chromosome from ST11 and 20% (~1.1 Mbp) from ST442 strains and that in strain Kp1832, a representative ST42 isolate in
(Fig. 1B). our strain collection. The gross organization between ST42 and
The ~1.1-Mbp chromosomal region in ST258 clade I and II ST258 strains indicates their distal genetic relatedness (Fig. 1B), a
strains contains the ~215-kb RD and the cps gene cluster (Fig. 2). finding consistent with MLST data. An SNP analysis confirmed
ICEKp258.2 is common to the prototype ST258 strains shown in that there was significant genome divergence between ST258 and
Fig. 1B but is absent from ST442 strain Kp13. To determine the ST42 strains. A total of 31,157 SNPs distinguished the three ST258
level of conservation of ICEKp258.2 among ST258 clinical iso- strains from the ST42 strain, Kp1832, and 27% of these SNPs
lates, we analyzed the DNA contigs of 83 additional ST258 ge- (8,444 SNPs) were located in the ~1.1-Mbp recombination region
nomes sequenced in our previous study (7). We found that (Fig. 3).
ICEKp258.2 is conserved in all of the queried ST258 genomes, and Since the two sequenced reference strains (NJST258_1 and
the insertion of this element in ST258 clade I and II genomes is at NJST258_2) were genotyped as ST258 clade II strains, we created
the same tRNA-Asn site (data not shown). a de novo genome sequence of the prototypic ST258 clade I strain

May/June 2014 Volume 5 Issue 3 e01355-14 ®

mbio.asm.org 3
Downloaded from mbio.asm.org on May 23, 2016 - Published by mbio.asm.org
Chen et al.

cps ICEKp258.2

cps ICEKp258.2 tonB

FIG 2 Upstream and downstream junction SNPs for the ~1.1-Mbp recombination fragment and cps region in ST258, ST442, and ST42 strains. The start site of
the replacement of the ~52-kb cps-harboring region is the same as that of the ~215-kb RD in ST258 II clades (7). Sequences were obtained from Kp1878 (an ST258
clade I strain), NJST258_1 (an ST258 clade II strain), JM45 (an ST11 strain), Kp1832 (an ST42 strain), and Kp13 (an ST442 strain).

(Kp1787) to use as a clade I reference genome. An ~450-kb region lectively, these findings suggest ST258 strains are hybrid strains
of the Kp1787 genome, which contains the entire ~215-kb RD, that arose from an ancestral ST11 strain that acquired an ~1.1-
was used as a reference to examine recombination events between Mbp contiguous chromosomal segment from an ST442-like
Kp1832 (ST42) and Kp1787 (an ST258 clade I strain). The com- strain by DNA recombination/replacement. The identification of
parison revealed a nearly identical region (2 SNPs) spanning distinct blaKPC-harboring elements in ST258, ST11, and ST442
~52 kb that contains the cps region; the alignment of the two strains indicates blaKPC was acquired by ST258 strains via horizon-
regions maps to the start of this DNA replacement at the same tal gene transfer (rather than by vertical gene transmission from
location in the RD region (Fig. 2). In addition, the ICEKp258.2 ST11 or ST442 parental strains) after the recombination events.
element is absent from strain Kp1832, and there is nucleotide
divergence outside the aforementioned ~52-kb region (Fig. 1 and DISCUSSION
2). The current rise of KPC-producing K. pneumoniae infections in
Comparative sequence analysis further showed that the neigh- U.S. health care facilities has been overwhelmingly associated with
boring sequences upstream and downstream from this ~52-kb strains typed as ST258. To better understand the evolutionary
region are identical in ST258 clade I and II strains (Fig. 2). In history of this epidemic clone, we compared the genome se-
addition, the SNP distribution among the 85 ST258 genomes re- quences of ST258 strains, single-locus variant ST11 strains, and
ported in our previous study revealed that 592 (89%) of the 664 other selected K. pneumoniae strain types. Notably, we discovered
SNPs in the RD are located within the ~52-kb cps-harboring re- that ST258 strains are hybrid strains comprised of genomic DNA
gion (7). Together, the genomic findings are consistent with the from ST11 (~80%) and ST442 (~20%)-like strains—presumably
hypothesis that clade I evolved rapidly through the acquisition of the product of a large chromosomal replacement event.
the cps region from an ST42 strain. The evidence provided above Recombination events and replacement of large chromosomal
strongly suggests that replacement of the original (presumably regions have been documented in various bacteria, and there are
ST258 clade II) cps region in clade I contributes largely to the reported examples where the hybrid strains are associated with
noted phylogenetic difference between the two ST258 clades. epidemiological success. Robinson and Enright were the first to
blaKPC-harboring genetic element. We and others have re- report a naturally occurring bacterial hybrid—in this case, in a
ported that the blaKPC gene in ST258 strains is carried exclusively Staphylococcus aureus strain known as ST239 (16). This hybrid
by a Tn3-based transposon, Tn4401 (5, 7, 14). Tn4401 is 10 kb in strain is a pandemic methicillin-resistant S. aureus (MRSA) strain
length, delimited by two 39-bp imperfect inverted repeat (IR) se- responsible for ~90% of the nosocomial infections throughout
quences, and harbors the blaKPC gene, a Tn3 transposase gene mainland Asia and much of South America (17). ST239 is com-
(tnpA), a Tn3 resolvase gene (tnpR), and two insertion sequences, prised of large chromosomal regions from two distantly related
ISKpn6 and ISKpn7 (15) (see Fig. S2 in the supplemental mate- lineages, ST8 and ST30. Approximately 20% of the ST8 genome
rial). In contrast, ST11 and ST442 strains harbor blaKPC- was replaced with an ~550-kb contiguous chromosomal fragment
containing elements that are distinct from those in ST258 strains from an ST30 donor strain, thereby creating ST239. This appar-
(see Fig. S2) and share only ~2 kb of sequence with Tn4401. Col- ently rare molecular event has not been explained or reproduced

4 ®

mbio.asm.org May/June 2014 Volume 5 Issue 3 e01355-14


Downloaded from mbio.asm.org on May 23, 2016 - Published by mbio.asm.org
Klebsiella pneumoniae ST258 Is a Hybrid Strain

46
21
_2

A-
BA
58

86
87

32
T2

12

C
45
17

13

18
S1
JS

C
JM
Kp

Kp

Kp
AT
N

H
NJST258_1 175/43 508/360 8180/8027 8056/7742 12829/12661 21035/217 31304/8566
NJST258_2 445/337 8117/8004 7993/7719 12766/12638 20966/194 31240/8543
Kp1787 8087/7968 8056/7771 12743/12609 21284/503 30927/8222
JM45 3621/3370 6784/6681 28841/8086 30395/7718
HS11286 8183/7915 28522/7761 30080/7414
ATCC BAA-2146 33493/12721 35032/12338
Kp13 31848/8590

(ST42)

(ST258 clade I)

(ST442) (ST11)

(ST258 clade II)

FIG 3 (A) SNP matrix for different K. pneumonaie strains. The matrix is illustrated as (total no. of SNPs/no. of SNPs in the ~1.1-Mbp recombination region).
Green shading indicates the number of SNPs in ST258 strains compared to that in ST11 strains. Orange shading indicates the number of SNPs in ST258 strains
compared to that in ST442 strains. (B) Phylogenetic analysis of the eight isolates based upon 52,135 concatenated SNPs in the core genome.

in the laboratory. In group B Streptococcus (GBS), large single homologous ~1.1-Mbp contiguous region from a strain in the ST442
chromosomal replacement events and multiple localized recom- lineage. This region, which includes the previously described region
bination events occur naturally and can be reproduced in the lab- of difference (RD) and capsular polysaccharide biosynthetic genes,
oratory (18). For GBS, conjugation is the molecular pathway for has molecular scars of multiple localized recombination events, sim-
genomic movement (19). It is worth noting that genetic replace- ilar to the phenomenon in Streptococcus agalactiae (18, 19). The find-
ment of the GBS cps region between unrelated sequence types is ing that ST442 and ST258 have a contiguous chromosomal region in
the common mechanism by which this species alters its surface common and that the nucleotide boundaries between the ST442 and
antigen composition (18). Similarly, cps region replacement- ST258 clade I and II genomes are indistinguishable (Fig. 2) is evidence
associated capsular switching has also been suggested as being an that the recombination event creating an ST11 and ST442 hybrid
intrinsic feature throughout the evolutionary history of Strepto- strain likely occurred once, thereby creating the ST258 clade II lineage
coccus pneumoniae (20). (Fig. 4).
Here we discovered that ST258 clade II strains are hybrid strains in Based on recent genome-scale studies, there have been numer-
which 20% of the K. pneumoniae ST11 genome was replaced with a ous putative chromosomal recombination events involving the

May/June 2014 Volume 5 Issue 3 e01355-14 ®

mbio.asm.org 5
Downloaded from mbio.asm.org on May 23, 2016 - Published by mbio.asm.org
Chen et al.

ST42
ST11-like
cps
(cps 258-1)
ICEKp258.2

ST258 ST258
clade II clade I
cps 258-2 cps 258-1

ST442-like
cps Kp13
(cps 258-2)
FIG 4 Hypothesized evolutionary history in K. pneumoniae ST258 strains.

region encoding the CPS biosynthetic machinery (7). ST258 the former is associated with a broad range of plasmids and car-
clades I and II have distinct cps regions, and this is also true of the bapenemases (KPC, VIM, IMP, NDM, and OXA-48) (10–12, 24–
three ST11 strains analyzed in this study (Fig. 1B), further sup- 28), whereas ST258 strains predominantly harbor KPC. Taken
porting the notion that DNA exchange in and around the cps together, the association of ICEKp258.2 with ST258 K. pneu-
regions may be a general mechanism used by K. pneumoniae to moniae strains raises the possibility that this element may contrib-
rapidly diversify and that novel clades arise through cps switching ute to epidemiological success of this sequence type. To investigate
between ST258 (clade I or II) and unrelated K. pneumoniae se- whether ICEKp258.2 could potentially be an “epidemic clone-
quence types. specific” target, we are currently investigating the impact of alter-
The presence, absence, and diversity of genetic landmarks ing the type IV pilus gene cluster and the type III restriction-
within the acquired ~1.1-Mbp region provide clues into ST258’s modification system in this element.
recent evolutionary origin and the extent of its genomic plasticity. Taken together, our findings underscore the role of recombi-
ICEKp258.2, which is absent in ST442, is present in both ST258 nation in the rapid evolution of clinical strains of K. pneumoniae
clades, where its chromosomal insertion site is conserved, suggest- in both creating hybrid clones and in more localized chromo-
ing that this ICE was acquired after the major genome recombi- somal replacements that alter antigenic presentation and ulti-
nation event that gave rise to ST258 (Fig. 4). In support of the mately divert the host response.
notion that ICEKp258.2 is a relatively recent acquisition, the G⫹C
content of ICEKp258.2 is 37.1%, significantly less than the MATERIALS AND METHODS
~57.5% G⫹C content of the entire K. pneumoniae chromosome. Sequence information. Data used in comparative analysis were down-
Taken together, these observations provide evidence that loaded from the NCBI database (http://www.ncbi.nlm.nih.gov/genome/
ICEKp258.2 is exogenous and was likely acquired once by ST258, genomes/815), including complete genome sequences and annotation of
before the recombination events involving the cps regions in K. pneumoniae isolates HS11286 (CP003200) (29), JM45 (CP006656),
clades I and II (Fig. 4). Moreover, the replacement of the ST258 ATCC BAA-2146 (CP006659) (30), Kp13 (CP003999) (8),
clade II cps region with that from ST42 (thus creating clade I) NJST258_1(CP006923) (7), and NJST258_2 (CP006918) (7). Additional
occurred after the acquisition of ICEKp258.2 (Fig. 4). sequence data were retrieved from our recent study on K. pneumoniae
ST258 (7).
Adler and colleagues investigated the association of the
Genome sequencing and assembly. Strain Kp1832 was selected from
ICEKp258.2 with ST258 by testing160 K. pneumoniae strains with one of the seven ST42 K. pneumoniae isolates that carry the same wzy and
diverse sequence types for the presence of pilV, a gene carried on wzi genes as ST258 cps-1 strains (7). Genomic DNA isolation and library
ICEKp258.2 (21). They found that pilV was present only in ST258 preparation were performed as described previously (7). The genome was
and genetically related strains. Based on sequence analysis, sequenced using an Illumina MiSeq platform, which generated 250-bp
ICEKp258.2 harbors a type IV pilus gene cluster and a type III paired-end reads. De novo assembly for Kp1832 and Kp1787 (a represen-
restriction-modification system. A type IV pilus could increase the tative ST258 clade I strain, selected from our previous study [7]) was
uptake and exchange of DNA, such as plasmids, as well as facilitate accomplished by using a combination of CLC genomic workbench (v
adherence to living and nonliving surfaces— e.g., the human gut 7.0.3; CLC Bio, Aarhus, Denmark), Mira (31), and Velvet (32). The best
or the environment (22)—which may in part explain the high assemblies from each method were combined using Geneious Pro soft-
ware in order to generate the supercontig for the cps-harboring element.
transmissibility of ST258 strains and the movement of KPC genes.
Comparative genomics analysis. Visualization of circular genome
Additionally, a type III restriction-modification system could comparisons was performed using the BLAST ring image generator
serve in “host specificity” regarding the exchange of certain com- (BRIG) (33). Prophages were identified by PHAST (34). Insertion se-
patible plasmids and other mobile elements (23). Restriction of quences were identified using the IS Finder database (http://www
plasmids and specific mobile elements may explain the differences -is.biotoul.fr). De novo assembled contigs from Kp1832 and Kp1787 were
observed between ST11 (which lacks ICEKp258.2) and ST258, as ordered and oriented relative to the NJST258_1 genome and then com-

6 ®

mbio.asm.org May/June 2014 Volume 5 Issue 3 e01355-14


Downloaded from mbio.asm.org on May 23, 2016 - Published by mbio.asm.org
Klebsiella pneumoniae ST258 Is a Hybrid Strain

bined together as a pseudochromosome using the Mauve contig mover veals remarkable genome plasticity and a wide repertoire of virulence and
(35). Multiple genome sequence alignments and comparison analysis resistance mechanisms. BMC Genomics 15:54. http://dx.doi.org/10.1186/
were then performed with Mauve (35). For core genomic analysis, SNPs 1471-2164-15-S2-P54.
located on the MGEs, including prophases, ICEs, and insertion elements, 9. Chen L, Chavda KD, Findlay J, Peirano G, Hopkins K, Pitout JD,
as well as those on rRNAs and tRNAs, were excluded. The concatenated Bonomo RA, Woodford N, Deleo FR, Kreiswirth BN. 14 April 2014.
Multiplex PCR for identification of two capsular types in epidemic KPC-
SNPs were used to generate a consensus phylogenetic tree by the maxi- producing Klebsiella pneumoniae ST258 strains. Antimicrob. Agents Che-
mum likelihood method based on the Tamura-Nei model with the MEGA mother. http://dx.doi.org/10.1128/AAC.02673-14.
5 software (36). The SNP distribution among different genome sequences 10. Qi Y, Wei Z, Ji S, Du X, Shen P, Yu Y. 2011. ST11, the dominant clone
was inferred from genome alignment using Mauve (35), and SNPs were of KPC-producing Klebsiella pneumoniae in China. J. Antimicrob. Che-
counted on a 1,000-nucleotide (nt) window based on the nucleotide po- mother. 66:307–312. http://dx.doi.org/10.1093/jac/dkq431.
sition on NJST258_1. 11. Pereira PS, de Araujo CF, Seki LM, Zahner V, Carvalho-Assef AP,
Nucleotide sequence accession numbers. Illumina short read data for Asensi MD. 2013. Update of the molecular epidemiology of KPC-2-
ST258 have been deposited in the Sequence Read Archive (SRA) database producing Klebsiella pneumoniae in Brazil: spread of clonal complex 11
under accession no. SRP036874 (7). The Illumina short read data for (ST11, ST437 and ST340). J. Antimicrob. Chemother. 68:312–316. http://
dx.doi.org/10.1093/jac/dks396.
Kp1832 have been deposited in the SRA database under accession no.
12. Chiu SK, Wu TL, Chuang YC, Lin JC, Fung CP, Lu PL, Wang JT, Wang
SRX512850. LS, Siu LK, Yeh KM. 2013. National surveillance study on carbapenem
non-susceptible Klebsiella pneumoniae in Taiwan: the emergence and
SUPPLEMENTAL MATERIAL rapid dissemination of KPC-2 carbapenemase. PLoS One 8:e69428. http://
Supplemental material for this article may be found at http://mbio.asm.org/ dx.doi.org/10.1371/journal.pone.0069428.
lookup/suppl/doi:10.1128/mBio.01355-14/-/DCSupplemental. 13. Breurec S, Guessennd N, Timinouni M, Le TA, Cao V, Ngandjio A,
Figure S1, EPS file, 25.9 MB. Randrianirina F, Thiberge JM, Kinana A, Dufougeray A, Perrier-Gros-
Figure S2, EPS file, 1.1 MB. Claude JD, Boisier P, Garin B, Brisse S. 2013. Klebsiella pneumoniae
resistant to third-generation cephalosporins in five African and two Viet-
namese major towns: multiclonal population structure with two major
ACKNOWLEDGMENTS international clonal groups, CG15 and CG258. Clin. Microbiol. Infect.
This work was supported in part by a grant (to B.N.K.) from the National 19:349 –355. http://dx.doi.org/10.1111/j.1469-0691.2012.03805.x.
Institutes of Health (1R01AI090155) and the Intramural Research Pro- 14. Cuzon G, Naas T, Truong H, Villegas MV, Wisell KT, Carmeli Y, Gales
gram of the National Institute of Allergy and Infectious Diseases, National AC, Venezia SN, Quinn JP, Nordmann P. 2010. Worldwide diversity of
Institutes of Health. Klebsiella pneumoniae that produce ␤-lactamase blaKPC-2 gene. Emerg.
B.N.K. discloses that he holds two patents that focus on using DNA Infect. Dis. 16:1349 –1356. http://dx.doi.org/10.3201/eid1609.091389.
15. Naas T, Cuzon G, Villegas MV, Lartigue MF, Quinn JP, Nordmann P.
sequencing to identify bacterial pathogens.
2008. Genetic structures at the origin of acquisition of the beta-lactamase
blaKPC gene. Antimicrob. Agents Chemother. 52:1257–1263. http://
REFERENCES dx.doi.org/10.1128/AAC.01451-07.
1. CDC. 2013. Antibiotic resistance threats in the United States, 2013. CDC, 16. Robinson DA, Monk AB, Cooper JE, Feil EJ, Enright MC. 2005. Evo-
Atlanta, GA. lutionary genetics of the accessory gene regulator (agr) locus in Staphylo-
2. Patel G, Bonomo RA. 2013. “Stormy waters ahead”: global emergence of coccus aureus. J. Bacteriol. 187:8312– 8321. http://dx.doi.org/10.1128/
carbapenemases. Front. Microbiol. 4:48. http://dx.doi.org/10.3389/ JB.187.24.8312-8321.2005.
fmicb.2013.00048. 17. Harris SR, Feil EJ, Holden MT, Quail MA, Nickerson EK, Chantratita
3. Munoz-Price LS, Poirel L, Bonomo RA, Schwaber MJ, Daikos GL, N, Gardete S, Tavares A, Day N, Lindsay JA, Edgeworth JD, de Len-
Cormican M, Cornaglia G, Garau J, Gniadkowski M, Hayden MK, castre H, Parkhill J, Peacock SJ, Bentley SD. 2010. Evolution of MRSA
Kumarasamy K, Livermore DM, Maya JJ, Nordmann P, Patel JB, during hospital transmission and intercontinental spread. Science 327:
Paterson DL, Pitout J, Villegas MV, Wang H, Woodford N, Quinn JP. 469 – 474. http://dx.doi.org/10.1126/science.1182395.
2013. Clinical epidemiology of the global expansion of Klebsiella pneu- 18. Bellais S, Six A, Fouet A, Longo M, Dmytruk N, Glaser P, Trieu-Cuot
moniae carbapenemases. Lancet Infect. Dis. 13:785–796. http:// P, Poyart C. 2012. Capsular switching in group B Streptococcus CC17
dx.doi.org/10.1016/S1473-3099(13)70190-7. hypervirulent clone: a future challenge for polysaccharide vaccine devel-
4. Tzouvelekis LS, Markogiannakis A, Psichogiou M, Tassios PT, Daikos opment. J. Infect. Dis. 206:1745–1752. http://dx.doi.org/10.1093/infdis/
GL. 2012. Carbapenemases in Klebsiella pneumoniae and other jis605.
Enterobacteriaceae: an evolving crisis of global dimensions. Clin. Micro- 19. Brochet M, Rusniok C, Couvé E, Dramsi S, Poyart C, Trieu-Cuot P,
biol. Rev. 25:682–707. http://dx.doi.org/10.1128/CMR.05035-11. Kunst F, Glaser P. 2008. Shaping a bacterial genome by large chromo-
5. Kitchel B, Rasheed JK, Patel JB, Srinivasan A, Navon-Venezia S, Car- somal replacements, the evolutionary history of Streptococcus agalactiae.
meli Y, Brolund A, Giske CG. 2009. Molecular epidemiology of KPC- Proc. Natl. Acad. Sci. U. S. A. 105:15961–15966. http://dx.doi.org/
producing Klebsiella pneumoniae isolates in the United States: clonal ex- 10.1073/pnas.0803654105.
pansion of multilocus sequence type 258. Antimicrob. Agents Chemother. 20. Wyres KL, Lambertsen LM, Croucher NJ, McGee L, von Gottberg A,
53:3365–3370. http://dx.doi.org/10.1128/AAC.00126-09. Liñares J, Jacobs MR, Kristinsson KG, Beall BW, Klugman KP, Parkhill
6. Schwaber MJ, Lev B, Israeli A, Solter E, Smollan G, Rubinovitch B, J, Hakenbeck R, Bentley SD, Brueggemann AB. 2013. Pneumococcal
Shalit I, Carmeli Y, Israel Carbapenem-Resistant Enterobacteriaceae capsular switching: a historical perspective. J. Infect. Dis. 207:439 – 449.
Working Group. 2011. Containment of a country-wide outbreak of http://dx.doi.org/10.1093/infdis/jis703.
carbapenem-resistant Klebsiella pneumoniae in Israeli hospitals via a na- 21. Adler A, Khabra E, Chmelnitsky I, Giakkoupi P, Vatopoulos A, Mathers
tionally implemented intervention. Clin. Infect. Dis. 52:848 – 855. http:// AJ, Yeh AJ, Sifri CD, De Angelis G, Tacconelli E, Villegas MV, Quinn
dx.doi.org/10.1093/cid/cir025. J, Carmeli Y. 2014. Development and validation of a multiplex PCR assay
7. DeLeo FR, Chen L, Porcella SF, Martens CA, Kobayashi SD, Porter AR, for identification of the epidemic ST-258/512 KPC-producing Klebsiella
Chavda KD, Jacobs MR, Mathema B, Olsen RJ, Bonomo RA, Musser pneumoniae clone. Diagn. Microbiol. Infect. Dis. 78:12–15. http://
JM, Kreiswirth BN. 2014. Molecular dissection of the evolution of dx.doi.org/10.1016/j.diagmicrobio.2013.10.003.
carbapenem-resistant multilocus sequence type 258 Klebsiella pneu- 22. Giltner CL, Nguyen Y, Burrows LL. 2012. Type IV pilin proteins: versa-
moniae. Proc. Natl. Acad. Sci. U. S. A. 111:4988 – 4993. http://dx.doi.org/ tile molecular modules. Microbiol. Mol. Biol. Rev. 76:740 –772. http://
10.1073/pnas.1321364111. dx.doi.org/10.1128/MMBR.00035-12.
8. Ramos PI, Picão RC, Almeida LG, Lima NC, Girardello R, Vivan AC, 23. Rao DN, Dryden DT, Bheemanaik S. 2014. Type III restriction-
Xavier DE, Barcellos FG, Pelisson M, Vespero EC, Médigue C, Vascon- modification enzymes: a historical perspective. Nucleic Acids Res. 42:
celos AT, Gales AC, Nicolás MF. 2014. Comparative analysis of the 45–55. http://dx.doi.org/10.1093/nar/gkt1373.
complete genome of KPC-2-producing Klebsiella pneumoniae Kp13 re- 24. Samuelsen Ø, Thilesen CM, Heggelund L, Vada AN, Kümmel A,

May/June 2014 Volume 5 Issue 3 e01355-14 ®

mbio.asm.org 7
Downloaded from mbio.asm.org on May 23, 2016 - Published by mbio.asm.org
Chen et al.

Sundsfjord A. 2011. Identification of NDM-1-producing Enterobacteri- moniae HS11286, a multidrug-resistant strain isolated from human spu-
aceae in Norway. J. Antimicrob. Chemother. 66:670 – 672. http:// tum. J. Bacteriol. 194:1841–1842. http://dx.doi.org/10.1128/JB.00043-12.
dx.doi.org/10.1093/jac/dkq483. 30. Leski T, Vora GJ, Taitt CR. 2012. Multidrug resistance determinants from
25. Lascols C, Peirano G, Hackel M, Laupland KB, Pitout JD. 2013. Sur- NDM-1-producing Klebsiella pneumoniae in the USA. Int. J. Antimicrob.
veillance and molecular epidemiology of Klebsiella pneumoniae isolates Agents 40:282–284. http://dx.doi.org/10.1016/j.ijantimicag.2012.05.019.
that produce carbapenemases: first report of OXA-48-like enzymes in 31. Chevreux B, Wetter T, Suhai S. 1999. Genome sequence assembly using
North America. Antimicrob. Agents Chemother. 57:130 –136. http:// trace signals and additional sequence information, p 45–56. In Computer
dx.doi.org/10.1128/AAC.01686-12. Science and Biology: Proceedings of the German Conference on Bioinfor-
26. Kristóf K, Tóth A, Damjanova I, Jánvári L, Konkoly-Thege M, Kocsis B, matics (GCB). GCB, Hannover, Germany.
Koncan R, Cornaglia G, Szego E, Nagy K, Szabó D. 2010. Identification 32. Zerbino DR, Birney E. 2008. Velvet: algorithms for de novo short read
of a blaVIM-4 gene in the internationally successful Klebsiella pneu- assembly using de Bruijn graphs. Genome Res. 18:821– 829. http://
moniae ST11 clone and in a Klebsiella oxytoca strain in Hungary. J. An- dx.doi.org/10.1101/gr.074492.107.
timicrob. Chemother. 65:1303–1305. http://dx.doi.org/10.1093/jac/ 33. Alikhan NF, Petty NK, Ben Zakour NL, Beatson SA. 2011. BLAST Ring
dkq133. Image Generator (Brig): simple prokaryote genome comparisons. BMC
27. Ma L, Lu PL, Siu LK, Hsieh MH. 2013. Molecular typing and resistance Genomics 12:402. http://dx.doi.org/10.1186/1471-2164-12-402.
mechanisms of imipenem-non-susceptible Klebsiella pneumoniae in 34. Zhou Y, Liang Y, Lynch KH, Dennis JJ, Wishart DS. 2011. PHAST: a fast
Taiwan: results from the Taiwan Surveillance of Antibiotic Resistance phage search tool. Nucleic Acids Res. 39:W347–W352. http://dx.doi.org/
(TSAR) study, 2002-2009. J. Med. Microbiol. 62:101–107. http:// 10.1093/nar/gkq1255.
dx.doi.org/10.1099/jmm.0.050492-0. 35. Darling AE, Mau B, Perna NT. 2010. progressiveMauve: multiple ge-
28. Voulgari E, Zarkotou O, Ranellou K, Karageorgopoulos DE, Vrioni G, nome alignment with gene gain, loss and rearrangement. PLoS One
Mamali V, Themeli-Digalaki K, Tsakris A. 2013. Outbreak of OXA-48 5:e11147. http://dx.doi.org/10.1371/journal.pone.0011147.
carbapenemase-producing Klebsiella pneumoniae in Greece involving an 36. Tamura K, Peterson D, Peterson N, Stecher G, Nei M, Kumar S. 2011.
ST11 clone. J. Antimicrob. Chemother. 68:84 – 88. http://dx.doi.org/ MEGA5: molecular evolutionary genetics analysis using maximum
10.1093/jac/dks356. likelihood, evolutionary distance, and maximum parsimony methods.
29. Liu P, Li P, Jiang X, Bi D, Xie Y, Tai C, Deng Z, Rajakumar K, Ou HY. Mol. Biol. Evol. 28:2731–2739. http://dx.doi.org/10.1093/molbev/
2012. Complete genome sequence of Klebsiella pneumoniae subsp. pneu- msr121.

8 ®

mbio.asm.org May/June 2014 Volume 5 Issue 3 e01355-14


Abundance of Antibiotic Resistance Genes in Bacteriophage following
Soil Fertilization with Dairy Manure or Municipal Biosolids, and
Evidence for Potential Transduction
Joseph Ross,a Edward Toppa,b
Agriculture and Agri-Food Canada, London, ON, Canadaa; Department of Biology, University of Western Ontario, London, ON, Canadab

Animal manures and municipal biosolids recycled onto crop production land carry antibiotic-resistant bacteria that can influ-
ence the antibiotic resistome of agricultural soils, but little is known about the contribution of bacteriophage to the dissemina-
tion of antibiotic resistance genes (ARGs) in this context. In this work, we quantified a set of ARGs in the bacterial and bacterio-
phage fractions of agricultural soil by quantitative PCR. All tested ARGs were present in both the bacterial and phage fractions.
We demonstrate that fertilization of soil with dairy manure or human biosolids increases ARG abundance in the bacterial frac-
tion but not the bacteriophage fraction and further show that pretreatment of dairy manure can impact ARG abundance in the
bacterial fraction. Finally, we show that purified bacteriophage can confer increased antibiotic resistance to soil bacteria when
combined with selective pressure. The results indicate that soilborne bacteriophage represents a substantial reservoir of antibi-
otic resistance and that bacteriophage could play a significant role in the horizontal transfer of resistance genes in the context of
an agricultural soil microbiome. Overall, our work reinforces the advisability of composting or digesting fecal material prior to
field application and suggests that application of some antibiotics at subclinical concentrations can promote bacteriophage-me-
diated horizontal transfer of ARGs in agricultural soil microbiomes.

O veruse of antibiotics has contributed to the spread of antibi-


otic resistance due to the release of antibiotics, antibiotic-
resistant bacteria, and antibiotic resistance genes (ARGs) into the
biosolids on the abundance and distribution of ARGs in bacteria
and bacteriophage was determined. In the case of dairy manure,
the effect of various preapplication treatments on the composition
environment (1). This phenomenon is mediated by horizontal of ARGs in the bacterial and bacteriophage fractions was deter-
gene transfer (HGT) of mobile genetic elements—such as plasmids, mined. Finally, we tested whether bacteriophage enriched from
transposons, and integrons— between bacterial cells through conju- biosolids conferred increased antibiotic resistance to soilborne
gation and viral transduction (2–4). bacteria when combined with selective pressure.
Transduction has been shown to occur in environmental matri-
ces, including freshwater and wastewater; moreover, bacterial 16S MATERIALS AND METHODS
ribosomal RNA sequences have been observed in the viral fraction of Field operations and soil sampling. The soil samples used in the experi-
wastewater, confirming the ability of bacteriophage to carry bacterial ments described here were obtained from field experiments undertaken dur-
genes (5, 6). In fact, while much attention has been paid to conjuga- ing the 2014 growing season on the Agriculture and Agri-Food Canada re-
search farm in London, Ontario, Canada (42.984°N, 81.248°W). The field
tion, more recent work has additionally implicated bacteriophage as a
installations and methods were described in detail by Marti et al. (17). Briefly,
major vehicle for horizontal gene transfer and recombination (7–10). the soil is a silt loam (gray-brown luvisol) with the following properties: a pH
Using high-throughput sequencing of murine fecal phage popu- of 7.5; a cation exchange capacity of 13.2; a sand, silt, and clay composition of
lations, Modi et al. (11) demonstrated that antibiotic treatment leads 18%, 67%, and 15%, respectively; and an organic matter content of 3.4%.
to the enrichment of genes conferring resistance to the administered Manure for application in the spring of 2014 was obtained from dairy farms in
drug as well as to unrelated antibiotics in the phage genome. Further- the London area, and biosolids that were aerobically digested and dewatered
more, bacteriophage from drug-treated mice provided cultured naive were obtained from the municipality of Tilsonburg, Ontario, Canada. The
microbiota with increased levels of resistance to the corresponding dairy manures were variously untreated (raw), anaerobically digested, me-
drug. Overall, this work concluded that antibiotic residues potentiate chanically dewatered, or composted. The dewatered and composted manures
the transduction-mediated dissemination of ARGs.
Antibiotic resistance genes have been found in the bacterio-
Received 22 July 2015 Accepted 1 September 2015
phage DNA fraction of various environmental matrices, such as
Accepted manuscript posted online 4 September 2015
activated sludge (12), urban sewage and river water (13), and
Citation Ross J, Topp E. 2015. Abundance of antibiotic resistance genes in
wastewater effluents from hospitals and wastewater treatment bacteriophage following soil fertilization with dairy manure or municipal
plants (14). The aforementioned studies indicate that bacterio- biosolids, and evidence for potential transduction. Appl Environ Microbiol
phage represents a reservoir of ARGs across a broad selection of 81:7905–7913. doi:10.1128/AEM.02363-15.
environments. However, it remains to be determined if bacterio- Editor: D. W. Schaffner
phage has such a role in agricultural soils. Address correspondence to Edward Topp, ed.topp@agr.gc.ca.
In the present study, a set of ARGs previously detected in ma- Supplemental material for this article may be found at http://dx.doi.org/10.1128
nured soils was quantified in bacteria and in bacteriophage recov- /AEM.02363-15.
ered from agricultural soil using quantitative PCR (qPCR) (15–17). Copyright © 2015, American Society for Microbiology. All Rights Reserved.
The impact of soil amendment either with dairy manure or with

November 2015 Volume 81 Number 22 Applied and Environmental Microbiology aem.asm.org 7905
Ross and Topp

were applied at a rate of 5 dry metric tons/ha, and the raw and the digested the reaction mixtures were composed of 1⫻ Green GoTaq Flexi buffer; 1.5
manures were applied at a rate of 80,000 liters/ha. mM MgCl2; 0.2 mM (each) dATP, dGTP, dCTP, and dTTP; 0.4 ␮M each
Soil cores (2 cm wide, 15 cm deep) were taken immediately after ma- primer; 1 U of GoTaq DNA polymerase; and deionized water to 25 ␮l. The
nure or biosolid application, as well as at 7 and 30 days postapplication reactions were carried out at the following stages of the extraction proce-
(days 0, 7, and 30, respectively), in order to assess any temporal impact on dure: for bacterial DNA, after the full procedure, including ethanol pre-
ARG composition. Six cores were sampled from each of three replicated cipitation; for bacteriophage particles, after Turbo DNase treatment but
plots using a T sampler rinsed with 70% ethanol between samplings. before proteinase K lysis; and for bacteriophage DNA, after the full pro-
Cores were bulked into a labeled Ziploc bag, mixed by hand until they cedure, including ethanol precipitation. The ideal result was a positive
were homogeneous, and transported to the laboratory in a cooler with signal from the bacterial DNA preparations and a negative result from
cool packs. Thus, three independent soil samples (each representing the bacteriophage particle preparations, indicating the absence of bacterial
average for 6 cores from a given plot) from control, dairy manure, and DNA outside the bacteriophage particles. Thus, any bacterial DNA de-
biosolids treatments were analyzed at each sampling time. tected in the enriched bacteriophage DNA fraction must be due to a legit-
Extraction of bacterial and phage DNA fractions from soil. Twenty- imate uptake of that DNA into the phage particle during the viral replica-
five-gram portions of soil were mixed with 25 ml of sterile SM buffer (100 tion cycle. PCR products were analyzed by agarose gel electrophoresis; a
mM NaCl, 8 mM MgSO4 · 7H2O, 50 mM Tris-HCl, pH 7.5) in a 50-ml representative gel is shown in Fig. S1 in the supplemental material. In all
Falcon tube and thoroughly mixed by vortexing for 1 h. The resulting cases, no bacterial DNA (above the levels seen for a no-template negative
slurry was centrifuged at 2,500 ⫻ g for 4 min to pellet the soil, and the control) was detected for either intact phage particles or purified phage
supernatant was passed through a 0.22-␮m-pore-size Durapore mem- DNA. To ensure that the lack of amplification from bacteriophage parti-
brane filter (Millipore) under vacuum. The filter was rinsed with 10 ml of cles or bacteriophage DNA was not simply due to the presence of PCR
SM buffer. Bacteria were retained on the filter, whereas bacteriophage inhibitors, these reactions were also performed in the presence of the same
passed through and was in the filtrate. DNA used for the positive control, with positive results being obtained
The membrane was transferred to a 15-ml Falcon tube and incubated (see Fig. S1 in the supplemental material).
with 1 ml GITC (5 M guanidine isothiocyanate, 100 mM EDTA, 0.5% Quantification of gene target copies. The primers and probes (Sigma-
N-lauroylsarcosine) buffer for 1 h at 75°C. DNA was extracted using a Aldrich, Toronto, ON, Canada) used in the present study are summarized
modification of the Qiagen DNeasy blood and tissue kit procedure. in Table 1. The resistance genes quantified were strA, strB, sul1, aadA, and
Briefly, 0.5 ml of buffer AL was added to each tube before vortexing for 1 blaOXA-20, which encode resistance to streptomycin (strA and strB), sulfa-
min, adding 0.5 ml 95% ethanol, vortexing again, and loading the con- methazine (sul1), aminoglycosides (aadA), and ␤-lactams (blaOXA-20).
tents of the tubes onto the provided spin columns in 700-␮l increments. Note that the amount of blaOXA-20 in dairy manure-amended soils was
The columns were then washed once with 500 ␮l of buffer AW1 and twice consistently below the limit of detection, and therefore, blaOXA-20 is not
with 700 ␮l of buffer AW2. The columns were centrifuged for 2 min at discussed. As a control for total bacterial DNA abundance among the
maximum speed to remove residual wash buffer, before they were eluted various samples, rrnS (encoding the 16S rRNA) was also quantified. PCR
twice with 50 ␮l buffer AE. The resulting 100 ␮l of DNA, now enriched for amplification was performed using a Bio-Rad CFX96 real-time PCR in-
bacterial DNA, was further purified and concentrated by ethanol precip- strument with Bio-Rad CFX Manager software, version 3.0. The reactions
itation, using a 1:10 volume of 3 M sodium acetate, 3 volumes of 95% were performed with the Brilliant II quantitative PCR (qPCR) master mix
ethanol, and RNA-grade glycogen (Life Technologies) as a coprecipitate. (Agilent, Toronto, ON, Canada) for the TaqMan PCR and the Brilliant II
The pellet was resuspended in 12 ␮l of 1⫻ TE (10 mM Tris-HCl, pH 7.5, SYBR green qPCR master mix (Agilent) for the SYBR green PCR. Two
1 mM EDTA), quantified with a NanoDrop ND1000 microspectropho- microliters of template DNA (corresponding to 2 ng of either bacterio-
tometer (NanoDrop Technologies, Wilmington, DE), and diluted to a phage or bacterial DNA) was added, and deionized water was used to
final concentration of 1 ng/␮l using deionized water. The DNA extract reach a final volume of 25 ␮l. Negative controls without template DNA
was aliquoted and stored at ⫺20°C. were run in triplicate. Each reaction was run in triplicate with the follow-
The soil filtrate was filtered once more using a 0.22-␮m-pore-size ing cycle conditions: 1 cycle at 95°C for 10 min followed by 40 cycles of
Sterivex syringe filter. The phage in the filtrate was concentrated using 95°C for 15 s, the annealing temperature indicated in Table 1 for 35 s, and
Amicon Ultra-15 filtration devices (30-kDa-molecular-mass cutoff) per 72°C for 1 min. For the SYBR green assay, a melting curve step was added
the manufacturer’s instructions. The resulting 400 to 600 ␮l was further in order to check the purity of the PCR product. This step consisted of a ramp
enriched for bacteriophage by ultracentrifugation on CsCl density gradi- of the temperature from 65 to 95°C at an increment of 0.5°C and a hold for 5
ents of 1.7, 1.5, and 1.35 g/ml as described in reference 18. The resulting 1 s for each step. The plasmids used to generate standard curves were created as
ml of fluid containing enriched phage was desalted using Amicon Ultra- described by Marti et al. (16). Standard curves were prepared with a 10-fold
0.5 filtration devices (30-kDa-molecular-mass cutoff) per the manufac- serial dilution of the known concentration of the plasmid solution for each
turer’s instructions. The 30 ␮l of concentrate was adjusted to 1 ml with marker, in order to have final concentrations of the plasmid ranging from 107
sterile SM buffer and stored at 4°C. To ensure the removal of nonphage to 10° copies per microliter. The identities of the quantified gene targets were
DNA prior to lysis of the phage particles, 0.5 ml of enriched bacteriophage ensured on the basis of hybridization when using TaqMan chemistry or melt-
was treated with 10 U of Turbo DNase (Life Technologies) for 1 h at 37°C ing behavior when using SYBR green. Gene abundance data are presented as
and then for 5 min at 65°C. To isolate bacteriophage DNA, 0.4 mg of the number of gene copies (GC) per nanogram of bacterial or phage DNA
proteinase K (final concentration, 0.7 mg/ml) was incubated with the included in the reaction mixture.
DNase-treated bacteriophage preparation for 1 h at 37°C to digest the Transduction of soil bacteria by biosolids-derived phage. Total bac-
capsid proteins. The resulting lysate was extracted twice with phenol- teriophage was extracted and enriched from biosolids (three independent
chloroform-isoamyl alcohol, and the DNA was further purified and con- replicates) essentially as described above for soil, except that the initial
centrated by ethanol precipitation as described above. centrifugation was increased to 15 min at 10,000 ⫻ g and the filtration
Each time that the above-described procedures were carried out, a steps were repeated two to three times to ensure the adequate removal of
series of conventional PCRs was performed in order to assess the quality of bacteria. Phage was stored at 4°C in SM buffer. In order to ensure that the
bacteriophage isolation. The presence of bacterial rrnS DNA (encoding aforementioned phage purification procedure, while stringent, yielded an
the 16S rRNA) was assessed using the primers described in Table 1. PCRs adequate number of infection-competent phage particles, the titer of co-
were carried out with the following cycle conditions: 1 cycle at 95°C for 10 liphage within the resulting bacteriophage enrichments was estimated
min followed by 30 cycles of 95°C for 15 s, 55°C for 15 s, and 72°C for 1 using the single-agar-layer method essentially as described in reference 23.
min. GoTaq Flexi reagents (Promega) used in the reaction mixtures, and Escherichia coli K-12 (grown in tryptone-yeast extract-glucose broth) was

7906 aem.asm.org Applied and Environmental Microbiology November 2015 Volume 81 Number 22
Antibiotic Resistance in Soil Phage

TABLE 1 Primers and probes used for conventional or quantitative PCR in this study
Primer specificity and Product Annealing Final primer
primer namea Sequence (5= to 3=)b size (bp) temp (°C) concn (nM) Target Reference
Universal bacteria
(conventional
PCR)
GM5-F CCTACGGGAGGCAGCAG 550 55 400 rrnS gene 19
907-R CCGTCAATTCCTTTGAGTTT 400

Universal bacteria
BACT1369F CGGTGAATACGTTCYCGG 123 59 300 rrnS gene 20
PROK1492R GGWTACCTTGTTACGACTT 300
TM1389F HEX-CTTGTACACACCGCCCGTC-BHQI 300

strA
strA-F TATGGTTGTTTGCCATGGTG 149 62 400 Streptomycin 15
strA-R TTCTCTTCGGCGTTAGCAAT 400 Phosphotransferase A

strB
strB-F ATCGCTTTGCAGCTTTGTTT 143 61 300 Streptomycin 21
strB-R ATGATGCAGATCGCCATGTA 300 Phosphotransferase B
strB-P HEX-ATGCCTCGGAACTGCGT-BHQI 200

sul1
sul1-F GACTGCAGGCTGGTGGTTAT 105 64 200 Sulfamethazine 16
resistance gene 1
sul1-R GAAGAACCGCACAATCTCGT 200

aadA
aadA-F CAGCGCAATGACATTCTTGC 293 63 200 Aminoglycoside 21
adenyltransferase A
aadA-R GTCGGCAGCGACA(C/T)CCTTCG 200
aadA-P HEX-TGGTAGGTCCAGCGGCGGAG-BHQI 300

blaOXA-20
blaOXA-20-F TGATGATTGTCGAAGCCAAA 100 60 400 Beta-lactamase, class D 22
(oxacillinase)
blaOXA-20-R GCCTGTAGGCCACTCTACCC 400
a
F, forward primer; R, reverse primer; P, probe.
b
HEX, 2=,4=,5=,7=-tetrachloro-6-carboxy-4,7-dichlorofluorescein succinimidyl ester; BHQI, black hole quencher 1.

used as the host, and tryptone-yeast extract-glucose agar was the medium. two 15-ml Falcon tubes (5 ml per tube). One tube served as a no-phage
Control plates that received the enriched bacteriophage preparation but control and received 500 ␮l of SM buffer. The other tube was amended
no strain K-12 bacteria had no visible colonies, corroborating the conclu- with 500 ␮l of biosolids-derived phage; this was performed for three in-
sion from the rrnS-specific PCR that the phage preparations were free of dependent biological isolates of phage and soil-derived supernatant. After
bacterial contamination. The average titer of coliphage was determined to 1 h of incubation at 30°C, ampicillin, cefoxitin, or sulfamethazine was
be about 300 PFU/ml in our enrichments. For comparison, Colomer- added to various final concentrations (see Table 2). The resulting trans-
Lluch et al. (13) reported coliphage titers of about 104 PFU/ml and 102 duction reaction mixtures were incubated for 3 days at 30°C, serially di-
PFU/ml in urban sewage and river water, respectively; Ashelford et al. (24) luted, and plated on either selective or control Chromocult agar. Thus, we
reported about 107 phage per gram of soil associated with plant roots in chose to focus on resistant versus total coliform bacteria as a subset of the
sugar beet fields. While our coliphage titer was low by comparison to the total population of potential bacterial transductants generated in this as-
titers in the aforementioned studies, it should be noted that these were say. Control plates lacked antibiotics (in order to count the total number
assessments of the bacteriophage titers in environmental matrices that of coliforms), while selective plates contained ampicillin (32 ␮g/ml), cefoxitin
had not undergone phage purification procedures and that the titer of (32 ␮g/ml), or sulfamethazine (512 ␮g/ml) in order to count the number of
K-12-infecting coliphage reported here is likely a vast underestimate of resistant coliform bacteria. Drug concentrations were based on breakpoints
the total number of infection-competent bacteriophage recovered. (i.e., MICs) reported by the Canadian Integrated Program for Antimicrobial
We chose to assess the impact of biosolids-derived phage on soilborne Resistance Surveillance (25). The frequency of resistant coliforms was calcu-
microbiota without the complication of soil particles, which might adsorb lated by dividing the resistant coliform count (in numbers of CFU per milli-
to antibiotics or bacteriophage and thus interfere with the effective free liter) by the total coliform count (also in numbers of CFU per milliliter)
concentration of drugs or phage particles. Accordingly, total bacteria were counted in the absence of antibiotics. Phage-only controls were also included
extracted from soil in an aqueous context as follows: 20 g of soil was mixed and yielded no bacterial growth. In all cases, the Chromocult plates were
with 20 ml of SM buffer in a 50-ml Falcon tube by vortexing for 1 h. The incubated at 37°C overnight. Note that these 3 antibiotics were chosen on the
resulting slurry was centrifuged at 2,000 ⫻ g for 2 min to remove most soil basis of an initial qualitative screen of 10 antibiotics (see Fig. S2 in the supple-
and large particulates, and the supernatant was removed and placed into mental material). These 10 antibiotics were chosen to represent several major

November 2015 Volume 81 Number 22 Applied and Environmental Microbiology aem.asm.org 7907
Ross and Topp

FIG 1 Abundance of gene targets in the bacterial and bacteriophage DNA fractions of soil amended with dairy manure. Bar graphs represent the number of gene
copies per nanogram of template DNA, as measured by qPCR. The gene targets quantified are indicated above the corresponding graphs. As indicated at the
bottom, soil either did not receive manure (control [Con]) or was amended with raw (Raw) dairy manure or manure that had been digested (Dig), dewatered
(DW), or composted (Comp) prior to application. Soil from these plots was sampled immediately after application, as well as 7 and 30 days later (days 0, 7, and
30, respectively). A no-template control (⫺) was also included in the qPCRs. Data are presented as the mean and standard deviation from three biological
replicates. *, a statistically significant difference between the bacterial and corresponding phage DNA fractions. A statistically significant difference between a
given manure treatment and the corresponding control not receiving manure is indicated by the letter a or b for the bacterial or phage fraction, respectively.

classes (e.g., penicillins, aminoglycosides, macrolides, cephalosporins, car- vary with manure application (Fig. 1, top). Soil carried about 105
bapenems, quinolones, sulfonamides). gene copies (GC) per ng of template DNA at day 0 and about 106
Calculations and statistics. Each condition was analyzed in triplicate. GC/ng DNA 7 and 30 days following manure application. The
In the qPCR experiments, three technical replicates were performed for increase in rrnS copy numbers at days 7 and 30 relative to the
each of three independent biological isolates for each sample. In the trans- numbers at day 0 was statistically significant for most of the cor-
duction experiment, we report the averages from three independent bio-
logical replicates. Statistically significant treatment effects were deter-
responding treatments, namely, the control, raw, digested, dewa-
mined using an unpaired t test without assuming equal variance. Data tered, and composted treatments for day 0 versus day 7 and the
were analyzed using SigmaPlot software, version 12.5 (Systat Software control, digested, and composted treatments for day 0 versus day
Inc.). The significance level was set at a P value of 0.05. 30. For simplicity, the results of these statistical analyses are not
indicated in Fig. 1. Overall, there was a time-dependent increase in
RESULTS bacterial abundance following manure application. The absence
Impact of manure application on abundance and distribution of any effect of manure preapplication treatment on rrnS abun-
of antibiotic resistance genes in bacteriophage and bacterial dance indicates that the overall abundance of bacteria was unaf-
fractions. The abundance of rrnS in the bacterial fraction did not fected by these treatments. Thus, any changes in ARG abundance

7908 aem.asm.org Applied and Environmental Microbiology November 2015 Volume 81 Number 22
Antibiotic Resistance in Soil Phage

seen between manure treatments would not simply reflect a gross sul1 were more abundant in the bacterial fraction of treated soil
variation in the overall bacterial abundance. than it that of control soil (Fig. 2). The abundance of these gene
As expected, the rrnS gene target was far less abundant in the targets declined thereafter through day 30. In contrast, aadA and
bacteriophage DNA fraction for all treatments, where it was pres- blaOXA-20 were not enriched by biosolids application. Further-
ent at a level of about 102 GC/ng DNA (Fig. 1, top). Importantly, more, other than aadA on day 30, both aadA and blaOXA-20 other-
this level was not significantly different from the background level wise remained at comparable levels in the treated and control soils
of rrnS measured in a no-template control. This means that any through the period of investigation.
ARGs observed in the phage fraction do not simply reflect the Every gene target was detected in the bacteriophage fraction in
rampant acquisition of bacterial genes overall, since the rrnS gene control soils, with the exception of strA, which was undetected in
is very rarely acquired by bacteriophage in our soil samples. As both the bacterial and bacteriophage fractions (Fig. 2). Only sul1
with the bacterial fraction, the rrnS copy number in the bacterio- was significantly less abundant in the bacteriophage fraction than
phage DNA fraction was consistent among all treatments, indicat- in the bacterial fraction for untreated soil. ARG abundance in the
ing little or no impact of manure application or pretreatment of bacteriophage fraction did not respond significantly to biosolids
manure on the overall bacterial DNA abundance in bacterio- treatment or time for most samples, with the exception of sul1
phage. abundance. On day 0, the abundance of this gene target increased
A no-template control was also evaluated for each of the ARG slightly but significantly in treated soil relative to that in control
primer/probe sets, and no gene targets were detected, indicating soil (Fig. 2). As was observed with dairy manure, the phage frac-
low background levels for each of the ARGs. Within the bacterial tion appears to harbor a constant reservoir of ARGs at the back-
fraction, each of the detected ARGs was significantly more abun- ground abundance, and the abundance in the phage fraction had
dant in soil receiving raw manure than in control soil not receiving little or no response to amendment with biosolids.
manure (Fig. 1). For instance, the level of sul1 increased from Taken together, these results suggest that soilborne bacteriophage
about 101 GC/ng DNA in control soil to about 105 GC/ng DNA in represents a reservoir of antibiotic resistance genes. Amendment of
soil amended with raw manure. Furthermore, ARG abundance soil with dairy manure or biosolids had no significant effect on the
was significantly higher for soils amended with digested and de- abundance of gene targets in the bacteriophage fraction but in-
watered manure than for control soil, indicating that these treat- creased the abundance in the bacterial fraction.
ments are ineffective at mediating the dissemination of ARGs by Potential for transduction. Aqueous suspensions of soil bac-
manure application. In contrast, composting of manure prior to teria were incubated for 3 days with or without bacteriophage
field application did not increase the abundance of ARG targets enriched from biosolids and with or without supplementation
above that in soils not receiving manure, indicating the efficacy of with antibiotics (Table 2). Note that we chose to quantify coliform
this practice. The ARG abundance in the bacterial fraction tran- bacteria as a representative subset of bacteria within the total soil-
siently increased on day 7 relative to that on day 0 and then de- borne microbiome. In the absence of added antibiotics, there was
creased by day 30. This suggests that there may have been an initial no effect of bacteriophage supplementation on the abundance of
proliferation of ARG-harboring bacteria in the days following ma- viable antibiotic-resistant coliform bacteria (Table 2). However,
nure application, followed by their decline by day 30. after 3 days in the presence of 1/10 the breakpoint concentration
In soils not receiving manure, each ARG quantified in the bac- of cefoxitin, the abundance of cefoxitin-resistant coliforms was
terial fraction was also detected in the phage fraction (Fig. 1). 3.7-fold higher in the presence than in the absence of bacterio-
Moreover, ARG abundance was not significantly lower in the phage (Table 2). Likewise, phage conferred a 6.3-fold increase in
phage fraction than in the corresponding bacterial fraction in soils the abundance of sulfamethazine-resistant coliforms when incu-
not receiving manure, with the exception of the abundances of bated in the presence of 1/100 of the sulfamethazine breakpoint
strA (not detected on day 7) and strB (not detected on day 7 or 30). concentration and a 7.1-fold increase in the presence of 1/10 of its
Strikingly, aadA was significantly more abundant in the phage breakpoint concentration (Table 2). Phage appeared to confer a
fraction than in the bacterial fraction on days 7 and 30 (Fig. 1, slight decrease (less than 2-fold) in the abundance of cefoxitin-
bottom). In stark contrast to the findings for the bacterial fraction, resistant coliforms when incubated in the presence of 1/100 of the
ARG abundance in the phage fraction did not generally respond cefoxitin breakpoint concentration. On the other hand, the abun-
significantly to manure treatment or time (Fig. 1). Instead, the dance of ampicillin-resistant coliforms did not vary with any
phage fraction appeared to harbor a constant reservoir of ARGs at treatment. Taken together, these results suggest that bacterio-
a background abundance roughly matching that seen in the bac- phage from biosolids increased the abundance of coliform bacte-
terial fraction in the absence of manure. This abundance did not ria resistant to sulfamethazine or cefoxitin in the presence but not
respond to amendment with manure, regardless of preapplication the absence of each antibiotic.
treatment.
Impact of biosolids application on abundance and distribu- DISCUSSION
tion of antibiotic resistance genes in bacteriophage and bacte- Distribution of antibiotic resistance genes in bacterial and bac-
rial fractions. The abundance of rrnS was between 104 and 105 teriophage fractions of agricultural soils. Several studies have
GC/ng DNA in untreated soil and did not increase significantly detected ARGs in the bacteriophage metagenome (or phageome)
following application of aerobically digested biosolids (Fig. 2). In of a wide variety of environmental matrices, including activated
fact, there was a significant decrease on days 0 and 7 relative to that sludge (9, 12), urban sewage and river water (13), and various
in untreated soil. Thus, any increase in ARG abundance in treated wastewater effluents from hospitals and wastewater treatment
versus control soil would not simply reflect an increase in bacterial plants (14, 26). When specific ARGs are detected by real-time PCR
DNA abundance. in the bacteriophage populations of the above-described environ-
Following biosolids application, the gene targets strA, strB, and ments, their levels are only about 10-fold lower than those in the

November 2015 Volume 81 Number 22 Applied and Environmental Microbiology aem.asm.org 7909
Ross and Topp

FIG 2 Abundance of gene targets in the bacterial and bacteriophage DNA fractions of soil amended with aerobically digested biosolids. Bar graphs represent the
number of gene copies per nanogram of template DNA, as measured by qPCR. The gene targets quantified are indicated above the corresponding graphs. As
indicated at the bottom of each graph, soil was untreated (control [Con]; sampled immediately before application) or received biosolids and was sampled
immediately after application (day 0) or at 7 and 30 days postapplication. A no-template control (⫺) was also included in the qPCRs. Data are presented as the
mean and standard deviation from three replicates. *, a statistically significant difference between the corresponding bacterial and phage DNA fractions. A
statistically significant difference between a given biosolids-treated time point and the corresponding untreated control is indicated by the letter a or b for the
bacterial or phage fraction, respectively.

corresponding bacterial fractions, on average (reviewed in refer- given that composting of the manure attenuated this enrichment
ence 2). The present study further demonstrates that the phageome of effect.
agricultural soil harbors ARGs. The ARGs tend to be present in In contrast to the responsiveness of the ARG target abun-
similar numbers in the corresponding bacterial and phage DNA dance in the bacterial fraction, ARG levels were virtually iden-
fractions from agricultural soils, at least in the absence of manure tical in the phageome, regardless of treatment or the time of
or biosolids application. ARG copy abundance in the bacterial sampling. It is unclear why ARG levels in the phageome did not
fraction rose sharply in response to dairy manure application and, respond to manure application or pretreatment in parallel to
in the case of strA, strB, and sul1, also rose in response to biosolids the bacteriome.
application. In the case of manure, some preapplication treatment Here, we used rrnS (the 16S rRNA gene) as an indicator of
options, specifically, composting, attenuated the enrichment ef- overall bacterial abundance; rrnS is also useful as a representative
fect. As measured by rrnS gene target copy abundance, there was gene for measurement of overall bacterial gene acquisition by bac-
no stimulation of total bacterial populations with the addition of teriophage (6). Since rrnS is ubiquitous in bacterial species, it
manure, regardless of manure treatment. In contrast, the abun- should represent a baseline level of bacterial gene acquisition by
dance of ARG target copies did respond to manure application. It phage. Notably, rrnS levels in the phage fraction of all soils sam-
is likely that the ARGs originated from bacteria that were intro- pled in this study were lower than those in the corresponding
duced by application of dairy manure and biosolids, especially bacterial fraction by several orders of magnitude (indeed, in most

7910 aem.asm.org Applied and Environmental Microbiology November 2015 Volume 81 Number 22
Antibiotic Resistance in Soil Phage

TABLE 2 Abundance of ampicillin-, cefoxitin-, and sulfamethazine-resistant coliforms recovered from soil suspensions following 3 days of
incubation in presence or absence of bacteriophage enriched from biosolidsa
Drug-resistant coliform count/total coliform count
Drug concnb during incubation Bacteriophage supplementation Ampicillin Cefoxitin Sulfamethazine
0 ⫺ 0.000337 ⫾ 0.000052 0.00724 ⫾ 0.00917 0.245 ⫾ 0.145
⫹ 0.00101 ⫾ 0.00099 0.00335 ⫾ 0.00273 0.129 ⫾ 0.037

1:100 ⫺ 0.00143 ⫾ 0.00142 0.00315 ⫾ 0.00044 0.00326 ⫾ 0.00291


⫹ 0.00252 ⫾ 0.01480 0.00168 ⫾ 0.00067* 0.0207 ⫾ 0.0132*

1:10 ⫺ 0.411 ⫾ 0.323 0.0512 ⫾ 0.0239 0.0342 ⫾ 0.0459


⫹ 0.116 ⫾ 0.031 0.192 ⫾ 0.060* 0.243 ⫾ 0.098*
a
Suspensions were incubated without the addition of antibiotics or with one of the antibiotics at either a 1:100 or a 1:10 clinical breakpoint concentration. Data represent the
mean ⫾ standard deviation (n ⫽ 3) of the count of resistant coliforms (in numbers of CFU per milliliter) normalized to the total coliform count (in numbers of CFU per milliliter).
*, statistically significant differences between phage treatment and no phage treatment.
b
1:100 and 1:10, final concentrations 100-fold and 10-fold, respectively, below the breakpoint concentrations of the indicated antibiotics (ampicillin, 32 ␮g/ml; cefoxitin, 32 ␮g/ml;
sulfamethazine, 512 ␮g/ml).

cases, rrnS levels in the phage fraction were not even above the doubtedly a widespread phenomenon in many environments. For
background rrnS levels, defined in control qPCRs lacking a tem- instance, retail chicken meat carries a number of phage capable of
plate). This suggests that bacterial gene uptake by phage is a rela- transferring antimicrobial resistance; of 243 phage randomly iso-
tively rare event, in agreement with previous estimates of about lated from chicken meat, about 25% was able to transduce into E.
one transduction event per every 108 phage infections (27). coli resistance to 1 or more of the 35 antimicrobials tested (29).
Potential transduction of soil coliform bacteria by biosolids- Based on factors like phage versus bacterial abundance, the num-
derived phage. We chose to focus on coliform bacteria as a subset ber of phage in a transduction-competent state, and the physical
of the total population of potential bacterial transductants gener- conditions of various environments, Muniesa et al. (8) concluded
ated in our experimental setup. This had the advantage of allowing that phage-mediated horizontal transfer between intestinal bacte-
us to quantify a relatively homogeneous set of bacterial species; the ria or between intestinal and indigenous bacteria in extraintestinal
counting of total viable bacteria from a complex matrix like soil is environments was probable. Moreover, bacteriophage isolated
cumbersome, and we would have no idea if increases in the pop- from antibiotic-treated mouse feces can confer antibiotic resis-
ulation of viable bacteria were due to the transduction of the var- tance to cultured murine intestinal microbiota (11). In-feed anti-
ious species or due to some shift in the complex dynamics of biotics induce prophage in swine fecal microbiomes (30), and
interspecies competition due to the perturbations of both antibi- horizontal gene transfer (including transduction) is subject to se-
otic selection and increased phage infective burden. By focusing lective pressure (31). For instance, a study examining the inci-
on coliforms, we thus took a more reductionist approach. A major dence of HGT of ARGs in thousands of microbial genomes found
drawback is that we likely grossly underestimated the true fre- that the genes involved in HGT events are 25-fold more likely to
quencies of transduction in agricultural soil, as well as the true become fixed in human-associated bacteria than in diverse envi-
extent of the influence of subclinical selective pressure. Neverthe- ronmental isolates, due to selective pressure (32). Combined with
less, our results serve as a proof of concept that subclinical con- our observation of probable transduction only in the presence of
centrations of some antibiotics can potentiate the phage-mediated selective pressure, it is tempting to speculate that transduction is
horizontal transfer of resistance genes into potential human generally potentiated by antibiotic selection.
pathogens in the context of an agricultural soil microbiome. It is unclear why ampicillin selection did not confer the trans-
We assume that the enrichment of antibiotic-resistant coli- duction of ampicillin resistance and why cefoxitin selection (at
forms in the presence of bacteriophage is due to transduction, 1/100 of the breakpoint concentration) caused a slight but signif-
although we use this term broadly, as the mechanism of gene icant decrease in cefoxitin resistance in the presence versus ab-
transfer and whether it is generalized or specialized (4) have not sence of phage. In the case of the latter, the decrease was quite
been proven. A few lines of evidence support the hypothesis that small (less than 2-fold), and a larger increase in resistance (nearly
transduction was at play. First, the effect occurred specifically 4-fold) was conferred by phage when cefoxitin selection was ap-
when enriched bacteriophage was incubated with soil-derived plied at 1/10 of the cefoxitin breakpoint concentration, perhaps
bacteria. Second, PCR analyses of the purified bacteriophage indicating that a lack of adequate selection by the lower cefoxitin
preparations yielded no evidence of bacterial DNA, and phage- concentration simply failed to promote transduction; the 2-fold
only plating controls gave no indication of bacterial growth, indi- decrease might thus represent an anomaly. It would be interesting
cating that the bacteriophage enrichments were not contaminated to test whether the transduction frequency also correlates with the
with bacterial DNA or viable coliform bacteria. The increased re- incubation time, as precedent exists for transduction efficiency
sistance in the presence of enriched bacteriophage must therefore peaking after a defined time period: in early transduction experi-
be specific to the phage; the resistance genes may have originated ments with soil, Zeph et al. (28) noted that optimal incubation
from bacteria in the biosolids from which the bacteriophage was times existed for P1 transduction of chloramphenicol resistance
enriched, or the bacteriophage may be promoting the transfer of into E. coli recipients. It is possible that different antibiotics are
resistance genes within the soil bacterial community. selecting for transduction by different sets of phage, each of which
Transduction is known to be feasible in soil (28) and is un- yields peak transduction after specific periods of time. Shousha et

November 2015 Volume 81 Number 22 Applied and Environmental Microbiology aem.asm.org 7911
Ross and Topp

al. (29) observed that of 243 bacteriophage randomly isolated the resistance reservoir and ecological network of the phage metagenome.
from chicken meat, about a quarter was able to transduce resis- Nature 499:219 –222. http://dx.doi.org/10.1038/nature12212.
tance to one or more of the five antibiotics tested into E. coli. 12. Calero-Cáceres W, Melgarejo A, Colomer-Lluch M, Stoll C, Lucena
F, Jofre J, Muniesa M. 2014. Sludge as a potential important source of
Resistance to kanamycin was transduced the most often, followed antibiotic resistance genes in both the bacterial and bacteriophage frac-
by that to chloramphenicol, while only a few phage transduced tions. Environ Sci Technol 48:7602–7611. http://dx.doi.org/10.1021
tetracycline or ampicillin resistance. It thus seems likely that some /es501851s.
ARGs are subject to transduction less frequently than others. In 13. Colomer-Lluch M, Jofre J, Muniesa M. 2011. Antibiotic resistance genes
our experiment, it is curious that ampicillin selection yielded no in the bacteriophage DNA fraction of environmental samples. PLoS One
6:e17549. http://dx.doi.org/10.1371/journal.pone.0017549.
transduction, while cefoxitin selection did, since beta-lactamases 14. Marti E, Variatza E, Balcazar J. 2014. Bacteriophages as a reservoir of
or ampC promoter or attenuator mutations (33) might reasonably extended spectrum ␤-lactamase and fluoroquinolone resistance genes in
yield resistance to both antibiotics. One possibility is that selection the environment. Clin Microbiol Infect 20:O456 –O459. http://dx.doi.org
with ampicillin did not yield an increase in transduction as robust /10.1111/1469-0691.12446.
as that seen for selection with cefoxitin. Thus, different types of 15. Rahube T, Marti R, Scott A, Tien YC, Murray R, Sabourin L, Zhang Y,
Duenk P, Lapen D, Topp E. 2014. Impact of fertilizing with raw or
selection might display different potentiating effects on the trans-
anaerobically digested sewage sludge on the abundance of antibiotic-
duction frequency. It would be interesting to test whether selec- resistant coliforms, antibiotic resistance genes, and pathogenic bacteria in
tion with various antibiotics during transduction yields increased soil and on vegetables at harvest. Appl Environ Microbiol 80:6898 – 6907.
resistance to unrelated antibiotics in our experimental setup. http://dx.doi.org/10.1128/AEM.02389-14.
Overall, our results suggest that the application of subclinical 16. Marti R, Tien YC, Murray R, Scott A, Sabourin L, Topp E. 2014. Safely
concentrations of antibiotics can promote bacteriophage-medi- coupling livestock and crop production systems: how rapidly do antibiotic
resistance genes dissipate in soil following a commercial application of
ated horizontal transfer of antibiotic resistance genes in agricul- swine or dairy manure? Appl Environ Microbiol 80:3258 –3265. http://dx
tural soil microbiomes. Further work is required to determine if .doi.org/10.1128/AEM.00231-14.
antibiotics entrained into soil through application of animal or 17. Marti R, Scott A, Tien Y-C, Murray R, Sabourin L, Zhang Y, Topp E.
human wastes might increase the risk of spreading antibiotic re- 2013. The impact of manure fertilization on the abundance of antibiotic
sistance to potential human pathogens. resistant bacteria and frequency of detection of antibiotic resistance genes
in soil, and on vegetables at harvest. Appl Environ Microbiol 79:5701–
5709. http://dx.doi.org/10.1128/AEM.01682-13.
ACKNOWLEDGMENTS 18. Thurber R, Haynes M, Breitbart M, Wegley L, Rohwer F. 2009. Labo-
This research was supported by competitive funding through the AAFC ratory procedures to generate viral metagenomes. Nat Protoc 4:470 – 483.
Growing Forward 2 program. J.R. was supported by the NSERC Visiting http://dx.doi.org/10.1038/nprot.2009.10.
Fellowship in Government Laboratories program. 19. Muyzer G, Dewaal E, Uitierlinden A. 1993. Profiling of complex micro-
We thank S. Gordon for valued technical assistance. bial populations by denaturing gradient gel electrophoresis analysis of
polymerase chain reaction-amplified genes coding for 16S rRNA. Appl
Environ Microbiol 59:695–700.
REFERENCES 20. Suzuki M, Taylor L, DeLong E. 2000. Quantitative analysis of small-
1. Martinez JL. 2009. Environmental pollution by antibiotics and by antibi- subunit rRNA genes in mixed microbial populations via 5=-nuclease as-
otic resistance determinants. Environ Pollut 157:2893–2902. http://dx.doi says. Appl Environ Microbiol 66:4605– 4614. http://dx.doi.org/10.1128
.org/10.1016/j.envpol.2009.05.051. /AEM.66.11.4605-4614.2000.
2. Muniesa M, Colomer-Lluch M, Jofre J. 2013. Potential impact of envi- 21. Walsh F, Ingenfeld A, Zampicolli M, Hilber-Bodmer M, Frey J, Duffy B.
ronmental bacteriophages in spreading antibiotic resistance genes. Future 2011. Real-time PCR methods for quantitative monitoring of streptomycin
Microbiol 8:739 –751. http://dx.doi.org/10.2217/fmb.13.32. and tetracycline resistance genes in agricultural ecosystems. J Microbiol
3. Marti E, Variatza E, Balcazar J. 2014. The role of aquatic ecosystems as Methods 86:150 –155. http://dx.doi.org/10.1016/j.mimet.2011.04.011.
reservoirs of antibiotic resistance. Trends Microbiol 22:36 – 41. http://dx 22. Bert F, Branger C, Lambert-Zechovsky N. 2002. Identification of PSE
.doi.org/10.1016/j.tim.2013.11.001. and OXA beta-lactamase genes in Pseudomonas aeruginosa using PCR-
4. Frost L, Leplae R, Summers A, Toussaint A. 2005. Mobile genetic restriction fragment length polymorphism. J Antimicrob Chemother 50:
elements: the agents of open source evolution. Nat Rev Microbiol 3:722– 11–18. http://dx.doi.org/10.1093/jac/dkf069.
732. http://dx.doi.org/10.1038/nrmicro1235. 23. United States Environmental Protection Agency. 2001. Method 1602:
5. Lang AS, Zhaxybayeva O, Beatty JT. 2012. Gene transfer agents: phage- male-specific (F⫹) and somatic coliphage in water by single agar layer
like elements of genetic exchange. Nat Rev Microbiol 10:472– 482. http: (SAL) procedure. United States Environmental Protection Agency, Wash-
//dx.doi.org/10.1038/nrmicro2802.
ington, DC.
6. Sander M, Schmieger H. 2001. Method for host-independent detection
24. Ashelford K, Day M, Fry J. 2003. Elevated abundance of bacteriophage
of generalized transducing bacteriophages in natural habitats. Appl Envi-
infecting bacteria in soil. Appl Environ Microbiol 69:285–289. http://dx
ron Microbiol 67:1490 –1493. http://dx.doi.org/10.1128/AEM.67.4.1490
.doi.org/10.1128/AEM.69.1.285-289.2003.
-1493.2001.
7. Penadés J, Chen J, Quiles-Puchalt N, Carpena N, Novick R. 2015. 25. Government of Canada. 2010. Canadian Integrated Program for Antimi-
Bacteriophage-mediated spread of bacterial virulence genes. Curr Opin crobial Resistance Surveillance (CIPARS) 2007. Public Health Agency of
Microbiol 23:171–178. http://dx.doi.org/10.1016/j.mib.2014.11.019. Canada, Guelph, ON, Canada.
8. Muniesa M, Imamovic L, Jofre J. 2011. Bacteriophages and genetic 26. Colomer-Lluch M, Jofre J, Muniesa M. 2014. Quinolone resistance
mobilization in sewage and faecally polluted environments. Microb Bio- genes (qnrA and qnrS) in bacteriophage particles from wastewater
technol 4:725–734. http://dx.doi.org/10.1111/j.1751-7915.2011.00264.x. samples and the effect of inducing agents on packaged antibiotic resis-
9. Parsley LC, Consuegra EJ, Kakirde KS, Land AM, Harper WF, Liles tance genes. J Antimicrob Chemother 69:1265–1274. http://dx.doi.org
MR. 2010. Identification of diverse antimicrobial resistance determinants /10.1093/jac/dkt528.
carried on bacterial, plasmid, or viral metagenomes from an activated 27. Chibani-Chennoufi S, Bruttin A, Dillmann M-L, Brüssow H. 2004.
sludge microbial assemblage. Appl Environ Microbiol 76:3753–3757. Phage-host interaction: an ecological perspective. J Bacteriol 186:3677–
http://dx.doi.org/10.1128/AEM.03080-09. 3686. http://dx.doi.org/10.1128/JB.186.12.3677-3686.2004.
10. Kenzaka T, Tani K, Nasu M. 2010. High-frequency phage-mediated gene 28. Zeph L, Onaga M, Stotzky G. 1988. Transduction of Escherichia coli by
transfer in freshwater environments determined at single-cell level. ISME bacteriophage P1 in soil. Appl Environ Microbiol 54:1731–1737.
J 4:648 – 659. http://dx.doi.org/10.1038/ismej.2009.145. 29. Shousha A, Awaiwanont N, Sofka D, Smulders FJ, Paulsen P, Szos-
11. Modi S, Lee H, Spina C, Collins J. 2013. Antibiotic treatment expands tak MP, Humphrey T, Hilbert F. 2015. Bacteriophages isolated from

7912 aem.asm.org Applied and Environmental Microbiology November 2015 Volume 81 Number 22
Antibiotic Resistance in Soil Phage

chicken meat and the horizontal transfer of antimicrobial resistance 32. Smillie C, Smith M, Friedman J, Cordero O, David L, Alm E. 2011.
genes. Appl Environ Microbiol 81:4600 – 4606. http://dx.doi.org/10 Ecology drives a global network of gene exchange connecting the
.1128/AEM.00872-15. human microbiome. Nature 480:241–244. http://dx.doi.org/10.1038
30. Allen H, Looft T, Bayles D, Humphrey S, Levine U, Alt D, Stanton T. /nature10571.
2011. Antibiotics in feed induce prophages in swine fecal microbiomes. 33. Mulvey M, Bryce E, Boyd D, Ofner-Agostini M, Land A, Simor A,
mBio 2:e00260-11. http://dx.doi.org/10.1128/mBio.00260-11. Paton S. 2005. Molecular characterization of cefoxitin-resistant Esche-
31. Perry J, Wright G. 2014. Forces shaping the antibiotic resistome. Bioes- richia coli from Canadian hospitals. Antimicrob Agents Chemother 49:
says 36:1179 –1184. http://dx.doi.org/10.1002/bies.201400128. 358 –365. http://dx.doi.org/10.1128/AAC.49.1.358-365.2005.

November 2015 Volume 81 Number 22 Applied and Environmental Microbiology aem.asm.org 7913
REVIEW doi:10.1038/nature10886

RNA-guided genetic silencing


systems in bacteria and archaea
Blake Wiedenheft1,2†, Samuel H. Sternberg3 & Jennifer A. Doudna1–4

Clustered regularly interspaced short palindromic repeat (CRISPR) are essential components of nucleic-acid-based
adaptive immune systems that are widespread in bacteria and archaea. Similar to RNA interference (RNAi) pathways
in eukaryotes, CRISPR-mediated immune systems rely on small RNAs for sequence-specific detection and silencing
of foreign nucleic acids, including viruses and plasmids. However, the mechanism of RNA-based bacterial immunity is
distinct from RNAi. Understanding how small RNAs are used to find and destroy foreign nucleic acids will provide new
insights into the diverse mechanisms of RNA-controlled genetic silencing systems.

B
acteria and archaea are the most diverse and abundant organisms Architecture and composition of CRISPR loci
on the planet, thriving in habitats that range from hot springs to The defining feature of CRISPR loci is a series of direct repeats
humans. However, viruses outnumber their microbial hosts in (approximately 20–50 base pairs) separated by unique spacer
every ecological setting, and the selective pressures imposed by these sequences of a similar length 11,33,34 (Fig. 2). The repeat sequences
rapidly evolving parasites has driven the diversification of microbial within a CRISPR locus are conserved, but repeats in different CRISPR
defence systems1–3. Historically, our understanding of antiviral immu- loci can vary in both sequence and length. In addition, the number
nity in bacteria has focused on restriction-modification systems, of repeat–spacer units in a CRISPR locus varies widely within and
abortive-phage phenotypes, toxin–antitoxins and other innate defence among organisms35.
systems4,5. More recently, bioinformatic, genetic and biochemical stud- The sequence diversity of these repetitive loci initially limited their
ies have revealed that many prokaryotes use an RNA-based adaptive detection and obscured their relationship, but computational methods
immune system to target and destroy genetic parasites (reviewed in refs have been developed for detecting repeat patterns rather than related
6–12). Such adaptive immunity, previously thought to occur only in sequences33,34,36–38. One of the first-generation pattern-recognition algo-
eukaryotes, provides an example of RNA-guided destruction of foreign rithms identified the repeat–spacer–repeat architecture in phylogeneti-
genetic material by a process that is distinct from RNA interference cally diverse bacterial and archaeal genomes, but related structures were
(RNAi) (Fig. 1). not identified in eukaryotic chromosomes39. Comparative analyses of
In response to viral and plasmid challenges, bacteria and archaea the sequences adjacent to the CRISPR loci have revealed an (A+T)-rich
integrate short fragments of foreign nucleic acid into the host chromo- ‘leader’ sequence that has been shown to serve as a promoter element
some at one end of a repetitive element known as CRISPR (clustered for CRISPR transcription39–42. In addition to the leader sequence, Jansen
regularly interspaced short palindromic repeat)13–15. These repetitive et al.39 identified a set of four CRISPR-associated (cas) genes known as
loci serve as molecular ‘vaccination cards’ by maintaining a genetic cas1–4 that are found exclusively in genomes containing CRISPRs. Based
record of prior encounters with foreign transgressors. CRISPR loci on sequence similarity to proteins of known function, Cas3 was predicted
are transcribed, and the long primary transcript is processed into a to be a helicase and Cas4 a RecB-like exonuclease39.
library of short CRISPR-derived RNAs (crRNAs)16–21 that each con- Subsequent bioinformatic analyses have shown that CRISPR loci are
tain a sequence complementary to a previously encountered invading flanked by a large number of extremely diverse cas genes32,43. The cas1
nucleic acid. Each crRNAs is packaged into a large surveillance complex gene is a common component of all CRISPR systems, and phylogenetic
that patrols the intracellular environment and mediates the detection analyses of Cas1 sequences indicate there are several versions of the
and destruction of foreign nucleic acid targets15,22–27. CRISPR system. Providing additional evidence for the classification
CRISPRs were originally identified in the Escherichia coli genome of distinct CRISPR types, neighbourhood analysis has identified con-
in 1987, when they were described as an unusual sequence element served arrangements of between four and ten cas genes that are found
consisting of a series of 29-nucleotide repeats separated by unique in association with CRISPR loci harbouring specific repeat sequences35.
32-nucleotide ‘spacer’ sequences28. Repetitive sequences with a similar These distinct immune systems have been divided into three major
repeat–spacer–repeat pattern were later identified in phylogenetically CRISPR types on the basis of gene conservation and locus organiza-
diverse bacterial and archaeal genomes, but the function of these repeats tion10. More than one CRISPR type is often found in a single organism,
remained obscure until many spacer sequences were recoginized as indicating that these systems are probably mutually compatible and could
being identical to viral and plasmid sequences29–31. This observation share functional components10. Despite the variation in number and
led to the hypothesis that CRISPRs provide a genetic memory of infec- diversity of cas genes, the distinguishing feature of all type I systems is that
tion29, and the detection of short CRISPR-derived RNA transcripts they encode a cas3 gene. The Cas3 protein contains an N-terminal HD
suggested that there may be functional similarities between CRISPR- phosphohydrolase domain and a C-terminal helicase domain32,39,43,44. In
based immunity and RNAi30,32. In this Insight, we review three stages of some type I systems, the Cas3 nuclease and helicase domains are encoded
CRISPR-based adaptive immunity and compare mechanistic aspects of by separate genes (cas3ʹʹ and cas3ʹ, respectively), but in each case they are
these immune systems to other RNA-guided genetic silencing pathways. thought to participate in degrading foreign nucleic acids22,44–46 (Fig. 2).
1
Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, Maryland 20815-6789, USA. 2Department of Molecular and Cell Biology, University of California, Berkeley, California 94720,
USA. 3Department of Chemistry, University of California, Berkeley, California 94720, USA. 4Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA.
†Present address: Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, USA

1 6 F E B R UA RY 2 0 1 2 | VO L 4 8 2 | N AT U R E | 3 3 1
© 2012 Macmillan Publishers Limited. All rights reserved
INSIGHT REVIEW

Type II CRISPR systems consist of just four cas genes, one of which cas genes in S. thermophilus demonstrated that csn2 (previously known
is always cas9 (formerly referred to as csn1). Cas9 is a large protein as cas7) is required for new spacer sequence acquisition14. This gene is
that includes both a RuvC-like nuclease domain and an HNH nucle- not conserved in other CRISPR types, which suggests that either the
ase domain. Studies in Streptococcus pyogenes and Streptococcus ther- mechanism of adaptation in S. thermophilus is distinct from the other
mophilus have indicated that Cas9 may participate in both CRISPR RNA types or that there are functional orthologues of Csn2 in other systems.
processing and target destruction14,15,17. Two variations of the type III Furthermore, gene deletion experiments in both S. thermophilus and
system have been identified (known as III-A and III-B). This division E. coli have shown that neither cas1 nor cas2 genes are required for
is supported by the functional differences reported in Staphylococcus CRISPR RNA processing or targeted interference22,53,54. These genetic
epidermidis and Pyrococcus furiosus47,48. The immune system in S. epi- studies suggest a role for Cas1 and Cas2 in the integration of foreign
dermidis (type III-A) targets plasmid DNA in vivo, whereas the purified DNA into the CRISPR.
components of the type III-B system in P. furiosus have been found to The role of Cas1 in CRISPR-mediated immunity is still uncertain;
cleave only single-stranded RNA substrates in vitro. The functional dis- however, biochemical and structural data indicate a function for Cas1 in
tinction between these two closely related systems suggests there could new–spacer–sequence acquisition54–56. Cas1 proteins from Pseudomo-
be other mechanistic differences between the distinct CRISPR subtypes. nas aeruginosa56, E. coli54 and Sulfolobus solfataricus55 have been purified
and studied biochemically. The Cas1 protein from S. solfataricus has
Integration of new information into CRISPR loci been shown to bind nucleic acids with high affinity (Kd ranging from 20
Acquisition of foreign DNA is the first step of CRISPR-mediated immu- to 50 nM), but without sequence preference55. The Cas1 protein from
nity (Fig. 2 and 3). During this stage, a short segment of DNA from an E. coli also binds to DNA with a preference for mismatched or abasic
invading virus or plasmid (known as the protospacer) is integrated pref- substrates57. This observation is consistent with a recent study show-
erentially at the leader end of the CRISPR locus14,15. Although metagen- ing a physical and genetic interaction between E. coli Cas1 and several
omic studies performed on environmental samples indicate that proteins associated with DNA replication and repair54.
CRISPRs evolve rapidly in dynamic equilibrium with resident phage Activity assays with Cas1 from P. aeruginosa and E. coli indicate that
populations13,49,50, the type II system in S. thermophilus is currently the Cas1 is a metal-dependent nuclease. The Cas1 protein from P. aeruginosa
only CRISPR system that has been shown to robustly acquire new phage is a DNA-specific nuclease, whereas the Cas1 protein from E. coli had a
or plasmid sequences in a pure culture. Phage-challenge experiments in nuclease activity on a wider range of nucleic acid substrates54,56. These
S. thermophilus have indicated that a small proportion of the cells in a in vitro assays suggest that Cas1 proteins interact with nucleic acids in a
population will typically incorporate a single virus-derived sequence at non-sequence-specific manner.
the leader end of a CRISPR locus14,15,51,52. The CRISPR-repeat sequence Crystal structures for five different Cas1 proteins are currently avail-
is duplicated for each new spacer seqenced added, thus maintaining the able (Protein Data Bank (PDB) identifiers: 3GOD, 3NKD, 3LFX, 3PV9
repeat–spacer–repeat architecture. Although the mechanism of spacer and 2YZS)54,56. Although the amino acid sequences for these proteins are
integration and replication of the repeat sequence is still unknown, extremely diverse (less than 15% sequence identity), their tertiary and
studies in S. thermophilus and E. coli have indicated that several Cas quaternary structures are similar. All Cas1 proteins seem to share a two-
proteins are involved in the process14,15,22,53. Mutational analysis of the domain architecture consisting of an N-terminal β-strand domain and a
CRISPR-mediated interference Eukaryotic RNA-interference

Foreign DNA Foreign RNA

Nucleus
CRISPR locus
Source piRNA locus miRNA locus
Repeat Repeat Repeat
CRISPR
Drosha
transcription

? miRNA siRNA
Cas or RNase III
crRNA RNA biogenesis piRNA 3′ Dicer
5′ 3′
crRNA-guided surveillance complex RNA-induced silencing complex
Cas protein(s) AGO/PIWI
Seed Seed
RNA-guided
5′ 3′ interference 5′

Target interference Target interference

Figure 1 | Parallels and distinctions between CRISPR RNA-guided act by restricting transposon mobility76–78. The biogenesis of piRNAs is
silencing systems and RNAi. CRISPR systems and RNAi recognize not yet fully understood. MicroRNAs (miRNAs) are also encoded on the
long RNA precursors that are processed into small RNAs, which act as chromosome, and primary miRNA transcripts form stable hairpin structures
sequence-specific guides for targeting complementary nucleic acids. In that are sequentially processed (shown by red triangles) by two RNase III
CRISPR systems, foreign DNA is integrated into the CRISPR locus, and long family endoribonucleases (Drosha and Dicer)79. miRNAs do not participate
transcripts from these loci are processed by a CRISPR-associated (Cas) or in genome defence but are major regulators of endogenous gene expression80.
RNase III family nuclease16–21,64. The short CRISPR-derived RNAs (crRNAs) Like crRNAs, eukaryotic piRNAs, siRNAs and miRNAs associate with
assemble with Cas proteins into large surveillance complexes that target proteins that facilitate complementary interactions with invading nucleic
destruction of invading genetic material15,22,24–27,48. In some eukaryotes, long acid targets27,60,69,79. In eukaryotes, the Argonaute proteins pre-order the 5ʹ
double-stranded RNAs are recognized as foreign, and a specialized RNase III region of the guide RNA into a helical configuration, reducing the entropy
family endoribonuclease (Dicer) cleaves these RNAs into short-interfering penalty of interactions with target RNAs69. This high-affinity binding site,
RNAs (siRNAs) that guide the immune system to invading RNA viruses76. called the ‘seed’ sequence, is essential for target sequence interactions. Recent
PIWI-interacting RNAs (piRNAs) are transcribed from repetitive clusters in studies indicate that the CRISPR system may use a similar seed-binding
the genome that often contain many copies of retrotransposons and primarily mechanism for enhancing target sequence interactions26,27,53,60.

3 3 2 | N AT U R E | VO L 4 8 2 | 1 6 F E B R UA RY 2 0 1 2
© 2012 Macmillan Publishers Limited. All rights reserved
REVIEW INSIGHT

C-terminal α-helical domain (Fig. 3). The C-terminal domain contains a saddle). Although this feature of the Cas1 structure did not initially stand
conserved divalent metal-ion binding site, and alanine substitutions of the out as a potential DNA-binding site, comparative analysis of the avail-
metal-coordinating residues inhibit Cas1-catalysed DNA degradation54,56. able Cas1 structures reveals a conserved set of positively charged residues
The metal ion is surrounded by a cluster of basic residues that form a along each of the β-hairpins that could contact the phosphate backbone.
strip of positive charge across the surface of the C-terminal domain. This The two β-hairpins, which are symmetrically related, might participate
positively charged surface may serve as an electrostatic snare to position in sequence-specific interactions with the CRISPR repeat, whereas the
nucleic-acid substrates near the catalytic metal ions56 (Fig. 3). The Cas1 large positively charged surface on the C-terminal α-helical domain could
protein forms a stable homodimer that is formed through interactions account for the high-affinity, non-sequence-specific interactions that have
between the two β-strand domains, which are related by a pseudo-two- been observed in vitro.
fold axis of symmetry54,56. This organization creates a saddle-like structure In spite of these structural studies and biochemical results, it is still only
that can be modelled onto double-stranded DNA without steric clashing. possible to speculate on the role of Cas1 in the integration of new spacer
β-hairpins, one from each of the two symmetrically related molecules, sequences, and many steps associated with the integration process still
hang on opposite faces of the double-stranded DNA (like stirrups on a need to be explained. For example, new spacer sequences are inserted

Invading virus

cas gene casette CRISPR

r Spacer Spacer
ce cas cas
spa Leader/promoter Repeat Repeat Repeat
o to
M Pr
PA
**

New spacer
Stage 1: Acquisition sequence
CRISPR trancription
Spacer Spacer
Leader/promoter Repeat Repeat Repeat Repeat
Repeat duplication

Stage 2: CRISPR RNA biogenesis


Type I Type II Type III

CRISPR transcription CRISPR transcription CRISPR transcription

RNase III
Leader CRISPR-
Leader specific
Leader endoribonuclease
tracrRNA tracrRNA tracrRNA ?
CRISPR- 5′ 3′
specific endoribonuclease

5′

3′3′ 5′ 3′ 5′ crRNA
5′ 3′
∼30-nt spacer trimming
No crRNA 3′ crRNA
trimming trimming
3′-Seed?
Cas3
5′ 5′-Seed?
Seed 5′ 3′
3′ ** 3′ PAM
PAM **
3′ 5′ 3′
3′
Target DNA
Ca
s3

Target either RNA or DNA


Target DNA

Stage 3: Interference

Figure 2 | Diversity of CRISPR-mediated adaptive immune systems end of the crRNA is trimmed by an unknown mechanism (green pacman,
in bacteria and archaea. A diverse set of CRISPR-associated (cas) right). In type II systems, a trans-acting antisense RNA (tracrRNA) with
genes (grey arrows) encode proteins required for new spacer sequence complementarity to the CRISPR RNA repeat sequence forms an RNA
acquisition (Stage 1), CRISPR RNA biogenesis (Stage 2) and target duplex that is recognized and cleaved by cellular RNase III (brown ovals)17.
interference (Stage 3). Each CRISPR locus consists of a series of direct This cleavage intermediate is further processed at the 5ʹ end resulting in
repeats separated by unique spacer sequences acquired from invading a mature, approximately 40-nucleotide crRNA with an approximately
genetic elements (protospacers). Protospacers are flanked by a short 20-nucleotide 3ʹ-handle. In each system, the mature crRNA associates with
motif called the protospacer adjacent motif (PAM, **) that is located on one or more Cas proteins to form a surveillance complex (green rectangles).
the 5ʹ (type I) or 3ʹ (type II) side in foreign DNA10,51,52,59,67. Long CRISPR Type I systems encode a Cas3 nuclease (blue pacman), which may be
transcripts are processed into short crRNAs by distinct mechanisms. In recruited to the surveillance complex following target binding24,27,44. A short
type I and III systems, a CRISPR-specific endoribonuclease (yellow ovals high-affinity binding site called a seed-sequence has been identified in some
and green circles, respectively) cleaves 8 nucleotides upstream of each type I systems27,60, and genetic experiments suggest that type II systems have
spacer sequence16,18–21,64. In type III systems, the repeat sequence on the 3ʹ a seed sequence located at the 3ʹ end of the crRNA spacer sequence53.

1 6 F E B R UA RY 2 0 1 2 | VO L 4 8 2 | N AT U R E | 3 3 3
© 2012 Macmillan Publishers Limited. All rights reserved
INSIGHT REVIEW

preferentially at the leader end of the CRISPR, but the mechanism of Crystal structures of CRISPR-specific endoribonucleases from two
leader end recognition is unknown. One simple model suggests that the other immune systems offer additional insights into the co-evolution-
leader sequence contains a recognition element that recruits the integra- ary relationship between these specialized enzymes and their cognate
tion machinery. It is equally possible that integration relies on single- RNAs16,19,21 (Fig. 4). In P. aeruginosa, Cas6f (previously known as Csy4)
stranded regions of the CRISPR DNA that are made available during specifically binds and cleaves the CRISPR-RNA-repeat 8 nucleotides
transcription. Transcription-associated recombination is involved in upstream of the spacer sequence, which leaves a similar 8-nucleotide
genome stability58, and a mechanism that couples integration together 5ʹ-handle on mature crRNAs19. The co-crystal structure of Cas6f bound
with transcription would link the process of adaptation to CRISPR RNA to its cognate RNA reveals interesting parallels between the method of
expression, ensuring that spacers from the most recent virus or plasmid RNA binding used by Cas6f and Cas6e18–20. Like Cas6e, the P. aeruginosa
are transcribed first. Cas6f protein recognizes the sequence and shape of a stable stem-loop
The integration machinery must be able to distinguish foreign DNA in the crRNA repeat sequence by interacting extensively with the major
from that of the host genome. The molecular cues that are involved in the groove of the double-stranded RNA. However, the structural elements
distinction of ‘self ’ from ‘non-self ’ are still unknown, but sequencing of responsible for this interaction are distinct between the two proteins18–20
CRISPR loci following phage challenge suggests that spacer sequences (Fig. 4). The Cas6f protein has a two-domain architecture, which con-
are not selected at random15,29,51,52,59,60. Mapping spacer sequences onto sists of an N-terminal ferredoxin-like fold similar to that in Cas6e, but
viral genomes reveals a short sequence motif proximal to the protospacer, its C-terminal domain is structurally distinct. An arginine-rich helix
which is referred to as the protospacer adjacent motif (PAM). PAM in the C-terminal domain of Cas6f inserts into the major groove of the
sequences are only a few nucleotides long, and the precise sequence var- crRNA duplex, and the bottom of the crRNA is positioned for sequence-
ies depending on the CRISPR system type59. This variation suggests that specific hydrogen-bonding contacts in the RNA major groove. These
one or more of the Cas proteins associated with each immune system is contacts position the scissile phosphate of the crRNA in the enzyme
involved in PAM recognition, but the mechanism governing this specific- active site so that cleavage occurs 8 nucleotides upstream of the spacer
ity is unknown. sequence18–20 (Fig. 4).
Although Cas6f and Cas6e recognize the sequence and shape of the
CRISPR RNA biogenesis crRNA hairpin in their respective systems, CRISPR RNA repeats in
Spacer acquisition is the first step of immunization, but successful protec- other CRISPR systems are thought to be unstructured35. For example,
tion from bacteriophage or plasmid challenge requires the CRISPR to be the Cas6 protein from P. furiosus associates with CRISPR transcripts that
transcribed and processed into short CRISPR-derived RNAs (crRNAs). are expected to contain unstructured repeats64. The specific recognition
crRNAs were first detected by small RNA profiling in Archaeoglobus of an unstructured RNA repeat requires a distinct mechanistic solution
fulgidus61 and S. solfataricus62. Northern-blot analysis using probes for RNA substrate discrimination. Remarkably, crystallographic studies
against the repeat sequence of the CRISPR revealed a ‘ladder-like’ pat- of the Cas6 protein from P. furiosus have revealed the same duplicated
tern of RNA consistent with a long precursor CRISPR RNA transcript ferredoxin-like fold observed in the Cas6e protein, but with a different
(pre-crRNA) that was processed at approximately 60-nucleotide inter- mode of RNA recognition involving the opposite face of the protein
vals. In fact, the 3ʹ ends of cloned crRNAs were mapped to the middle (Fig. 4). In Cas6, the two ferredoxin-like folds clamp the 5ʹ end of the
of the CRISPR repeat61, which suggested that the repeat sequence was single-stranded RNA repeat sequence in place21. Although the RNA in
recognized and cleaved. this structure is disordered in the enzyme active site, biochemical studies
The need for crRNAs in CRISPR-mediated defence was demon- have shown that cleavage occurs 8 nucleotides upstream of the spacer
strated initially by investigation of a CRISPR-specific endoribonucle- sequence16,64. While the nucleotide sequences at the cleavage site vary for
ase in E. coli called Cas6e (formerly known as Cse3 or CasE)22. Cas6e each of the different Cas6 proteins, all Cas6 family endoribonucleases
specifically binds and cleaves within each repeat sequence of the long cleave their cognate RNA 8 nucleotides upstream of the spacer sequence
pre-crRNA, resulting in a library of crRNAs that each contain a unique using a metal-ion-independent mechanism.
spacer sequence flanked by fragments of the adjacent repeats. Mutation Despite advances in our understanding of crRNA biogenesis, the
of a conserved histidine blocks crRNA biogenesis and leaves the cell diversity of cas genes has obscured identification of the protein fac-
susceptible to phage infection22. tors responsible for CRISPR RNA processing in some systems. Type II
The Cas6e protein consists of a double ferredoxin-like fold that selec- immune systems consist of four cas genes, none of which have a detect-
tively associates with specific RNA repeats and does not associate with able sequence similarity to known CRISPR-specific endoribonucleases.
DNA or CRISPR RNAs containing a non-cognate repeat sequence Recently, a different CRISPR RNA processing mechanism has been
18,20,22,63
(Fig. 4). Crystal structures of Cas6e bound to a CRISPR RNA reported that involves RNase-III-mediated cleavage of double-stranded
repeat reveal a combination of sequence- and structure-specific interac- regions of the CRISPR RNA repeats17. The first indication of this mecha-
tions that explain the molecular mechanism of substrate recognition18,20. nism came from deep sequencing of RNA from S. pyogenes. An abundant
The repeat sequence of the E. coli CRISPR is partially palindromic, and transcript containing a 25-nucleotide sequence that was complemen-
the RNA forms a stable (approximately 20-nucleotide) stem loop22,35. A tary to the CRISPR repeat was identified. This RNA, termed tracrRNA
positively charged β-hairpin in Cas6e interacts with the major groove of (trans-activating CRISPR RNA), is coded on the opposite strand and just
the RNA duplex, which positions the 3ʹ strand of the crRNA stem along a upstream of the CRISPR locus. Genetic and biochemical experiments
conserved, positively charged cleft on one face of the protein18,20 (Fig. 4). demonstrated that tracrRNA and pre-crRNA are co-processed by RNase
RNA binding induces a conformational change that disrupts the bot- III, which produces cleavage products with a 2 nucleotide 3ʹ overhang17.
tom base pair of the stem and positions the scissile phosphate within In vivo processing of CRISPR RNAs required Cas9 (previously known as
the enzyme active site for site-specific cleavage20. CRISPR RNA cleav- Csn1), although a precise role for this enzyme in RNA processing has not
age occurs 8 nucleotides upstream of the spacer sequence, which results yet been defined. The essential role of cellular proteins that are not solely
in 61-nucleotide mature crRNAs consisting of a 32-nucleotide spacer involved in CRISPR-mediated defence, such as RNase III, indicates that
flanked by 8 nucleotides of the repeat sequence on the 5ʹ end (known different host factors may be involved as ancillary components of these
as the 5ʹ-handle) and 21 nucleotides of the remaining repeat sequence immune systems.
on the 3ʹ end (Fig. 4). Cas6e remains tightly bound to the 3ʹ stem-loop20
and may serve as a nucleation point for assembly of a large effector com- crRNA-guided interference
plex, Cascade (CRISPR-associated complex for antiviral defence), that is The third stage of CRISPR-mediated immunity is target interference
required for phage silencing in the next stage of the immune system22,24,26 (Fig. 2). Here crRNAs associate with Cas proteins to form large CRISPR-
(discussed later). associated ribonucleoprotein complexes that can recognize invading

3 3 4 | N AT U R E | VO L 4 8 2 | 1 6 F E B R UA RY 2 0 1 2
© 2012 Macmillan Publishers Limited. All rights reserved
REVIEW INSIGHT

a b
Leader recognition

Leader
CRISPR DNA Repeat Repeat

β-hairpin Protospacer selection


PAM
~20 Å
Viral DNA Protospacer

DNA binding? 0 10 20 30
Base pairs

Recombination/repair
New ‘spacer’ acquisition

Leader
CRISPR DNA Repeat Repeat Repeat

Figure 3 | Steps leading to new spacer integration. a, The Cas1 protein metal ions (green sphere). b, CRISPR adaptation occurs by integrating
forms a stable homodimer where the two molecules (green and grey) are fragments of foreign nucleic acid preferentially at the leader end of the
related by a pseudo-two-fold axis of symmetry (PBD ID: 3GOD)54,56. This CRISPR, forming new repeat-spacer units in the process. Protospacers
organization creates a saddle-like structure in the N-terminal domain, are chosen non-randomly and may be selected from regions flanking the
in which β-hairpins (blue) from each symmetrically related molecule protospacer adjacent motif (PAM). Coordinated cleavage of the foreign
hang (like stirrups) that are separated by approximately 20 Å, and may DNA and integration of the protospacer into the leader-end of the CRISPR
interact with the phosphodiester backbone of double-stranded DNA. An occurs through a mechanism that duplicates the repeat sequence and
electrostatic surface representation (bottom) reveals a cluster of basic thus preserves the repeat-spacer-repeat architecture of the CRISPR locus.
residues (blue) that form a positively charged strip across the metal- Although the protein components required for this process have not been
binding surface of the C-terminal domain. This strip may serve as an conclusively identified, Cas1 and other general recombination or repair
electrostatic trap that positions DNA substrates proximally to catalytic factors have been implicated (blue ovals)32,54,56.

nucleic acids. Foreign nucleic acids are identified by base-pairing interac- Cas proteins directly participate in target binding. Recent bio-
tions between the crRNA spacer sequence and a complementary sequence chemical studies have shown that CRISPR-associated complexes
from the intruder. Phage- and plasmid-challenge experiments performed facilitate target recognition by enhancing sequence-specific
in several model systems have demonstrated that crRNAs complementary hybridization between the CRISPR RNA and complementary target
to either the coding or the non-coding strand of the invading DNA can sequences27. A short high-affinity binding site located at one end of
provide immunity14,22,47,60,65,66. This is indicative of an RNA-guided DNA- the crRNA spacer sequence governs the efficiency of target binding,
targeting system, and indeed a pathway for DNA silencing has recently and viruses that acquired a single mismatch in this region were able
been demonstrated in S. thermophilus15. DNA sequencing and Southern to escape detection by the immune system60. This high-affinity bind-
blots indicated that both strands of the target DNA are cleaved within ing site is functionally analogous to the ‘seed’ sequence (Fig. 1) that
the region that is complementary to the crRNA spacer sequence15. This has been identified in eukaryotic microRNAs (miRNAs)68. Struc-
mechanism efficiently eliminates foreign DNA sequences, which have tural and biochemical studies have shown that Argonaute proteins
been specified by the spacer region of the crRNA, but avoids targeting facilitate target recognition by pre-ordering the nucleotides at the
the complementary DNA sequences in the CRISPR region of the host 5ʹ end of the miRNA in a helical configuration69. This pre-ordering
chromosome. The mechanism for distinguishing self from non-self is reduces the entropic penalty that is associated with helix forma-
built into the crRNA. The spacer sequence of each crRNA is flanked by tion and provides a thermodynamic advantage for target binding
a portion of the adjacent CRISPR repeat sequence, and any complemen- within this region. A similar mechanism may occur during crRNA
tarity beyond the spacer into the adjacent repeat region signals self and target binding, providing an interesting example of convergent evo-
prevents the destruction of the host chromosome67. lution between CRISPR-based immunity in prokaryotes and RNAi
However, not all CRISPR systems target DNA. In vitro experiments in eukaryotes (Fig. 1).
using enzymes from the type III-B CRISPR system of P. furiosus have Structural and biochemical studies have been performed on
shown that this system cleaves target RNA rather than DNA48. All DNA CRISPR-associated complexes isolated from three different type I
targeting systems encode a complementary DNA sequence for each CRISPR systems24–27,48. These complexes������������������������
seem to share some gen-
crRNA in the CRISPR locus and therefore require a mechanism for distin- eral morphological features, but the precise special arrangement of
guishing self (CRISPR locus) from non-self (invading DNA). In contrast, the Cas proteins and their interactions with the crRNA have been
systems that target RNA may not be required to make this distinction unclear. Sub-nanometre-resolution structures of the CRISPR-asso-
because most CRISPR loci are transcribed only in one direction and thus ciated complex from E. coli (Cascade) have recently been determined
do not generate complementary RNA targets. CRISPR systems that tar- using cryo-electron microscopy26. This complex is comprised of an
get RNA may be uniquely capable of defending against viruses that have unequal stoichiometry of 5 functionally essential Cas proteins and
RNA-based genomes. However, adaptation of the CRISPR in response a 61-nucleotide crRNA22,24,26. The structure reveals a sea-horse-
to a challenge by an RNA-based virus will probably require the invading shaped architecture in which the crRNA is displayed along a helical
RNA to be reverse-transcribed into DNA before it can be integrated into arrangement of protein subunits that protect the crRNA from deg-
the CRISPR locus. radation26. The 5ʹ and 3ʹ ends of the crRNA form unique structures

1 6 F E B R UA RY 2 0 1 2 | VO L 4 8 2 | N AT U R E | 3 3 5
© 2012 Macmillan Publishers Limited. All rights reserved
INSIGHT REVIEW

Type I Type II Type III

Cleavage
Cleavage Cleavage Cleavage

Cleavage

Cas6e Cas6f tracrRNA RNase III Cas6

pre-crRNA:

Cas9 RNase III Cas6


Cas6e/Cas6f

3′ trimming ?
5′ trimming ?
crRNAs:

8-nt 5′-handle 20-nt 3′-handle 8-nt 5′-handle

Figure 4 | Diverse mechanisms of CRISPR RNA biogenesis. CRISPR removes leftover repeat sequences from the 5ʹ end. Cas6 (PDB ID: 3PKM)
RNA repeats are specifically recognized and cleaved by diverse in type III-B CRISPR systems specifically recognizes single-stranded
mechanisms. In type I CRISPR systems, Cas6e (PDB ID: 2Y8W) and Cas6f RNA, upstream of the scissile phosphate, on a face of the protein opposite
(PDB ID: 2XLK) recognize the major groove of the crRNA stem-loop that of the previously identified active site residues16,21,64. The remainder of
primarily through electrostatic interactions using a β-hairpin and α-helix, the repeat substrate probably wraps around the protein (red dashed line)
respectively18,19,20. Cleavage occurs at the double-stranded–single-stranded to allow cleavage 8 nucleotides upstream of the repeat-spacer junction.
junction (black arrows), leaving an 8-nt 5ʹ-handle on mature crRNAs. In Subsequent 3ʹ trimming (red arrows) generates mature crRNAs of two
type II CRISPR systems, tracrRNA hybridizes to the pre-crRNA repeat discrete lengths. The N-terminal domain of all Cas 6 family proteins
to form duplex RNAs that are substrates for endonucleolytic cleavage by adopts a ferredoxin-like fold (light blue).The C-terminal domain of Cas6
host RNase III (PDB ID: 2EZ6), an activity that may also require Cas9 and Cas6e also adopts a ferredoxin-like fold but the C-terminal domain of
(ref. 17). Subsequent trimming (red arrows) by an unidentified nuclease Cas6f is structurally distinct (dark blue).

that are anchored at opposite ends of the Cascade complex, dis- Laboratory strains of bacteria are grown in high-density bioreactors for
playing the 32-nucleotide spacer sequence for base-pairing with many different applications in the food industry, and they are becoming
complementary targets. increasingly important in the production of biofuels. CRISPR systems
The structure of Cascade bound to a 32-nucleotide target sequence26 offer a natural mechanism for adapting economically important bacteria
reveals a concerted conformational change that could be a signal for for resistance against multiple phages.
recruiting Cas3. Cas3 — the trans-acting nuclease of type I CRISPR sys- The biochemical activities of various Cas proteins may have use-
tems — may function as a target ‘slicer’ in a similar way to Argonaute in ful applications in molecular biology in much the same way that DNA
RNAi pathways22,44,46,70. Although Cas3 was implicated previously in the restriction enzymes have revolutionized cloning and DNA manipulation.
process of self versus non-self discrimination, recent studies have dem- A wide range of CRISPR-specific endoribonucleases that recognize small
onstrated that Cascade recognizes the PAM directly and that mutations RNA motifs with high affinity expand the number of tools available for
in the PAM decrease Cascade’s affinity for the target60. The importance manipulating nucleic acids. In addition, a crRNA-guided ribonucleopro-
of the PAM is highlighted by the recovery of phage and plasmid escape tein complex in P. furiosus was shown to cleave target RNAs48. Site-specific
mutants, which frequently contain a single mutation in the PAM15,51–53,60. cleavage of target RNA molecules could have a range of uses, from gen-
The structure of Cascade indicates that the PAM is positioned near the erating homogeneous termini after in vitro transcription to targeting a
‘tail’ of the sea-horse-shaped complex. High-resolution structures and specific intracellular messenger RNA for inactivation in a similar way to
mutational analysis of the nucleic acid and protein components in this RNAi. CRISPRs also provide a new mechanism for limiting the spread of
and related systems are needed to determine the mechanisms of target antibiotic resistance or the transfer of virulence factors by blocking hori-
authentication and degradation. zontal gene transfer15,47. In addition, CRISPRs participate in a regulatory
mechanism that alters biofilm formation in P. aeruginosa74,75. Although
Applications of CRISPR structure and function the clinical relevance of CRISPRs remains to be demonstrated, the oppor-
The sequence diversity of CRISPR loci, even within closely related tunities for creative implementation of this new gene-regulation system
strains, has been used for high-resolution genotyping and forensic medi- are perceivably vast.
cine. This technique, known as spoligotyping (spacer oligotyping), has
been applied successfully to the analysis of human pathogens, including Future directions of CRISPR biology
Mycobacterium tuberculosis71, Corynebacterium diphtheriae72 and Salmo- The discovery of some of the fundamental mechanisms of CRISPR-
nella enterica73. Spoligotyping was developed long before the function based adaptive immunity has raised new questions and highlighted the
of CRISPRs was understood, but now that studies have begun to reveal areas with the greatest potential for future research. Although CRISPR
the biological function and mechanism of CRISPR-mediated genetic RNA processing and targeting steps are now understood in some detail,
silencing, new opportunities for creative applications have emerged. how and when target sequences are identified during a phage infection

3 3 6 | N AT U R E | VO L 4 8 2 | 1 6 F E B R UA RY 2 0 1 2
© 2012 Macmillan Publishers Limited. All rights reserved
REVIEW INSIGHT

or plasmid transformation are still unclear. Furthermore, why DNA or 26. Wiedenheft, B. et al. Structures of the RNA-guided surveillance complex from a
bacterial immune system. Nature 477, 486–489 (2011).
RNA target sequences are chosen, and their fate once they are bound This paper presented the first sub-nanometer resolution structures
to a crRNA-targeting complex is not well understood. In addition, the of Cascade, showing how the crRNA is accommodated within a large
mechanisms by which foreign sequences are selected and integrated ribonucleoprotein complex that is involved in foreign nucleic acid surveillance.
27. Wiedenheft, B. et al. RNA-guided complex from a bacterial immune system
into CRISPR loci are almost entirely unknown. Some CRISPR loci seem enhances target recognition through seed sequence interactions. Proc. Natl
to be considerably more active than others, at least under laboratory Acad. Sci. USA 108, 10092–10097 (2011).
conditions, so selection of the model organisms will be important. The 28. Ishino, Y., Shinagawa, H., Makino, K., Amemura, M. & Nakata, A. Nucleotide
diversity and prevalence of CRISPR systems throughout microbial com- sequence of the iap gene, responsible for alkaline phosphatase isozyme
conversion in Escherichia coli, and identification of the gene product. J. Bacteriol.
munities ensures that new findings and applications in this field will be 169, 5429–5433 (1987).
forthcoming in the years ahead. ■ 29. Bolotin, A., Ouinquis, B., Sorokin, A. & Ehrlich, S. D. Clustered regularly
interspaced short palindrome repeats (CRISPRs) have spacers of
extrachromosomal origin. Microbiology 151, 2551–2561 (2005).
1. Hoskisson, P. A. & Smith, M. C. Hypervariation and phase variation in the
30. Mojica, F. J., Diez-Villasenor, C., Garcia-Martinez, J. & Soria, E. Intervening
bacteriophage ‘resistome’. Curr. Opin. Microbiol. 10, 396–400 (2007).
sequences of regularly spaced prokaryotic repeats derive from foreign genetic
2. Rodriguez-Valera, F. et al. Explaining microbial population genomics through
elements. J. Mol. Evol. 60, 174–182 (2005).
phage predation. Nature Rev. Microbiol. 7, 828–836 (2009).
31. Pourcel, C., Salvignol, G. & Vergnaud, G. CRISPR elements in Yersinia pestis
3. Weinbauer, M. G. Ecology of prokaryotic viruses. FEMS Microbiol. Rev. 28,
acquire new repeats by preferential uptake of bacteriophage DNA, and provide
127–181 (2004).
additional tools for evolutionary studies. Microbiology 151, 653–663 (2005).
4. Labrie, S. J., Samson, J. E. & Moineau, S. Bacteriophage resistance mechanisms.
32. Makarova, K. S., Grishin, N. V., Shabalina, S. A., Wolf, Y. I. & Koonin, E. V.
Nature Rev. Microbiol. 8, 317–327 (2010).
A putative RNA-interference-based immune system in prokaryotes:
5. Stern, A. & Sorek, R. The phage-host arms race: shaping the evolution of
computational analysis of the predicted enzymatic machinery, functional
microbes. Bioessays 33, 43–51 (2010).
analogies with eukaryotic RNAi, and hypothetical mechanisms of action. Biol.
6. Al-Attar, S., Westra, E. R., van der Oost, J. & Brouns, S. J. Clustered regularly
Direct 1, 7 (2006).
interspaced short palindromic repeats (CRISPRs): the hallmark of an ingenious
33. Grissa, I., Vergnaud, G. & Pourcel, C. CRISPRFinder: a web tool to identify
antiviral defense mechanism in prokaryotes. Biol. Chem. 392, 277–289 (2011).
clustered regularly interspaced short palindromic repeats. Nucleic Acids Res.
7. Deveau, H., Garneau, J. E. & Moineau, S. CRISPR/Cas system and its role in
phage-bacteria interactions. Annu. Rev. Microbiol. 64, 475–493 (2010). 35, W52–W57 (2007).
8. Horvath, P. & Barrangou, R. CRISPR/Cas, the immune system of bacteria and 34. Rousseau, C., Gonnet, M., Le Romancer, M. & Nicolas, J. CRISPI: a CRISPR
archaea. Science 327, 167–170 (2010). interactive database. Bioinformatics 25, 3317–3318 (2009).
9. Karginov, F. V. & Hannon, G. J. The CRISPR system: small RNA-guided defense 35. Kunin, V., Sorek, R. & Hugenholtz, P. Evolutionary conservation of sequence and
in bacteria and archaea. Mol. Cell 37, 7–19 (2010). secondary structures in CRISPR repeats. Genome Biol. 8, R61 (2007).
10. Makarova, K. S. et al. Evolution and classification of the CRISPR-Cas systems. 36. Bland, C. et al. CRISPR recognition tool (CRT): a tool for automatic detection of
Nature Rev. Microbiol. 9, 467–477 (2011). clustered regularly interspaced palindromic repeats. BMC Bioinformatics 8, 209
11. Marraffini, L. A. & Sontheimer, E. J. CRISPR interference: RNA-directed adaptive (2007).
immunity in bacteria and archaea. Nature Rev. Genet. 11, 181–190 (2010). 37. Dsouza, M., Larsen, N. & Overbeek, R. Searching for patterns in genomic data.
12. Sorek, R., Kunin, V. & Hugenholtz, P. CRISPR-a widespread system that provides Trends Genet. 13, 497–498 (1997).
acquired resistance against phages in bacteria and archaea. Nature Rev. 38. Edgar, R. C. PILER-CR: fast and accurate identification of CRISPR repeats. BMC
Microbiol. 6, 181–186 (2008). Bioinformatics 8, 18 (2007).
13. Andersson, A. F. & Banfield, J. F. Virus population dynamics and acquired virus 39. Jansen, R., Embden, J. D., Gaastra, W. & Schouls, L. M. Identification of genes
resistance in natural microbial communities. Science 320, 1047–1050 (2008). that are associated with DNA repeats in prokaryotes. Mol. Microbiol. 43,
14. Barrangou, R. et al. CRISPR provides acquired resistance against viruses in 1565–1575 (2002).
prokaryotes. Science 315, 1709–1712 (2007). 40. Pougach, K. et al. Transcription, processing and function of CRISPR cassettes in
This study provided the first direct evidence for CRISPR immune system Escherichia coli. Mol. Microbiol. 77, 1367–1379 (2010).
function, demonstrating that new spacer acquisition provides resistance 41. Pul, U. et al. Identification and characterization of E. coli CRISPR-cas promoters
against future phage infection in a cas gene-dependent manner. and their silencing by H-NS. Mol. Microbiol. 75, 1495–1512 (2010).
15. Garneau, J. E. et al. The CRISPR/Cas bacterial immune system cleaves 42. Westra, E. R. et al. H-NS-mediated repression of CRISPR-based immunity in
bacteriophage and plasmid DNA. Nature 468, 67–71 (2010). Escherichia coli K12 can be relieved by the transcription activator LeuO. Mol.
This study showed that CRISPRs can acquire new spacers upon plasmid Microbiol. 77, 1380–1393 (2010).
challenge and provided the first example of crRNA-guided cleavage of 43. Haft, D. H., Selengut, J., Mongodin, E. F. & Nelson, K. E. A guild of 45 CRISPR-
double-stranded DNA in vivo. associated (Cas) protein families and multiple CRISPR/Cas subtypes exist in
16. Carte, J., Wang, R., Li, H., Terns, R. M. & Terns, M. P. Cas6 is an endoribonuclease prokaryotic genomes. PLoS Comput. Biol. 1, e60 (2005).
that generates guide RNAs for invader defense in prokaryotes. Genes Dev. 22, 44. Sinkunas, T. et al. Cas3 is a single-stranded DNA nuclease and ATP-dependent
3489–3496 (2008). helicase in the CRISPR/Cas immune system. EMBO J. 30, 1335–1342 (2011).
17. Deltcheva, E. et al. CRISPR RNA maturation by trans-encoded small RNA and 45. Han, D. & Krauss, G. Characterization of the endonuclease SSO2001 from
host factor RNase III. Nature 471, 602–607 (2011). Sulfolobus solfataricus P2. FEBS Lett. 583, 771–776 (2009).
This study identified a new CRISPR RNA processing pathway that involves 46. Mulepati, S. & Bailey, S. Structural and biochemical analysis of the nuclease
a trans-acting RNA complementary to the CRISPR repeat sequence and domain of the clustered regularly interspaced short palindromic repeat
demonstrated that processing of this duplex is mediated by cellular RNase III. (CRISPR) associated protein 3(CAS3). J. Biol. Chem. 286, 31896–31903
18. Gesner, E. M., Schellenberg, M. J., Garside, E. L., George, M. M. & MacMillan, A. M. (2011).
Recognition and maturation of effector RNAs in a CRISPR interference pathway 47. Marraffini, L. A. & Sontheimer, E. J. CRISPR interference limits horizontal gene
Nature Struct. Mol. Biol. 18, 688–692 (2011). transfer in staphylococci by targeting DNA. Science 322, 1843–1845 (2008).
19. Haurwitz, R. E., Jinek, M., Wiedenheft, B., Zhou, K. & Doudna, J. A. Sequence- 48. Hale, C. R. et al. RNA-guided RNA cleavage by a CRISPR RNA-Cas protein
and structure-specific RNA processing by a CRISPR endonuclease. Science complex. Cell 139, 945–956 (2009).
329, 1355–1358 (2010). Most CRISPR systems appear to target DNA, but this article reported the
20. Sashital, D. G., Jinek, M. & Doudna, J. A. An RNA induced conformational isolation of a multisubunit ribonucleoprotein complex (Cmr-complex) from P.
change required for CRISPR RNA cleavage by the endonuclease Cse3. Nature furriosus that cleaves target RNAs (not DNA) at a fixed distance from the 3΄
Struct. Mol. Biol. 18, 680–687 (2011). end of the crRNA-guide.
21. Wang, R., Preamplume, G., Terns, M. P., Terns, R. M. & Li, H. Interaction of the 49. Tyson, G. W. & Banfield, J. F. Rapidly evolving CRISPRs implicated in acquired
Cas6 riboendonuclease with CRISPR RNAs: recognition and cleavage. Structure resistance of microorganisms to viruses. Environ. Microbiol. 10, 200–207
19, 257–264 (2011). (2008).
22. Brouns, S. J. et al. Small CRISPR RNAs guide antiviral defense in prokaryotes. 50. Snyder, J. C., Bateson, M. M., Lavin, M. & Young, M. J. Use of cellular CRISPR
Science 321, 960–964 (2008). (clusters of regularly interspaced short palindromic repeats) spacer-based
This paper revealed that long CRISPR RNA transcripts are processed microarrays for detection of viruses in environmental samples. Appl Environ
into small RNAs (crRNAs) by a dedicated endoribonuclease and that the Microbiol 76, 7251–7258 (2010).
processed RNAs are bound by a Cas protein complex (Cascade), which 51. Deveau, H. et al. Phage response to CRISPR-encoded resistance in
together with Cas3 are required for phage protection. Streptococcus thermophilus. J. Bacteriol. 190, 1390–1400 (2008).
23. Hale, C., Kleppe, K., Terns, R. M. & Terns, M. P. Prokaryotic silencing (psi)RNAs in 52. Horvath, P. et al. Diversity, activity, and evolution of CRISPR loci in Streptococcus
Pyrococcus furiosus. RNA 14, 2572–2579 (2008). thermophilus. J. Bacteriol. 190, 1401–1412 (2008).
24. Jore, M .M. et al. Structural basis for CRISPR RNA-guided DNA recognition by 53. Sapranauskas, R. et al. The Streptococcus thermophilus CRISPR/Cas system
Cascade. Nature Struct. Mol. Biol. 18, 529–536 (2011). provides immunity in Escherichia coli. Nucleic Acids Res. 39, 9275–9282
25. Lintner, N. G. et al. Structural and functional characterization of an archaeal (2011).
clustered regularly interspaced short palindromic repeat (crispr)-associated 54. Babu, M. et al. A dual function of the CRISPR-Cas system in bacterial antivirus
complex for antiviral defense (CASCADE). J. Biol. Chem. 286, 21643–21656 immunity and DNA repair. Mol. Microbiol. 79, 484–502 (2011).
(2011). 55. Han, D., Lehmann, K. & Krauss, G. SSO1450--a CAS1 protein from Sulfolobus

1 6 F E B R UA RY 2 0 1 2 | VO L 4 8 2 | N AT U R E | 3 3 7
© 2012 Macmillan Publishers Limited. All rights reserved
INSIGHT REVIEW

solfataricus P2 with high affinity for RNA and DNA. FEBS Lett. 583, 1928– effector enzyme of the CRISPR interference. EMBO J. 30, 4616–4627 (2011).
1932 (2009). 71. Groenen, P. M., Bunschoten, A. E., van Soolingen, D. & van Embden, J. D.
56. Wiedenheft, B. et al. Structural basis for DNase activity of a conserved protein Nature of DNA polymorphism in the direct repeat cluster of Mycobacterium
implicated in CRISPR-mediated antiviral defense. Structure 17, 904–912 tuberculosis; application for strain differentiation by a novel typing method. Mol.
(2009). Microbiol. 10, 1057–1065 (1993).
57. Chen, C. S. et al. A proteome chip approach reveals new DNA damage 72. Mokrousov, I., Limeschenko, E., Vyazovaya, A. & Narvskaya, O. Corynebacterium
recognition activities in Escherichia coli. Nature Methods 5, 69–74 (2008). diphtheriae spoligotyping based on combined use of two CRISPR loci.
58. Aguilera, A. The connection between transcription and genomic instability. Biotechnol. J. 2, 901–906 (2007).
EMBO J. 21, 195–201 (2002). 73. Liu, F. et al. Novel virulence gene and clustered regularly interspaced short
59. Mojica, F. J., Diez-Villasenor, C., Garcia-Martinez, J. & Almendros, C. Short motif palindromic repeat (CRISPR) multilocus sequence typing scheme for subtyping
sequences determine the targets of the prokaryotic CRISPR defence system. of the major serovars of Salmonella enterica subsp. enterica. Appl. Environ.
Microbiology 155, 733–740 (2009). Microbiol. 77, 1946–1956 (2011).
60. Semenova, E. et al. Interference by clustered regularly interspaced short 74. Cady, K. C. & O’Toole, G. A. Non-identity targeting of yersinia-subtype CRISPR-
palindromic repeat (CRISPR) RNA is governed by a seed sequence. Proc. Natl prophage interaction requires the Csy and Cas3 proteins. J. Bacteriol. 193,
Acad. Sci. USA 108, 10098–10103 (2011). 3433–3445 (2011).
61. Tang, T. H. et al. Identification of 86 candidates for small non-messenger 75. Zegans, M. E. et al. Interaction between bacteriophage DMS3 and host CRISPR
RNAs from the archaeon Archaeoglobus fulgidus. Proc. Natl Acad. Sci. USA 99, region inhibits group behaviors of Pseudomonas aeruginosa. J. Bacteriol. 191,
7536–7541 (2002). 210–219 (2009).
62. Tang, T. H. et al. Identification of novel non-coding RNAs as potential antisense 76. Obbard, D. J., Gordon, K. H. J., Buck, A. H. & Jiggins, F. M. The evolution of RNAi
regulators in the archaeon Sulfolobus solfataricus. Mol. Microbiol. 55, 469–481 as a defence against viruses and transposable elements. Philos. Trans. R. Soc. B
(2005). Biol. Sci. 364, 99–115 (2009).
63. Ebihara, A. et al. Crystal structure of hypothetical protein TTHB192 from 77. Aravin, A. A., Hannon, G. J. & Brennecke, J. The Piwi-piRNA pathway provides an
Thermus thermophilus HB8 reveals a new protein family with an RNA adaptive defense in the transposon arms race. Science 318, 761–764 (2007).
recognition motif-like domain. Protein Sci. 15, 1494–1499 (2006). 78. Aravin, A. A. et al. Double-stranded RNA-mediated silencing of genomic tandem
64. Carte, J., Pfister, N. T., Compton, M. M., Terns, R. M. & Terns, M. P. Binding and repeats and transposable elements in the D-melanogaster germline. Curr Biol
cleavage of CRISPR RNA by Cas6. RNA 16, 2181–2188 (2010). 11, 1017–1027 (2001).
65. Gudbergsdottir, S. et al. Dynamic properties of the Sulfolobus CRISPR/Cas and 79. Bartel, D. P. MicroRNAs: target recognition and regulatory functions. Cell 136,
CRISPR/Cmr systems when challenged with vector-borne viral and plasmid 215–233 (2009).
genes and protospacers. Mol. Microbiol. 79, 35–49 (2011) 80. Guo, H., Ingolia, N. T., Weissman, J. S. & Bartel, D. P. Mammalian microRNAs
66. Manica, A., Zebec, Z., Teichmann, D. & Schleper, C. In vivo activity of CRISPR- predominantly act to decrease target mRNA levels. Nature 466, 835–840
mediated virus defence in a hyperthermophilic archaeon. Mol. Microbiol. 80, (2010).
481–491 (2011).
67. Marraffini, L. A. & Sontheimer, E. J. Self versus non-self discrimination during Acknowledgements B.W. is a Howard Hughes Medical Institute Fellow of the Life
CRISPR RNA-directed immunity. Nature 463, 568–571 (2010). Sciences Research Foundation. S.H.S. acknowledges support from the National
This study identified a mechanism for distingishing self from non-self, Science Foundation and National Defense Science & Engineering Graduate
which relies on base-pairing potential in regions outside the crRNA Research Fellowship programs. J.A.D. is an Investigator of the Howard Hughes
spacer sequence. Medical Institute.
68. Bartel, D. P. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell
116, 281–297 (2004). Author Information Reprints and permissions information is available at
69. Parker, J. S., Parizotto, E. A., Wang, M., Roe, S. M. & Barford, D. Enhancement www.nature.com/reprints. The authors declare no competing financial
of the seed-target recognition step in RNA silencing by a PIWI/MID domain interests. Readers are welcome to comment on the online version of this
protein. Mol. Cell 33, 204–214 (2009). article at www.nature.com/nature. Correspondence should be addressed to
70. Beloglazova, N. et al. Structure and activity of the Cas3 HD nuclease MJ0384, an J.A.D. (doudna@berkeley.edu).

3 3 8 | N AT U R E | VO L 4 8 2 | 1 6 F E B R UA RY 2 0 1 2
© 2012 Macmillan Publishers Limited. All rights reserved
Prospects & Overviews

Review essays
The phage-host arms race:
Shaping the evolution of microbes
Adi Stern and Rotem Sorek

Bacteria, the most abundant organisms on the planet, Introduction


are outnumbered by a factor of 10 to 1 by phages that
Phage-host relationships have been studied intensively since
infect them. Faced with the rapid evolution and turnover
the early days of molecular biology. In the late 1970s, while
of phage particles, bacteria have evolved various mech- viruses were found to be ubiquitous, it was assumed that they
anisms to evade phage infection and killing, leading to were present in relatively low numbers and that their effect on
an evolutionary arms race. The extensive co-evolution of microbial communities was low [1]. With the increasing avail-
both phage and host has resulted in considerable diver- ability of new molecular techniques that allow studies of
microbial communities without the need to culture them
sity on the part of both bacterial and phage defensive
[2], it is now realized that viruses greatly outnumber bacteria
and offensive strategies. Here, we discuss the unique in the ocean and other environments, with viral numbers
and common features of phage resistance mechanisms (107–108/mL) often 10-fold larger than bacterial cell counts
and their role in global biodiversity. The commonalities (106/mL) [3–5]. Thus, bacteria are confronted with a constant
between defense mechanisms suggest avenues for the threat of phage predation.
discovery of novel forms of these mechanisms based on The Red Queen hypothesis (Box 1) posits that competitive
environmental interactions, such as those displayed by hosts
their evolutionary traits.

.
and parasites, will lead to continuous variation and selection
towards adaptation of the host, and counter-adaptations on
Keywords: the side of the parasite. Arguably, nowhere is this evolutionary
arms race; bacteria; evolution; phage; resistance trend so pronounced as in phage-microbe interactions. This is
due to the extremely rapid evolution and turnover of phage
particles [6], causing acute pressure on microbial communities
to evade infection and killing by phages. In fact, the arms race
between phages and bacteria is predicted to have had an
impact on global nutrient cycling [7], on global climate
[6, 7], on the evolution of the biosphere [8], and also on the
evolution of virulence in human pathogens [9].
This review focuses on the evolution of three of the most
well-studied microbial defense mechanisms against phages:
the restriction-modification (RM) system, the recently discov-
ered ‘‘clustered regularly interspersed palindromic repeats’’
(CRISPR) loci together with CRISPR-associated (cas) genes,
and the abortive infection (Abi) system (summarized in
DOI 10.1002/bies.201000071
Table 1). We first describe these defense systems, and the
counter-adaptations that evolved in the phage to allow escape
Department of Molecular Genetics, Weizmann Institute of Science,
Rehovot, Israel from bacterial defense. Next, we discuss features that are
*Corresponding author:
common to many microbial defense systems, such as rapid
Rotem Sorek evolution, tendency for lateral gene transfer (LGT), and the
E-mail: rotem.sorek@weizmann.ac.il selfish nature of these systems. We elaborate on defense
Abbreviations: systems that have gained new functions in the host genome.
Abi, abortive infection; Cas, CRISPR-associated sequence; CRISPR, Finally, the exciting hypothesis that many other prokaryotic
clustered regularly interspersed palindromic repeats; crRNA, CRISPR RNA;
LGT, lateral gene transfer; MTase, methyltransferase; REase, restriction defense systems are still to be discovered is discussed. Our
endonuclease; RM, restriction modification; TA, toxin-antitoxin. review mainly focuses on the evolutionary angle of the phage-

Bioessays 33: 43–51,ß 2010 WILEY Periodicals, Inc. www.bioessays-journal.com 43


A. Stern and R. Sorek Prospects & Overviews ....
material and another that protects host genetic material from
Box 1 restriction (Fig. 1A). Both activities are mediated by recog-
nition of a specific DNA sequence, on average 4–8 bp long.
The Red Queen hypothesis
Review essays

Protection is normally conferred by modification (usually


methylation) of specific bases in this recognition sequence
The Red Queen hypothesis was originally proposed by
in the host genome. Accordingly, all non-methylated DNA
Van Valen [10], and is also termed the evolutionary arms
recognition sequences are recognized as foreign and are
race hypothesis. As the Red Queen tells Alice in Lewis
cleaved. Genetically, the minimal composition of RM systems
Carroll’s Through the Looking-Glass: ‘‘Now, here, you
consists of a methyltransferase (MTase) gene that performs the
see, it takes all the running you can do, to keep in the
defense activity and a restriction endonuclease (REase) gene
same place. If you want to get somewhere else, you must
that performs the foreign restriction activity. Since the REases
run at least twice as fast as that!’’ The original theory
undergo rapid evolution, it is often the presence of an MTase
proposed that in tight co-evolutionary interactions, such
that serves as the basis for identification of RM systems in
as those in a prey-predator relationship, changes (e.g.,
newly sequenced genomes [14].
running faster) on the one side may lead to near extinc-
Phages have evolved to evade the ubiquitous RM systems
tion of the other side. The only way the second side can
in a variety of ways (Fig. 1B). Some phages have acquired an
maintain its fitness is by counter-adaptation (running
MTase, or stimulate the host MTase so that it confers protec-
even faster). This will lead to an uneasy balance between
tion to the phage genome [15]. Other phages code for proteins
prey and predator, where species have to constantly
that target and shut down the REase. One interesting example
evolve to stay at the same fitness level. The metaphor
is the Ocr protein of phage T7, which blocks the active site of
of an evolutionary arms race has been found to be
some REases by mimicking 24 bp of bent B-form DNA [16].
relevant for many biological processes, but nowhere is
Alternatively, some phages incorporate unusual bases in their
this metaphor as apt as in host-parasite relationships.
genomes, thus throwing off the REase. For example, some
Bacillus subtilis phages replace thymine with 5-hydroxyme-
thyluracil [15]. These phages code a protein that then further
inhibits the host protein uracil-DNA glycosylase from cleaving
host arms race; for deeper mechanistic descriptions of phage uracil bases from the phage DNA [17, 18]. Interestingly, this
resistance systems we refer the readers to an excellent recent inhibitory protein is also a DNA mimic (reviewed in ref. 19).
review [11]. In addition, the discussion here is restricted to The T-even phages T2, T4, and T6 also contain unusual bases
active defense mechanisms; passive host adaptations, such as in their genomes and may further post-synthetically glycosy-
mutations at the phage receptors, are not discussed. late their DNA to avoid REase restriction [15]. Yet another
evasion mechanism that has been reported is alteration of
the restriction recognition sites. For example, some phages
Restriction-modification systems: employ ‘‘palindrome avoidance’’: since Type II REases often
Degradation of foreign DNA recognize symmetrical (palindromic) sequences, some phages
tend to avoid containing such sites in their genome [20].
Arguably, the most well-studied phage defense mechanism is The well-studied model organisms Escherichia coli and T4
the RM system [12], which is present in over 90% of sequenced phage display a fascinating example of a co-evolutionary arms
bacterial and archaeal genomes [13]. This system is composed race [11]. The battle purportedly begins with the T4 phage genome,
of two activities: one that restricts incoming foreign genetic which contains the modified base hydroxymethylcytosine instead

Table 1. A summary of the major defense mechanisms described in this article

Name Mechanism Phylogenetic breadth


Restriction-modification The restriction enzyme cleaves specific patterns Appear in 90% of all sequenced
in the incoming foreign DNA, while the modification prokaryotic genomes [13]
enzyme protects host DNA from cleavage by
unique biochemical modification
CRISPR/Cas Fragments of phage DNA are integrated into Appear in 40% and 90% of all
CRISPR loci, which are then transcribed and sequenced bacterial and archaeal
processed into short non-coding RNAs. These genomes, respectively [27]. The
RNAs, along with the associated Cas proteins, distribution of CRISPR/Cas-bearing
guide the way to interfere with the phage nucleic species across the phylogenetic tree
acids, in an as-yet-unknown mechanism of life is highly patchy [38, 59]
Abortive infection Premature cellular death occurs upon phage entry, Currently known Abi systems display
blocking the expansion of the phage to neighboring a sporadic phylogenetic distribution in
cells. Notably, Abi systems include a large collection Gamma-proteobacteria and in Firmicutes [44],
of mechanisms with little or no known evolutionary yet recent reports suggest that some
relationship, apart from a very similar phenotype systems may have an even broader
phylogenetic range [54, 111]

44 Bioessays 33: 43–51,ß 2010 WILEY Periodicals, Inc.


.... Prospects & Overviews A. Stern and R. Sorek

CRISPR/Cas: Acquired adaptive immunity


The existence of a unique nucleotide arrangement in E. coli,

Review essays
comprised of a cluster of direct repeats interspersed with
variable sequences (termed spacers; both around 30 bp long),
was first described in 1987 [25]. These clusters, together with
associated cas genes (Fig. 2A), were later found to exist in
40% of sequenced bacterial genomes and 90% of archaeal
genomes [26–28]. The first inkling that this system comprises a
phage-defense mechanism arose when spacer sequences were
found to be highly similar to DNA from foreign origin, i.e. from
phage, plasmid or transposon DNA [29–31]. In 2007, exper-
imental evidence was presented that this system indeed con-
fers resistance to phage infection [32]. This led to the striking
insight that similar to vertebrates, prokaryotes possess
acquired (and inherited) immunity [27, 33–35]. Following
phage infection, it has been found that a small portion of
bacterial cells integrated new spacers identical to the phage
genomic sequence (termed proto-spacer), resulting in CRISPR-
mediated phage resistance [32, 36, 37]. Further experiments
have shown that the CRISPR locus is transcribed into a single
RNA transcript, which is then further cleaved by the Cas
proteins to generate smaller CRISPR RNA (crRNA) units, each
including one targeting spacer [37, 38]. These units then
interfere with the incoming foreign genetic material by comp-
lementary base-pairing with either DNA [36, 37] or RNA [39]
from the foreign element (Fig. 2A).
While the study of CRISPR/Cas is still in its infancy,
examples of phages that are resistant to CRISPR/Cas interfer-
ence have nevertheless been noted (Fig. 2B). Following the
first round of infection and acquisition of novel spacers by the
bacterial CRISPR, phages that have mutated, recombined or
lost their proto-spacer target sequence in the second round of
infection are then resistant to CRISPR [32, 40–42]. It also seems
likely that phages have evolved mechanisms that directly
Figure 1. Restriction-modification (RM) defense system. A: A general target the CRISPR/Cas machinery. To date, only vague hints
illustration of function, exemplified by type II RM enzymes. B: of such mechanisms exist. For example, it has been shown
Examples of strategies employed by phages to evade restriction: that one of the proteins encoded by the T7 phage phosphor-
(1) incorporation of unusual bases protects from restriction [15]; ylates the CasB protein [43]. It remains to be shown whether
(2) masking of the restriction sites by phage proteins [112]; (3) stimu-
this feat of the phage affects CRISPR/Cas functioning.
lation of MTase activity causes the phage DNA to be protected;
(4) neutralization of REase by phage proteins that mimic DNA [113].
Abortive infection: Cellular suicide
of cytosine [15]. To counter-attack the phage, E. coli K-12 If a phage has successfully entered the host cell and avoided
possesses a unique form of REase, the McrBC enzyme, which restriction by the host RM systems and by CRISPR, it proceeds to
cleaves only modified DNA substrates (such as that of T4) [21]. develop, replicate, and release its progeny. Abi is a collective
The T4 phage rises to the challenge by glucosylating its term describing host mechanisms that interrupt with phage
genome, and is thus impervious to McrBC [15]. However, development at different stages of phage transcription, genome
E. coli cT596 encodes the GmrS-GmrD system that can also replication, and phage packaging [44]. Abi-mediated resistance
restrict glucosylated DNA [22, 23]. In continuation of the leads to death (suicide) of the cell, and is thought to occur since
battle, some T4 phages encode the IPI protein, which in its corruption of host functions has already been initiated by the
processed form (IPI) disables the GmrS-GmrD system [24]. phage. However, this death confers an advantage to surround-
Evidently, the battle is far from an end, since bacterial strains ing bacterial cells since it confines the infection to the sacrificed
have been found to overcome the IPI protein of T4 [24]. In cell and prevents the spread of infectious particles.
fact, it is likely that many bacterial defense systems, Although several Abi systems have been discovered, with
coupled with their cognate phage evasion strategies, have the majority of them encoded on plasmids, the mechanism by
undergone similar attack and counter-attack cycles, where which they operate remains largely unknown. Frequently, Abi
a change on one side selects for changes that can overcome is mediated by a single gene encoded on a plasmid or on a
the opponent. prophage, which displays little or no homology to any known

Bioessays 33: 43–51,ß 2010 WILEY Periodicals, Inc. 45


A. Stern and R. Sorek Prospects & Overviews ....
A) Repeats
cas genes
CRISPR
Review essays

locus
Spacers
transcription

CRISPR
pre-crRNA
Cas processing
proteins
Processed,
mature crRNAs

B)

?
P

Figure 2. The CRISPR/Cas system. A: Mechanism of action: tran- Since Abi genes have a toxic effect on their host, they are
scription from the repeat-spacer CRISPR locus generates a long under tight regulation [50]. In fact, many similarities can be
non-coding RNA, with repeats that may sometimes assume a sec- drawn between Abi systems (and also RM systems) and toxin-
ondary structure. Cleavage of the repeat sequences by the Cas
antitoxin (TA) systems. TA systems are composed of a stable
proteins generates crRNAs that target the phage DNA or RNA, and
interfere with phage infection. B: Phages can evade CRISPR interfer-
toxin and an unstable antitoxin [51]. Normally, the antitoxin
ence by mutation or recombination of the targeted proto-spacer binds and inhibits the toxin. However, a decrease in the levels
sequence. Another putative evasion mechanism is phosphorylation of the unstable antitoxin activates the toxin and leads to
of the Cas proteins. However, this remains to be verified. growth arrest or cell death. It has been shown that this situ-
ation occurs in response to phage infection in two known TA
modules, mazEF and hok-sok, which can thus cause Abi [52,
53]. Moreover, the AbiQ system was recently found to function
proteins. Some Abi genes have been shown to target phage as a protein-RNA TA pair [54], albeit exactly how this system
genes involved in DNA replication [45, 46], and others have interferes with phage replication remains unknown. Thus, it
been shown to target the host translation apparatus [47, 48]. appears that some TA systems may be a subtype of Abi
For instance, in E. coli K-12, the Lit protease encoded by the systems. Interestingly, RM systems may also be viewed as
defective e14 prophage is only activated in the presence of a TA systems, since loss of the MTase (which like the antitoxin
short polypeptide called Gol, which is produced by the T4 is often unstable) results in a toxic effect of the REase [55, 56].
phage. Once active, this protease cleaves the translation
elongation factor Ef-Tu, thus leading to translational arrest
and cell death (Fig. 3A). Similarly, the Prr protein (also part of Commonalities among phage defense
a defective prophage in E. coli cT196) cleaves tRNALys in systems
response to the presence of the T4 peptide Stp [49].
Mutations in these phage peptides suppress activation of One of the most striking features common to all phage defense
Lit or Prr and rescue the infecting phage [47, 49] (Fig. 3B). systems is their high genetic variability, which occurs as a

46 Bioessays 33: 43–51,ß 2010 WILEY Periodicals, Inc.


.... Prospects & Overviews A. Stern and R. Sorek

to counteract the invading phage, thus contributing to the


extensive co-evolution with the prokaryotic parasites.
However, there is an interesting alternative view, not necess-

Review essays
arily mutually exclusive, which may explain the high varia-
bility and mobility of phage defense systems. Accordingly,
these systems are selfish elements, as elaborated in the next
section.

Defense systems: A burden or a benefit?


In this section, the advantages and disadvantages of microbial
immune systems are reviewed. Such a discussion is incom-
plete without examining the pros and cons of the phage-host
relationship. Evidently, for a single cell, phage predation, and
killing of the host are a strong disadvantage. However, phages
may also contribute directly to host fitness by supplying a pool
of new and possibly beneficial genes [73]. As such, phages
serve as vessels of LGT, which is one of the most important
forces in microbial evolution (Box 2).
What is the associated cost of encoding phage defense
mechanisms? An obvious primary cost is the energy cost
linked to carrying additional genetic cargo. The fact that,

Figure 3. The Abi system. A: Illustration of the mechanism of the


E. coli K-12 Abi Lit system. When the T4 phage peptide Gol is syn-
thesized, it binds and activates the bacterial (prophage-encoded) Lit Box 2
protein, which then cleaves the elongation factor EF-Tu. This leads
to the arrest of protein synthesis and to bacterial cell death, with the Role of defense systems in limiting LGT
phage trapped inside. B: A mutation at the Gol polypeptide reduces
the activation of Lit, and rescues the phage. LGT is the process whereby genetic material is incorp-
orated from a non-parental organism. There are three
major mechanisms of LGT: (a) transduction by phages;
consequence of the co-evolutionary arms race with phages (b) transformation of naked DNA; and (c) bacterial con-
[57]. This is manifested in the enormous numbers and types of jugation, which involves transfer of DNA via direct con-
RM systems [12, 58], of CRISPR subtypes [59, 60] and in the nections generated between a pair of cells. All three
variety of Abi systems [44]. Moreover, the gene sequences of mechanisms involve the entry of foreign DNA into the
these systems often display a high evolutionary rate ([61–63]; host cell. LGT is recognized as one of the most important
A. Stern and R. Sorek, unpublished data). This variability is mechanisms of genetic innovation in both bacteria and
most easily exemplified in the diversity of RM systems: four archaea [82–84]. In fact, it is now realized that much of
inherently different groups of RM systems exist, classified as the genomic diversity in prokaryotes is a consequence of
Type I–IV based on their subunit composition, mode of action, LGT, rather than allelic differences at the same loci [85,
and cofactor requirement (reviewed in ref. 12). Each type of RM 86]. However, when viewed from the point of view of a
system includes an assortment of restriction enzymes that single cell, LGT has a negligible chance of contributing to
recognize different recognition sequences. For instance, the fitness of the organism. Evidently, transduction by
nearly 4000 Type II enzymes are known of today, which lytic phages may severely compromise the survival of a
are further divided into 11 overlapping subclasses [13, 64]. bacterial colony. Plasmids and transposons may also
Interestingly, Type II REase sequences have frequently been decrease the fitness of an organism by integrating into
found to be ORFan sequences, i.e. have no significant sim- crucial regions of the genomes, by adding an energetic
ilarity to any other protein [65, 66]. Initially, this was seen as cost involved with replication of excess DNA, and by
evidence for convergent evolution [67]. However, crystallo- expressing harmful genes (e.g. refs. 87–89). Finally, even
graphic studies have shown structural similarity among if a new beneficial gene sequence is obtained, there is
Type II REases, most likely representing a rapid evolutionary only a small probability that it will integrate into the
rate since their divergence from a common ancestor [68]. current cellular network of an organism without causing
Another trait common to all defense systems presented deleterious effects [90, 91]. Thus, defense mechanisms
here is their propensity to undergo LGT, sometimes between such as those described here are crucial for maintaining
distantly related prokaryotes [69–72]. Often these systems the genetic identity of organisms, and protecting it
reside on plasmids, on prophages, or on genomic islands of against the constant bombardment of potentially detri-
foreign origin, or are linked to transposase genes. This mobi- mental foreign DNA.
lity allows rapid acquisition and dissemination of new systems

Bioessays 33: 43–51,ß 2010 WILEY Periodicals, Inc. 47


A. Stern and R. Sorek Prospects & Overviews ....
often, only a small portion of a given bacterial population such as mismatch repair by the MutHLS complex, binding
bears a plasmid or a prophage with a given defense mechan- of the replication-initiation complex to methylated OriC, and
ism may be seen as evidence for this cost. A second intriguing regulation of bacterial pathogenicity [93]. On the other hand,
Review essays

cost of encoding a phage defense mechanism is the risk of the CcrM methylase has been shown to affect the cell cycle in
autoimmunity. Autoimmunity, classically defined in mamma- alpha proteobacteria that encode this gene [93].
lian immune systems, is the failure of the immune system to Another interesting example of exaptation of RM systems
recognize what is self and what is foreign, resulting in an is evident in phase-variable Type III RM systems [94], whose
immune response against self. In RM systems, this is a clear genes can be reversibly inactivated due to tandem repeat tracts
danger since the restriction enzyme is often more stable than in their sequences. These repeats initiate a mechanism called
the protecting methylase [74]. Abi mechanisms are equally slipped-strand mispairing, leading to a change in the number
lethal, since errant function of the Abi gene will lead to cell of repeats after DNA replication and possible frame-shift
death. Finally, the CRISPR system is also not ‘‘immune’’ to mutations [94]. Several regulatory roles have been suggested
errors, and the frequent acquisition of self genetic material for phase-variable RM systems: (a) to allow regulated removal
also leads to spacers that target the self-genome, potentially of the barrier against foreign DNA, thus allowing potentially
incurring autoimmunity ([75], see also ref. 76). beneficial uptake of DNA [95, 96], (b) autolytic self-DNA
Paradoxically, phages themselves often bear anti-phage degradation or ‘‘bacterial suicide’’ [97–99] (discussed further
defense systems, enabling their acquisition by host cells. To below), and (c) epigenetic gene regulation via differential meth-
cite a few examples, the HindIII RM gene complex was found ylation of the genome [100]. The latter phenomenon, which
on a cryptic prophage in the Haemophilus influenzae genome allows switching different genes on and off, has been linked
[77], the abiN Abi gene is encoded on a prophage in to pathogenicity of bacterial species by allowing colonization,
Lactococcus lactis subsp. cremoris S114, and even a CRISPR immune evasion, and adaptation to novel environments [94].
array was found within a Clostridium difficile prophage [78]. TA modules, which include some Abi systems, are also
These results initially seem counterintuitive, since what known to participate in a variety of other cellular processes.
benefit is there for the phage to carry such systems? One For example, one of the most studied TA loci, mazEF, aborts
possible explanation is that this allows superinfection exclu- translation by cleaving mRNA molecules in response to differ-
sion, thus preventing other phages from infecting an already ent stress signals [101, 102], one of which is phage infection
infected cell [11, 78, 79]. However, this phenomenon has also [52]. An ongoing debate exists whether this action is reversible
been viewed as evidence for the selfish nature of phage or not: reversible effects have been attributed to bacteriostatic
defense mechanisms. effects, which allow reduced growth rate of each cell during
The behavior of defense mechanisms as selfish mobile nutritional stress [101, 102]. On the other hand, irreversible
elements has been extensively discussed for the case of RM effects of mazEF have been attributed to programmed cell
systems [55, 80], but is also applicable for many Abi systems death that occurs in a subpopulation of cells, permitting
that operate as TA systems, which also have ‘‘selfish’’ proper- the survival of the population of a whole [103].
ties [51, 81]. The main lines of evidence in favor of this view is Interestingly, in Myxococcus xanthus it was shown that the
that (a) RM systems destroy any other invading RM system, toxin MazF exists without the antitoxin MazE, and has
(b) any attempt to lose the RM system will result in the death of adopted a key transcriptional regulator as an alternative anti-
the host, and (c) RM systems are prone to extensive mobility, toxin [104]. MazF mediates programmed cell death during
and are often associated with plasmids, phages, transposons, multicellular development of this organism. To summarize,
and integrons [55]. These characteristics of the RM systems these accounts exemplify the broad evolutionary diversifica-
lead to an increase of their relative frequency in the bacterial tion of different microbial defense mechanisms, and their
population. Thus, according to this hypothesis, the defense potential to cross boundaries from phage-encoded mechan-
incurred by RM systems on host cells is a mere by-product of isms, to anti-phage systems, to regulatory host mechanisms.
the fact that RM systems defend themselves.

Conclusions
Exaptations: Alternative functions of
defense systems Despite our growing understanding of microbial immunity,
much still remains obscure. Do defense systems work separ-
Intriguingly, a number of anti-phage defense systems have ately or in unison? What is the cost of each system? Which
evolved to gain a distinct function in cellular regulation that is phages are targeted by which systems? For instance, while
independent of phage restriction. Such evolutionary events most characterized defense systems work against double-
were coined ‘‘exaptations’’ [92], a term used to describe the stranded DNA phages, RNA viruses might also be abundant
use of a biological structure or function for a purpose other [105]. Defense systems that target such viruses are yet to be
than that for which it initially evolved. For instance, several discovered.
RM systems have lost their REase activity, leaving an orphan The recently discovered CRISPR system epitomizes our
MTase that can now take part in epigenetic modifications. The incomplete understanding of the complexity of bacterial
Dam methylase in E. coli and the CcrM methylase in defense systems. The discovery that almost half of all prokar-
Caulobacter crescentus, both of which originated from RM yotes possess acquired inherited immunity came as a surprise
systems [93], are two such examples. Methylation by Dam to the scientific community, given our initial tendency to view
has been linked to several important regulatory processes, prokaryotes as less complex organisms. However, since it is

48 Bioessays 33: 43–51,ß 2010 WILEY Periodicals, Inc.


.... Prospects & Overviews A. Stern and R. Sorek

now realized that prokaryotes are faced with a constant threat 4. Chibani-Chennoufi S, Bruttin A, Dillmann ML, Brussow H. 2004.
Phage-host interaction: an ecological perspective. J Bacteriol 186:
of predation, it is becoming clearer that the microbial immune 3677.
system must be highly complex. Nevertheless, the CRISPR 5. Wommack KE, Colwell RR. 2000. Virioplankton: viruses in aquatic

Review essays
system and the Abi systems have a highly sporadic distri- ecosystems. Microbiol Mol Biol Rev 64: 69.
6. Fuhrman JA. 1999. Marine viruses and their biogeochemical and eco-
bution across the prokaryotic phylogeny (Table 1). logical effects. Nature 399: 541–8.
Furthermore, while RM systems are present in 90% of pro- 7. Suttle CA. 2007. Marine viruses – major players in the global ecosystem.
karyotic species [13], this system is far from infallible, since it Nat Rev Microbiol 5: 801–12.
has been shown that phages have a non-negligible probability 8. Comeau AM, Krisch HM. 2005. War is peace – dispatches from the
bacterial and phage killing fields. Curr Opin Microbiol 8: 488–94.
of escaping the REase and being methylated by the MTase. 9. Brussow H, Canchaya C, Hardt WD. 2004. Phages and the evolution of
[106]. Thus, all three mechanisms reviewed here are either bacterial pathogens: from genomic rearrangements to lysogenic con-
somewhat leaky, or span only part of the phylogenetic breadth version. Microbiol Mol Biol Rev 68: 560–602.
10. Van Valen L. 1973. A new evolutionary law. Evol Theor 1: 1–30.
of bacteria and archaea. Given the intensive arms race 11. Labrie SJ, Samson JE, Moineau S. 2010. Bacteriophage resistance
between bacteria and phages, it seems probable that there mechanisms. Nat Rev Microbiol 8: 317–27.
are other yet unknown defense mechanisms waiting to be 12. Tock MR, Dryden DT. 2005. The biology of restriction and anti-restric-
tion. Curr Opin Microbiol 8: 466–72.
revealed.
13. Roberts RJ, Vincze T, Posfai J, Macelis D. 2010. REBASE – a data-
So how does one set about the search for novel prokaryotic base for DNA restriction and modification: enzymes, genes and
defense systems, and how does one learn more about existing genomes. Nucleic Acids Res 38: D234–6.
ones? It is likely that many solutions to these questions will be 14. Roberts RJ, Vincze T, Posfai J, Macelis D. 2005. REBASE – restriction
enzymes and DNA methyltransferases. Nucleic Acids Res 33:
made possible, thanks to advances in sequencing technologies D230.
such as whole-transcriptome studies and metagenomics. For 15. Kruger DH, Bickle TA. 1983. Bacteriophage survival: multiple mech-
instance, a seminal work by Andersson and Banfield [42] anisms for avoiding the deoxyribonucleic acid restriction systems of their
hosts. Microbiol Rev 47: 345–60.
reconstructed viral and host population genomes from com- 16. Bandyopadhyay PK, Studier FW, Hamilton DL, Yuan R. 1985.
munity genomic data, and showed how CRISPR spacers cor- Inhibition of the type I restriction-modification enzymes EcoB and
relate with the location of coexisting viruses and hosts. In EcoK by the gene 0.3 protein of bacteriophage T7. J Mol Biol 182:
addition, controlled experimental evolution studies, such as 567–78.
17. Wang Z, Mosbaugh DW. 1989. Uracil-DNA glycosylase inhibitor gene
those performed in Pseudomonas fluorescens and its phage, of bacteriophage PBS2 encodes a binding protein specific for uracil-
enable a direct survey of the changes that occur in the phage DNA glycosylase. J Biol Chem 264: 1163–71.
and host population with and without infection [107–109]. 18. Wang Z, Mosbaugh DW. 1988. Uracil-DNA glycosylase inhibitor of
bacteriophage PBS2: cloning and effects of expression of the inhibitor
These studies are also likely to shed light on current defense gene in Escherichia coli. J Bacteriol 170: 1082–91.
systems and possibly discover novel ones. 19. Putnam CD, Tainer JA. 2005. Protein mimicry of DNA and pathway
More specifically, the search for novel defense mechan- regulation. DNA Repair (Amst) 4: 1410–20.
20. Rocha EP, Danchin A, Viari A. 2001. Evolutionary role of restriction/
isms may be based on the well-established common properties
modification systems as revealed by comparative genome analysis.
of both eukaryotic and prokaryotic immune systems: their Genome Res 11: 946–58.
high rate of evolution, and their tendency to undergo LGT. 21. Raleigh EA, Wilson G. 1986. Escherichia coli K-12 restricts DNA con-
Thus, it may be that a reservoir of novel defense mechanisms taining 5-methylcytosine. Proc Natl Acad Sci USA 83: 9070–4.
22. Bair CL, Rifat D, Black LW. 2007. Exclusion of glucosyl-hydroxyme-
lies in the most variable regions of bacterial and archaeal thylcytosine DNA containing bacteriophages is overcome by the injected
genomes, known as genomic islands [110]. A strategy that protein inhibitor IPI. J Mol Biol 366: 779–89.
focuses on such islands to search for novel phage resistance 23. Bair CL, Black LW. 2007. A type IV modification dependent restriction
nuclease that targets glucosylated hydroxymethyl cytosine modified
mechanisms might lead, in the future, to surprising
DNAs. J Mol Biol 366: 768–78.
discoveries. 24. Rifat D, Wright NT, Varney KM, Weber DJ, et al. 2008. Restriction
endonuclease inhibitor IPI of bacteriophage T4: a novel structure for a
dedicated target. J Mol Biol 375: 720–34.
Acknowledgments 25. Ishino Y, Shinagawa H, Makino K, Amemura M, et al. 1987. Nucleotide
sequence of the iap gene, responsible for alkaline phosphatase isozyme
R.S. is an EMBO Young Investigator. He was supported, in conversion in Escherichia coli, and identification of the gene product.
part, by the ISF-FIRST program (grant 1615/09), NIH J Bacteriol 169: 5429.
R01AI082376-01, ERC-StG, the Wolfson Family Trust miRNA 26. Jansen R, van Embden JDA, Gaastra W, Schouls LM. 2002.
Identification of genes that are associated with DNA repeats in prokar-
research program, the Minerva foundation, and the Yeda-Sela
yotes. Mol Microbiol 43: 1565–75.
Center for basic research. A.S. was supported by the Clore 27. Sorek R, Kunin V, Hugenholtz P. 2008. CRISPR – a widespread system
post-doctoral fellowship. that provides acquired resistance against phages in bacteria and arch-
aea. Nat Rev Microbiol 6: 181–6.
28. Grissa I, Vergnaud G, Pourcel C. 2007. The CRISPRdb database and
tools to display CRISPRs and to generate dictionaries of spacers and
References repeats. BMC Bioinformatics 8: 172.
29. Bolotin A, Quinquis B, Sorokin A, Ehrlich SD. 2005. Clustered regularly
1. Torrella F, Morita RY. 1979. Evidence by electron micrographs for a interspaced short palindrome repeats (CRISPRs) have spacers of
high incidence of bacteriophage particles in the waters of Yaquina Bay, extrachromosomal origin. Microbiology 151: 2551–61.
Oregon: ecological and taxonomical implications. Appl Environ 30. Mojica FJ, Diez-Villasenor C, Garcia-Martinez J, Soria E. 2005.
Microbiol 37: 774–8. Intervening sequences of regularly spaced prokaryotic repeats derive
2. Riesenfeld CS, Schloss PD, Handelsman J. 2004. Metagenomics: from foreign genetic elements. J Mol Evol 60: 174–82.
genomic analysis of microbial communities. Annu Rev Genet 38: 525– 31. Pourcel C, Salvignol G, Vergnaud G. 2005. CRISPR elements in
52. Yersinia pestis acquire new repeats by preferential uptake of bacterio-
3. Bergh O, Borsheim KY, Bratbak G, Heldal M. 1989. High abundance of phage DNA, and provide additional tools for evolutionary studies.
viruses found in aquatic environments. Nature 340: 467–8. Microbiology 151: 653–63.

Bioessays 33: 43–51,ß 2010 WILEY Periodicals, Inc. 49


A. Stern and R. Sorek Prospects & Overviews ....
32. Barrangou R, Fremaux C, Deveau H, Richards M, et al. 2007. CRISPR 59. Haft DH, Selengut J, Mongodin EF, Nelson KE. 2005. A guild of 45
provides acquired resistance against viruses in prokaryotes. Science CRISPR-associated (Cas) protein families and multiple CRISPR/Cas
315: 1709–12. subtypes exist in prokaryotic genomes. PLoS Comput Biol 1: e60.
33. Horvath P, Barrangou R. 2010. CRISPR/Cas, the immune system of 60. Kunin V, Sorek R, Hugenholtz P. 2007. Evolutionary conservation of
Review essays

bacteria and archaea. Science 327: 167–70. sequence and secondary structures in CRISPR repeats. Genome Biol 8:
34. Marraffini LA, Sontheimer EJ. 2010. CRISPR interference: RNA- R61.
directed adaptive immunity in bacteria and archaea. Nat Rev Genet 61. Sharp PM, Kelleher JE, Daniel AS, Cowan GM, et al. 1992. Roles of
11: 181–90. selection and recombination in the evolution of type I restriction-modi-
35. van der Oost J, Jore MM, Westra ER, Lundgren M, et al. 2009. fication systems in enterobacteria. Proc Natl Acad Sci USA 89: 9836–40.
CRISPR-based adaptive and heritable immunity in prokaryotes. 62. Murray NE, Daniel AS, Cowan GM, Sharp PM. 1993. Conservation of
Trends Biochem Sci 34: 401–7. motifs within the unusually variable polypeptide sequences of type I
36. Marraffini LA, Sontheimer EJ. 2008. CRISPR interference limits hori- restriction and modification enzymes. Mol Microbiol 9: 133–43.
zontal gene transfer in staphylococci by targeting DNA. Science 322: 63. Zheng Y, Roberts RJ, Kasif S. 2004. Identification of genes with fast-
1843. evolving regions in microbial genomes. Nucleic Acids Res 32: 6347–57.
37. Brouns SJ, Jore MM, Lundgren M, Westra ER, et al. 2008. Small 64. Roberts RJ, Belfort M, Bestor T, Bhagwat AS, et al. 2003.
CRISPR RNAs guide antiviral defense in prokaryotes. Science 321: 960– A nomenclature for restriction enzymes, DNA methyltransferases, hom-
4. ing endonucleases and their genes. Nucleic Acids Res 31: 1805–12.
38. Makarova KS, Grishin NV, Shabalina SA, Wolf YI, et al. 2006. 65. Kroger M, Hobom G, Schutte H, Mayer H. 1984. Eight new restriction
A putative RNA-interference-based immune system in prokaryotes: endonucleases from Herpetosiphon giganteus-divergent evolution in a
computational analysis of the predicted enzymatic machinery, functional family of enzymes. Nucleic Acids Res 12: 3127.
analogies with eukaryotic RNAi, and hypothetical mechanisms of action. 66. Mullings R, Bennett SP, Brown NL. 1988. Investigation of sequence
Biol Direct 1: 7. homology in a group of type-II restriction/modification isoschizomers.
39. Hale CR, Zhao P, Olson S, Duff MO, et al. 2009. RNA-guided Gene 74: 245–51.
RNA cleavage by a CRISPR RNA-Cas protein complex. Cell 139: 67. Wilson G, Murray NE. 1991. Restriction and modification systems.
945–56. Annu Rev Genet 25: 585–627.
40. Deveau H, Barrangou R, Garneau JE, Labonte J, et al. 2008. Phage 68. Venclovas C, Timinskas A, Siksnys V. 1994. Five-stranded beta-sheet
response to CRISPR-encoded resistance in Streptococcus thermophi- sandwiched with two alpha-helices: a structural link between restriction
lus. J Bacteriol 190: 1390–400. endonucleases EcoRI and EcoRV. Proteins 20: 279–82.
41. Heidelberg JF, Nelson WC, Schoenfeld T, Bhaya D. 2009. Germ 69. Nelson KE, Clayton RA, Gill SR, Gwinn ML, et al. 1999. Evidence for
warfare in a microbial mat community: CRISPRs provide insights into lateral gene transfer between Archaea and bacteria from genome
the co-evolution of host and viral genomes. PLoS One 4: e4169. sequence of Thermotoga maritima. Nature 399: 323–9.
42. Andersson AF, Banfield JF. 2008. Virus population dynamics and 70. Jeltsch A, Pingoud A. 1996. Horizontal gene transfer contributes to the
acquired virus resistance in natural microbial communities. Science wide distribution and evolution of type II restriction-modification sys-
320: 1047–50. tems. J Mol Evol 42: 91–6.
43. Qimron U, Tabor S, Richardson CC. 2010. New details about bac- 71. Haaber J, Moineau S, Hammer K. 2009. Activation and transfer of the
teriophage T7-host interactions. Microbe 5: 117–20. chromosomal phage resistance mechanism AbiV in Lactococcus lactis.
44. Chopin MC, Chopin A, Bidnenko E. 2005. Phage abortive infection in Appl Environ Microbiol 75: 3358–61.
lactococci: variations on a theme. Curr Opin Microbiol 8: 473–9. 72. Godde JS, Bickerton A. 2006. The repetitive DNA elements called
45. Durmaz E, Klaenhammer TR. 2007. Abortive phage resistance mech- CRISPRs and their associated genes: evidence of horizontal transfer
anism AbiZ speeds the lysis clock to cause premature lysis of phage- among prokaryotes. J Mol Evol 62: 718–29.
infected Lactococcus lactis. J Bacteriol 189: 1417. 73. Gogarten JP, Townsend JP. 2005. Horizontal gene transfer, genome
46. Bidnenko E, Ehrlich D, Chopin MC. 1995. Phage operon involved in innovation and evolution. Nat Rev Microbiol 3: 679–87.
sensitivity to the Lactococcus lactis abortive infection mechanism 74. Naito T, Kusano K, Kobayashi I. 1995. Selfish behavior of restriction-
AbiD1. J Bacteriol 177: 3824. modification systems. Science 267: 897–9.
47. Georgiou T, Yu YTN, Ekunwe S, Buttner MJ, et al. Specific peptide- 75. Stern A, Keren L, Wurtzel O, Amitai G, et al. 2010. Self-targeting by
activated proteolytic cleavage of Escherichia coli elongation factor Tu. CRISPR: gene regulation or autoimmunity? Trends Genet 26: 335–40.
1998. Proc Natl Acad Sci USA 95: 2891. 76. Marraffini LA, Sontheimer EJ. 2010. Self versus non-self discrimination
48. Morad I, Chapman-Shimshoni D, Amitsur M, Kaufmann G. 1993. during CRISPR RNA-directed immunity. Nature 463: 568–71.
Functional expression and properties of the tRNA (Lys)-specific core 77. Hendrix RW, Smith MC, Burns RN, Ford ME, et al. 1999. Evolutionary
anticodon nuclease encoded by Escherichia coli prrC. J Biol Chem 268: relationships among diverse bacteriophages and prophages: all the
26842. world’s a phage. Proc Natl Acad Sci USA 96: 2192–7.
49. Snyder L. 1995. Phage-exclusion enzymes: a bonanza of biochemical 78. Sebaihia M, Wren BW, Mullany P, Fairweather NF, et al. 2006. The
and cell biology reagents? Mol Microbiol 15: 415–20. multidrug-resistant human pathogen Clostridium difficile has a highly
50. Blower TR, Fineran PC, Johnson MJ, Toth IK, et al. 2009. Mutagenesis mobile, mosaic genome. Nat Genet 38: 779–86.
and functional characterization of the RNA and protein components of 79. Lu MJ, Henning U. 1994. Superinfection exclusion by T-even-type
the toxIN abortive infection and toxin-antitoxin locus of Erwinia. coliphages. Trends Microbiol 2: 137–9.
J Bacteriol 191, 6029–39. 80. Nakayama Y, Kobayashi I. 1998. Restriction-modification gene com-
51. Gerdes K, Christensen SK, Lobner-Olesen A. 2005. Prokaryotic toxin- plexes as selfish gene entities: roles of a regulatory system in their
antitoxin stress response loci. Nat Rev Microbiol 3: 371–82. establishment, maintenance, and apoptotic mutual exclusion. Proc
52. Hazan R, Engelberg-Kulka H. 2004. Escherichia coli mazEF-mediated Natl Acad Sci USA 95: 6442–7.
cell death as a defense mechanism that inhibits the spread of phage P1. 81. Makarova KS, Wolf YI, Koonin EV. 2009. Comprehensive comparative-
Mol Genet Genomics 272: 227–34. genomic analysis of type 2 toxin-antitoxin systems and related mobile
53. Pecota DC, Wood TK. 1996. Exclusion of T4 phage by the hok/sok killer stress response systems in prokaryotes. Biol Direct 4: 19.
locus from plasmid R1. J Bacteriol 178: 2044. 82. Gogarten JP, Doolittle WF, Lawrence JG. 2002. Prokaryotic evolution
54. Fineran PC, Blower TR, Foulds IJ, Humphreys DP, et al. 2009. The in light of gene transfer. Mol Biol Evol 19: 2226–38.
phage abortive infection system, ToxIN, functions as a protein-RNA 83. Koonin EV, Makarova KS, Aravind L. 2001. Horizontal gene transfer in
toxin-antitoxin pair. Proc Natl Acad Sci USA 106: 894–9. prokaryotes: quantification and classification. Annu Rev Microbiol 55:
55. Kobayashi I. 2001. Behavior of restriction-modification systems as 709–42.
selfish mobile elements and their impact on genome evolution. 84. Dagan T, Artzy-Randrup Y, Martin W. 2008. Modular networks and
Nucleic Acids Res 29: 3742–56. cumulative impact of lateral transfer in prokaryote genome evolution.
56. Hayes F. 2003. Toxins-antitoxins: plasmid maintenance, programmed Proc Natl Acad Sci USA 105: 10039–44.
cell death, and cell cycle arrest. Science 301: 1496–9. 85. Whitaker RJ, Banfield JF. 2006. Population genomics in natural
57. Hoskisson PA, Smith MC. 2007. Hypervariation and phase variation in microbial communities. Trends Ecol Evol 21: 508–16.
the bacteriophage ‘resistome’. Curr Opin Microbiol 10: 396–400. 86. Medini D, Serruto D, Parkhill J, Relman DA, et al. 2008. Microbiology in
58. Orlowski J, Bujnicki JM. 2008. Structural and evolutionary classifi- the post-genomic era. Nat Rev Microbiol 6: 419–30.
cation of Type II restriction enzymes based on theoretical and exper- 87. Modi RI, Adams J. 1991. Coevolution in bacterial-plasmid populations.
imental analyses. Nucleic Acids Res 36: 3552. Evolution 45: 656–67.

50 Bioessays 33: 43–51,ß 2010 WILEY Periodicals, Inc.


.... Prospects & Overviews A. Stern and R. Sorek

88. Zund P, Lebek G. 1980. Generation time-prolonging R plasmids: cor- 101. Pedersen K, Christensen SK, Gerdes K. 2002. Rapid induction and
relation between increases in the generation time of Escherichia coli reversal of a bacteriostatic condition by controlled expression of toxins
caused by R plasmids and their molecular size. Plasmid 3: 65–9. and antitoxins. Mol Microbiol 45: 501–10.
89. Bouma JE, Lenski RE. 1988. Evolution of a bacteria/plasmid associ- 102. Christensen SK, Pedersen K, Hansen FG, Gerdes K. 2003. Toxin-

Review essays
ation. Nature 335: 351–2. antitoxin loci as stress-response-elements: ChpAK/MazF and ChpBK
90. Kedzierska B, Glinkowska M, Iwanicki A, Obuchowski M, et al. 2003. cleave translated RNAs and are counteracted by tmRNA. J Mol Biol 332:
Toxicity of the bacteriophage lambda cII gene product to Escherichia coli 809–19.
arises from inhibition of host cell DNA replication. Virology 313: 622–8. 103. Engelberg-Kulka H, Amitai S, Kolodkin-Gal I, Hazan R. 2006.
91. Lercher MJ, Pal C. 2008. Integration of horizontally transferred genes Bacterial programmed cell death and multicellular behavior in bacteria.
into regulatory interaction networks takes many million years. Mol Biol PLoS Genet 2: e135.
Evol 25: 559–67. 104. Nariya H, Inouye M. 2008. MazF, an mRNA interferase, mediates pro-
92. Brosius J, Gould SJ. 1992. On ‘‘genomenclature’’: a comprehensive grammed cell death during multicellular Myxococcus development. Cell
(and respectful) taxonomy for pseudogenes and other ‘‘junk DNA’’. Proc 132: 55–66.
Natl Acad Sci USA 89: 10706–10. 105. Lang AS, Rise ML, Culley AI, Steward GF. 2009. RNA viruses in the
93. Marinus MG, Casadesus J. 2009. Roles of DNA adenine methylation in sea. FEMS Microbiol Rev 33: 295–323.
host-pathogen interactions: mismatch repair, transcriptional regulation, 106. Korona R, Levin BR. 1993. Phage-mediated selection and the
and more. FEMS Microbiol Rev 33: 488–503. evolution and maintenance of restriction-modification. Evolution 47:
94. Srikhanta YN, Maguire TL, Stacey KJ, Grimmond SM, et al. 2005. The 556–75.
phasevarion: a genetic system controlling coordinated, random switch- 107. Paterson S, Vogwill T, Buckling A, Benmayor R, et al. 2010.
ing of expression of multiple genes. Proc Natl Acad Sci USA 102: 5547– Antagonistic coevolution accelerates molecular evolution. Nature 464:
51. 275–8.
95. Ando T, Xu Q, Torres M, Kusugami K, et al. 2000. Restriction-modi- 108. Buckling A, Wei Y, Massey RC, Brockhurst MA, et al. Antagonistic
fication system differences in Helicobacter pylori are a barrier to inter- coevolution with parasites increases the cost of host deleterious
strain plasmid transfer. Mol Microbiol 37: 1052–65. mutations. Proc Biol Sci 2006. 273: 45–9.
96. Donahue JP, Israel DA, Peek RM, Blaser MJ, et al. 2000. Overcoming 109. Wichman HA, Badgett MR, Scott LA, Boulianne CM, et al. 1999.
the restriction barrier to plasmid transformation of Helicobacter pylori. Different trajectories of parallel evolution during viral adaptation.
Mol Microbiol 37: 1066–74. Science 285: 422–4.
97. Dybvig K, Sitaraman R, French CT. 1998. A family of phase-variable 110. Juhas M, van der Meer JR, Gaillard M, Harding RM, et al. 2009.
restriction enzymes with differing specificities generated by high-fre- Genomic islands: tools of bacterial horizontal gene transfer and evol-
quency gene rearrangements. Proc Natl Acad Sci USA 95: 13923–8. ution. FEMS Microbiol Rev 33: 376–93.
98. Saunders NJ, Peden JF, Hood DW, Moxon ER. 1998. Simple sequence 111. Pandey DP, Gerdes K. 2005. Toxin-antitoxin loci are highly abundant in
repeats in the Helicobacter pylori genome. Mol Microbiol 27: 1091–8. free-living but lost from host-associated prokaryotes. Nucleic Acids Res
99. Hamilton HL, Dillard JP. 2006. Natural transformation of Neisseria 33: 966–76.
gonorrhoeae: from DNA donation to homologous recombination. Mol 112. Iida S, Streiff MB, Bickle TA, Arber W. 1987. Two DNA antirestriction
Microbiol 59: 376–85. systems of bacteriophage P1, darA, and darB: characterization of darA-
100. Seib KL, Peak IR, Jennings MP. 2002. Phase variable restriction- phages. Virology 157: 156–66.
modification systems in Moraxella catarrhalis. FEMS Immunol Med 113. Dryden DT, Tock MR. 2006. DNA mimicry by proteins. Biochem Soc
Microbiol 32: 159–65. Trans 34: 317–9.

Bioessays 33: 43–51,ß 2010 WILEY Periodicals, Inc. 51


Gene, 27 (1984) 131-149 131
Elsevier

GENE 919

The use of transposon Tn5 mutagenesis in the rapid generation of correlated physical and genetic
maps of DNA segments cloned into multicopy plasmids - a review?

(Escherichia coli; rhizobia; agrobacteria; recombinant DNA; nitrogen fixation; transposition frequency;
insertional specificity; polarity; bacteriophage L and Mu; eukaryotic genes)

F.J. de Bruijn * and J.R. Lupski **

* Department of Cellular and Developmental Biology, Harvard University, Cambridge. MA 02138. Department of Molecular
Biology. Massachusetts General Hospital,Jackson 11, Boston, MA 02114, and ** Biochemistry Department, New York Universit?
Medical Center, 550 First Avenue, New York, NY 10016 (U.S.A.) Tel. (212) 340-5137

(Received August 15th, 1983)


(Accepted September 6th, 1983)

SUMMARY

The properties of transposon Tn5 that render it useful for in vivo mutagenesis of cloned DNA sequences
are reviewed. Transposition frequency, insertional specificity, polarity and stability of Tn5 insertion mutations
are among the topics discussed. Examples are cited from the published literature which illustrate the applications
of Tn5 mutagenesis to the analysis of cloned prokaryotic and eukaryotic genes. A methods section is included
which outlines precisely how to carry out transposon Tn5 mutagenesis analysis of cloned DNA segments.

INTRODUCTION

Transposable genetic elements carrying antibiotic


resistance genes (Tn elements or transposons) have
+ Dedicated to the memory of Ahmad I. Bukhari, our friend and
mentor, who inspired writing this review.
proven to be extremely useful tools in bacterial
* To whom correspondence and reprint requests should be sent genetics (see Kleckner et al., 1977):
at his present address: Max-Planck-Institut fur Ziichtungsfor- (1) The transposition of a Tn element into a gene
schung, Abt. Schell, D-5000 Koln, Vogelsang 30 (F.R.G.) Tel. usually leads to inactivation of that gene, with such
(49)-221-5062315 or 5062341.
insertion mutations being generally nonleaky and
Abbreviations: Ap’, ampicillin resistance; LB, Luria broth, SM
stable.
buffer, YM broth, see MATERIALS AND METHODS, section (2) Such insertion mutations in multi-gene ope-
a; Cm’, chloramphenicol resistance; Em’, erythromycin resis- rons usually exert polar effects on genes located
tance; IR, inverted repeat; Km’, kanamycin resistance; kb, kilo- downstream from the insertion site. This is presum-
base pairs; m.o.i., multiplicity of infection; M-STET, see MATE- ably due to the presence of transcriptional as well as
RIALS AND METHODS, section el; Nm’, neomycin resis-
translational termination signal sequences carried by
tance; NPT II, neomycin phosphotransferase type II; p.f.u.,
plaque-forming units; RIA, radioimmune assay; SDS, sodium
the Tn element.
dodecyl sulfate; Sm’, streptomycin resistance; Tc’, tetracycline (3) A Tn insertion mutation represents a new
resistance; ::, novel joint. genetic (e.g., antibiotic resistance) and physical

0378-I 119/84/$03.00 0 1984 Elsevier Science Publishers


132

marker at the site of insertion. The latter can be Tn5 mutagenesis has proven extremely useful in
physically identified, e.g., by DNA-DNA hetero- the molecular genetic analysis of the Gram-negative
duplexing or by restriction analysis. Rhizobiaeceae family. Rhizobia induce the formation
In this review we shall focus on the properties of of specialized structures (nodules) on the roots or
a 5700-bp transposon Tn5 (Berg et al., 1975; Berg, stems of their host plant, in which the bacteria carry
1977), encoding the NPTII, which confers resistance out the nitrogen-fixation process (see Sprent, 1979).
to a number of aminoglycoside antibiotics, such as Agrobacteria induce the formation of crown-gall
neomycin and kanamycin (and streptomycin upon tumors on their host plant (see Kahl and Schell,
certain bacterial species, such as Rhizobium meliloti; 1982). Since the phenotype of any bacterial mutation
Putnoky et al., 1983) upon its host (Berg et al., 1978). in a gene involved in the symbiotic associations is
Transposon Tn.5 is composed of two inverted only observable in plantum, conventional muta-
repeats of 1535 bp (IS.50; Berg, 1983) containing genesis and screening techniques are of limited use.
genes responsible for transposition and its regu- A plasmid vector for the introduction of transposon
lation, flanking an approximately 2700-bp region Tn5 and the finding that Tn5 is capable of trans-
carrying the structural gene for NPTII (see Rezni- position and inducing insertion mutations (see
koff, 1982; for an exhaustive review of the biology of below; Beringer et al., 1978; Van Vliet et al., 1978),
Tn5, see Berg and Berg, 1983). made possible the pre-selection of random muta-
Tn5 mutagenesis has been successfully employed tions, thereby effectively reducing the number of
in the genetic analysis of a number of bacterial infected plants to be screened. This has resulted in
species. TnS-induced insertion mutations have been the identification of a number of Rhizobium loci in-
generated in chromosomal genes and operons of volved in nodulation (nod) and nitrogen fixation (nif)
Escherichia coli and Salmonella typhimurium, such as (Beringer et al., 1978; Rolfe et al., 1980; 1981; Dun-
his (Kleckner et al., 1977; 1979; Biek and Roth, can, 1981; Meade et al., 1982; Scott et al., 1982;
1980), lac (Berg et al., 1980; Miller et al., 1980), ilv Buchanan-Wollaston et al., 1980; Lamb et al., 1982;
(Berg et al., 1979), trg (Harayama et al., 1979), ksg Ma et al., 1982; Forrai et al., 1983) and Agrobac-
(Fouts and Barbour, 1981), uvrD, mutB and mutD terium loci involved in tumor formation and opine
(Shanabruch et al., 1981) and pnp (Portier, 1980). catabolism (Garlinkel and Nester, 1980; and for
Tn5 mutagenesis has been used to generate muta- reviews see Bevan and Chilton, 1982 and Kahl and
tions and then to facilitate cloning of genes of bac- Schell, 1982).
terial species such as Caulobacter crescentus Ruvkun and Ausubel (1981) extended the Tn5
(Purucker et al., 1982), Myxococcus xanthus (Kuner mutagenesis methodology for the Rhizobiaceae by
and Kaiser, 1980; Shimkets et al., 1983) and Erwinia developing a general method for site-directed Tn5
herbicola (Gantotti et al., 1981). Tn5 mutagenesis mutagenesis. It involves the cloning of Rhizobium (or
has also been used to study genes coding for bio- Agrobacterium) nif, nod or tumor formation genes
luminescence of Vibrio fischeri (Engebrecht et al., into multicopy plasmids, followed by Tn5 muta-
1983) and Vibrio harvqyi (Belas et al., 1982) IgAl genesis in E. coli, reintroduction of the TnS-mutated
proteases of Haemophilus infuenzae (Bricker et al., sequences into Rhizobium (or Agrobacterium) and
1983) heme biosynthesis in R. meliloti (Leong et al., forced gene-replacement of the corresponding Rhizo-
1982), phase variation of Salmonella (Silverman bium (or Agrobacterium) wild-type sequences with
and Simon, 1980), naphthalene degradation genes the TnS-mutated counterparts. Such analysis has
(Schell, 1983), type I fimbriae of Klebsiella pneu- made possible the combined physical and genetic
moniae (Purcell and Clegg, 1983). and acetohy- characterization of additional nod and nifgenes in a
droxy acid synthetase in S. typhimurium (Shaw et al., variety of Rhizobium species (Ruvkun et al., 1982;
1980). The structure and organization of bacterial Ausubel, 1982; Corbin et al., 1981; 1983; Fuhrmann
episomes and plasmids such as mini-F (Phua et al., and Hennecke, 1982; Scott et al., 1982) and the
1982) and CloDF13 (Hakkaart et al., 1981) and construction of correlated physical and genetic maps
bacteriophages such as Pl (Heilmann et al., 1980) of the regions of the tumor inducing (Ti) plasmid of
and Mu (Giphart-Gassler et al., 1982) have also A. tumefaciens (Garlinkel et al., 1981; Klee et al.,
been facilitated by the use of Tn5 mutagenesis. 1983; for reviews see Bevan and Chilton, 1982; Kahl
and Schell, 1982).
133

TnS-induced mutations have been used to clone of parameters of transposon Tn5 mutagenesis such
genes that lack a readily identifiable phenotype, to as vectors for the introduction of Tn5, transposition
direct suppressive integration of ColEl derivatives frequency and insertional specificity of this trans-
into the chromosome of dnaA strains of E. coli poson, stability and polarity of TnS-induced inser-
(Sasakawa and Yoshikawa, 1980) and to construct tion mutations and convenient restriction sites within
transposable genetic elements carrying genes of Tn5 for physical mapping, based on our observations
interest by flanking such genes with two complete as well as the observations from the literature cited.
copies of Tn.5 (Guarente et al., 1980), ,or by cloning In the interest of clarity, we will cite a number of the
the gene(s) of interest directly within the transposon best studied cases to illustrate our points, but will not
Tn5 sequences (Meyer et al., 1979). The selectable attempt to provide an exhaustive reference list for
genetic marker introduced by Tn5 into cryptic plas- each point discussed. In addition, we include a
mids or those without a readily selectable phenotype “cook-book” style Materials and Methods section,
have been useful for the genetic transfer and identifi- describing the experimental techniques involved in
cation of such TnS-tagged plasmids (Johnston et al., the Tn5 mutagenesis protocol we employ.
1978; Lamb et al., 1982; Hooykaas et al., 1981).
Finally, a simple deletion derivative of transposon
Tn5 has been proposed to be useful in the creation
of a “mobile recombinational switch”, since it is MATERIALS AND METHODS
capable of turning on the expression of genes down-
stream from its insertion site in one orientation, but (a) Media and solutions (see Miller, 1972)
not the other (Berg, 1980).
Tn5 mutagenesis for construction of correlated A broth: 10 g Bactotryptone, 2.5 g NaCl per liter.
physical and genetic maps was first successfully Plates are solidified with 1.1% agar.
applied to the characterization of the chromosomal A top agar: 1 broth plus 0.6% agar.
nitrogen fixation (nif) gene cluster of K. pneumoniue YM broth: 2 broth plus 0.2% maltose plus 0.01%
by Riedel et al. (1979). These authors physically yeast extract.
mapped a large number of transposon insertions in LB (Luria broth): 10 g Bactotryptone, 5 g yeast
the nifgene cluster (isolated and genetically mapped extract, 5 g NaCl per liter with the pH adjusted to 7.5
by Merrick et al., 1978; 1980), by cleavage of total with NaOH.
chromsomal DNA of strains carrying nif: :Tn.5 inser- SM buffer: 0.1 M NaCl, 0.02 M Tris pH 7.5,
tions with appropriate restriction endonucleases (see 0.01 M MgSO,, 0.01% gelatin.
Fig. 2), followed by Southern transfer and probing of
the resulting filters with cloned subfragments of the (b) Bacteriophage and bacterial strains
nifregion. Altered patterns of hybridization, due to
the presence of transposon insertions allowed the The A::Tn5 phage was originally constructed by
physical mapping of the transposon. This combined Dr. Douglas Berg (Berg et al., 1975). The 1 bacterio-
with the genetic mapping studies resulted in the phage vector we used as a source of Tn5 was 1467,
construction of a correlated physical and genetic which was obtained from Dr. Nancy Kleckner.
map of the nzf region by unambiguously ordering Phage 1467 has the following genotype: lb221
Tn-induced insertion mutations, providing an esti- rex: :Tn5 ~1857, Oam29, Pam80. The b22 1 mutation
mation of the minimum sizes of individual genes and is a deletion in the 1 genome that removes att, the
an assignment of specific genes to restriction frag- phage attachment site. This prevents the. bacterio-
ments (Riedel et al., 1979). phage from undergoing lysogeny. Gene rex is non-
In this review we will focus on an extension of the essential for Agrowth and in this case contains a Tn5
transposon Tn5 mutagenesis methodology described insertion. The ~1857 mutation results in a tempera-
by Riedel et al. (1979), namely the rapid construction ture-sensitive repressor. Both the 0 and P genes are
of correlated physical and genetic maps of pro- involved in phage i DNA replication, so amber
karyotic (and eukaryotic) DNA segments cloned mutations in these genes will prevent phage replica-
into multicopy plasmids. We will discuss a number tion in a Su” background.
134

Bacterial strain Ql (fhr-1 Zeu-6 thi-1 IucYl, supE44 physically verified using one of the rapid plasmid
ton,421 T5’ $80’) or strain LE392 (F ~, hsdR5 14 (rk , preparation protocols described below. One must be
mk ) supE44 supF58 lacy1 galK2 gal722 metB1 certain that the plasmid is a monomer. Plasmid-
trpR.5.5 A- ) is used as host for propagating A467. In bearing cells are then grown to saturation in 5 ml YM
our experience LE392 appears to contain a better broth (supplemented with antibiotic) at 30°C. The
suppressor and thus gives higher titre phage stock. culture is diluted lOO-fold and grown at 30’ C in 5 ml
Any bacterial strain that is neither Su’ nor 3, YM broth plus antibiotics to an A,,, of 0.8 (approx.
resistant can be used as the host for plasmids to be 10” cells/ml). Then 1 ml of cells is mixed with 1,: :Tn5
mutagenized with A: :TnS. phage at an m.o.i. of 1- 10 and placed at 30°C for
2 h. Infected cells are then plated in 0.2-ml aliquots
(c) Preparation of 12467 (L::TnS) phage stocks onto LB agar plates containing 20 pg/ml kanamycin
and the antibiotic used to select for the plasmid. The
Cells of LE392 are infected with 2: :Tn5 phage and plates are incubated at 30°C for 48 h. Km’ colonies
incubated overnight at 37 o C to obtain fresh plaques. are washed off the plates by adding 5 ml 209, sucrose
Simultaneously, a fresh culture of LE392 is grown in solution (20 O0 sucrose, 10 mM Tris pH 7.8, 1 mM
5 ml YM broth to saturation. The next day the EDTA) to each plate using a sterile glass spreading
LE392 culture is diluted in LB and regrown to an rod. The cell suspension is decanted into a sterile
AhOO(Bausch and Lomb Spectronic 20) of approx. 30-ml Corex centrifuge tube and the cells are collect-
0.8. One fresh plaque from the above plate is picked ed by centrifugation at 7000 rev./min for 10 min in a
with a sterile Pasteur pipette and placed into 1 ml of Beckman JA-17 rotor. The cells are resuspended in
SM buffer (see Maniatis et al., 1982). Then 0.25 ml 5 ml sucrose solution or LB broth and plasmid DNA
of SM buffer containing phage is added to 0.5 ml of is isolated by the cleared lysate procedure of Clewell
the regrown LE392 cells. After mixing, the cells are and Helinski (1969) or using a scaled-up (5 x )
incubated at 37°C for 20 min to allow phage version ofthe alkaline lysis protocol described below.
absorption. To the cell and phage mixture 7.5 ml of Plasmid DNA thus prepared is then used to trans-
melted ,? top agar equilibrated at 45 “C is added. form competent cells of the desired bacterial strain,
Aliquots of 2.5 ml are then spread per A plate. An in order to screen for the phenotype of the Tn5
equivalent amount of LE392 cells with no phage mutated plasmid. When characterizing Tn5 inser-
added is plated as a control. The plates are incubated tions into a cloned gene with a readily observable
at 37°C for 6-8 h and inspected for lysis by com- phenotype, the bacterial strain of choice will carry a
paring with the LE392 control. When confluent lysis mutation in such gene, and the TnS-mutated plas-
is observed on the plates containing the A::TnS-in- mids are directly screened for their ability to comple-
fected cells, and the control plates show a visible ment the mutation. For example, in the case of the
bacterial lawn, the phage is harvested by scraping the Tn5 mutagenesis analysis of the cloned glnALG
top agar into a sterile screw-top Corex centrifuge (glnAR) operon of K. pneutnoniae, plasmid DNA
tube and adding 2.5 ml SM buffer plus 0.5 ml chlo- from I::TnS-infected cells was used to transform
roform. The mixture is then vortexed thoroughly, competent cells of a GlnA R- K. pneumoniue
incubated on ice for 5 min, and centrifuged in a strain, and the transformants were screened for their
Beckman JA-17 rotor at 10000 rev./min, 4°C for GlnA and GlnR phenotype (De Bruijn and Ausubel.
5 min. After decanting the supernatant into a sterile 1981; see Fig. 4). Thus ghzA : :Tn5 and
screw-top tube, a few drops of chloroform are added glnR(LG): :Tn5 insertions were genetically identified.
and the lysate is stored at 4°C. The phage are then When characterizing Tn5 insertions into a cloned
titered on LE392 (see Miller, 1972). gene without a genetically identifiable phenotype,
such as a eukaryotic gene, DNA from A: :Tn5 infect-
(d) Mutagenesis of plasmid DNA with transposon ed cells is used to transform any desired strain of
Tn5 E. coli, followed by screening of the transformants by
other means such as antibody binding assays (see
The plasmid to be mutagenized is transformed EXPERIMENTAL, SeCtiOn j).
into any Su’ /1” bacterial strain and its presence
135

(e) Rapid plasmid minipreps (2) Alkali lysis method [modified from method by
Bimboim and Doly (1979)]
(1) Boiling method [modified from methods by De A plasmid-bearing strain is inoculated from a
Bruijn and Ausubel (1981), Holmes and Quigley single colony into 3 ml of LB plus appropriate
(1982) and Lupski et al. (1982)] antibiotic. The cells are grown to A,,, (Spectronic
Cells from a 5-ml saturated culture, grown in LB 20) of approximately 0.4-0.5, and 300 pg chloram-
plus the appropriate antibiotic, are placed in a 15-ml phenicol is added to amplify the plasmid. After
Corex tube and pelleted by centrifuging at allowing approx. 5 h for amplification, the cells are
7000 rev./min for 10 min in a Beckman JA-17 rotor. collected in a 1.5-ml eppendorf tube by performing
The cell pellet is resuspended in 50 ~1of 25 % sucrose 2 x 30 s centrifugations in an eppendorf table-top
by vigorous vortexing. To this suspension 300 ~1 of centrifuge. The pellet is resuspended in 200 ~1resus-
M-STET (5% Triton X-100,50 mM EDTA, 50 mM pension buffer (25 mM Tris pH 8.0,50 mM glucose,
Tris pH 8, 5 % sucrose) is added. After mixing, 50 ~1 10 mM EDTA, 2 mg/ml lysozyme, 50 pg/ml RNase)
of 20 mg/ml lysozyme in H,O is added. This mixture and incubated on ice for 5 min or until the viscosity
is gently vortexed, transferred to a 1.5-ml eppendorf changes. It is then mixed with 200 ~1 of freshly pre-
tube using a Pasteur pipette, heated in a boiling water pared alkali lysing buffer (0.5% SDS, 0.2 N NaOH)
bath for 2 min, and placed on ice. The tubes are spun until the solution becomes clear; then 200 ~1 3 M
for 15 min in an eppendorf table-top centrifuge K * acetate pH 4.8 is added, and a white precipitate
placed in the cold room. The viscous pellet is gently appears. This is mixed well, but gently, and incubated
removed with a pipette, and to the supematant 50 ~1 for 5 min at 0°C. The solution is centrifuged for
of 3 M K. acetate (pH 4.8) and 500 ~1of isopropanol 5 min at 4°C and the supematant is transferred to
are added. The DNA is allowed to precipitate at another eppendorf tube. The DNA is precipitated by
- 20’ C for l-2 h, then pelleted by centrifugation for adding 500 ~1isopropanol and placing it at - 20’ C.
15 min in the eppendorf centrifuge kept at 4’ C. The After centrifuging for 5 min in the cold room, the
white pellet is dried and resuspended in 120 ~1H,O. pellet is resuspended in 50 ~1 H,O and passed over
The chromosomal DNA remains as a pellet, while a G-50 Sephadex microcolumn as described above.
the plasmid DNA readily goes into solution. A quick
spin is performed to again pellet this chromosomal
DNA, and the resuspended plasmid DNA is passed
over a 400 ~1 Sephadex G-50 (medium) column EXPERIMENTAL
equilibrated with 10 mM Tris pH 7.5, 1 mM EDTA.
The Sephadex minicolumns are made by puncturing (a) Vectors used for transposon Tn5 mutagenesis
the bottom of a 400 ~1 eppendorf tube, using a 20
gauge needle heated over a bunsen burner, covering A number of different vectors have been employed
this hole with a small glass wool plug, followed by for the generation of TnS-induced insertion muta-
sterilization and placing the tube inside a larger tions. For the mutagenesis of bacterial genera such
1.5-ml eppendorf tube which acts as a support. The as Rhizobium, Agrobacterium, Caulobacterium and
400~~1eppendorf is then tilled to overflowing with Erwinia, a promiscuous Pl-type R-factor (RP4),
equilibrated G-50 Sephadex and centrifuged at top carrying a copy of bacteriophage Mu and of Tn5
speed for 75 s in an IEC table-top centrifuge to pack (pJB4JI; Beringer et al., 1978) has been utilized.
the column. The 120~1 of resuspended plasmid This plasmid is extremely unstable in the above-
DNA is then loaded on top of the packed column mentioned bacterial species, presumably due to Mu-
and the tube spun again in a 1.5-ml eppendorf tube mediated illegitimate recombination events upon
at top speed for 45 s. In this manner 24 minipreps introduction, and is therefore called a “suicide plas-
can be done simultaneously. Enough purified plas- mid”.
mid DNA is usually recovered from 5 ml of liquid Selection for Km’ or Nm’ afer mating the plasmid
culture to perform 10-12 restriction enzyme digests. across from E. coli results in the transposition of the
Tn5 from the “suicide plasmid” into the genome of
the recipient cell, followed by loss of the plasmid
136

vector DNA sequences (Beringer et al., 1978; Van mutagenesis has been made possible even in bac-
Vliet et al., 1978; Ely and Croft, 1982; Gantotti et al., terial species such as S. typhimurium, which are
1981; Duncan, 1981; Walton and Moseley, 1981). normally not sensitive to A infection. Palva and
However, Tn5 mutagenesis using pJB4JI and deri- Lindstrom (198 1) have shown that introduction of
vative vectors has generated ambiguous results in the E. coli malB region, encoding the 3, receptor
some bacterial species such as Rhizobium meliloti protein (LamB), into S. typhimurium allows II phage
(Meade et al., 1982). These authors reported that a adsorption and DNA injection, but not replication,
large percentage of the induced insertion mutations in this bacterium.
carried phage Mu DNA sequences in addition to Other vectors useful for the introduction of Tn5
Tn5 sequences at the site of insertion. Therefore, include plasmid ColEl derivatives carrying a tem-
when using this Tn5 mutagenesis protocol, caution perature sensitive replication mutation in addition to
should be exercised in analyzing presumptive Tn5- the Tn5 transposon, allowing the selection for Tn5
induced insertion mutations; the resulting strains transposition at elevated temperatures (Laird and
should be checked for the presence of both Tn5 and Young, 1980; Meade, H., Brown, S. and Ausubel,
Mu DNA sequences by Southern blotting (Meade F., unpublished). This allows Tn5 mutagenesis in
et al., 1982; Forrai et al., 1983). any bacterial species which can be transformed with
An alternative vector has been constructed con- ColEl type plasmid DNA. In addition, strains
sisting of the pBR325 replicon (Bolivar et al., 1977) containing a single copy of Tn5 integrated into the
into which the mobilization (mob) site of the promi- chromosome, either as a simple Tn5 insertion or as
scuous P-type plasmid RP4 has been inserted and part of an integrated /i : : Tn5 prophage, have been
carrying a copy of Tn5 (pSUP2021; Simon et al., used to generate Tn5 insertions (for examples see
1983). Such a plasmid can be mobilized at a very Fuhrmann and Hennecke, 1982; Pannekoek et al.,
high frequency to bacterial species such as Rhizobium 1980; Hakkaart et al., 1981).
or Agrobacterium, when provided in tram wit transfer
functions. Once transferred conjugally, it is unable to
replicate in the recipient Rhizobium or Agrobacterium (b) Transposon mutagenesis protocol
cells, and addition of Km or Nm to the growth
medium selects for the transposition of the Tn5 from For transposon Tn5 mutagenesis of DNA seg-
the plasmid replicon into the genome of the cell. This ments cloned into (multicopy) plasmids in E. coli, the
vector may circumvent some of the problems asso- ;i : : Tn5 (1467) vector has proven to be the most
ciated with the pJB4JI plasmid described above. useful one, due to the ease with which high titer
For Tn5 mutagenesis in E. coli, S. typhimurium, stocks of this conditionally defective phage can be
and Myxococcus xanthus, bacteriophage vectors prepared (see MATERIALS AND METHODS,
have been used. In the case of S. t_vphimurium a section c), the high frequency with which Tn5 trans-
derivative of P22, incapable of lytic growth or stable poses from the A : : Tn5 vector, and the efficiency
lysogenization and carrying Tn5 (DB 2062; D. Bot- with which the vector is lost (segregated away) after
stein) has been employed (see for example Shana- Tn5 transposition. The protocol routinely used is
bruch et al., 1981). In Myxococcus bacteriophage outlined in Fig. 1 and can be broken down into the
Pl : : Tn5, capable of adsorption and injection of following steps:
phage DNA, but incapable of self-propagation in (1) Infection of E. coli cells harboring the (multi-
this bacterium, has been used (Kuner and Kaiser, copy) plasmid carrying the cloned DNA segment of
1981). In bacterial species that are sensitive to interest with the A: : Tn5 (see MATERIALS AND
infection by phage A, a ,? derivative deleted for the att METHODS, section d).
site and carrying amber mutations in genes 0 and P (2) Growth of the infected cells on plates contain-
(rendering it incapable of integration or replication in ing kanamycin (or neomycin) and the antibiotic used
Su” strains) with Tn5 inserted in a nonessential gene to select for the plasmid, to select for Tn5 trans-
(2467; Lb221 rex : : Tn5 ~1857 Oam29 Pam80; Berg position from 2 : : Tn5 into the chromosome of the
et al., 1975; N. Kleckner, unpublished observations) cell or into the resident plasmids.
has proven to be most effective. Recently i : : Tn5 (3) Preparation of plasmid DNA from the cells
137

Tn5 MUTAGENESIS PROTOCOL

® INFECT PLATE LB+Cm(or····)


+ Km

((¡ E.coli ). 5
>.: :Tn5 +
( >,: replicot1on plosmid (Cm r
and intec;¡rotion or Te r etc) r
Cm r Km colonies
defic1ent)

Wild type
or EXTRACT
mutant . - - - TRANSFORM . . - - PLASMID
strain DNA

r r SCREEN EXTRACT RESTRICTION


Select Cm Km PHENOTYPE PLASMID - + MAP
DNA
Fig. l. Tn5 mutagenesis protocol. The general transposon Tn5 mutagenesis protocol employed in our studies includes the following
steps: (1) lnfection of cells harboring the recombinant plasmid to be mutagenized with .t:Tn5 (see MATERIALS AND METHODS,
section d). (2) Plating of the infected cells on LB plates containing Km (to select for Tn5) and the antibiotic corresponding to the
resistance gene carried by the plasmid vector (such as Cm, Te, or Ap). (3) Incubation ofthe plates at 30 º C for 48 h. (4) Extraction
of plasmid DNA from the Km' (and Cm', Te' or Ap') cells thus obtained (see MATERIALS AND METHODS, section d).
(5) Transformation of cells from mutant strains of E. co/i, in case the genetic phenotype ofthe Tn5-mutated plasmids can be determined
by genetic complementation, and wild-type cells if another method ofphenotypic identification is employed (see EXPERIMENTAL,
sectionj). (6) Screening ofthe phenotype ofthe resulting transformants (see MATERIALS AND METHODS, section d). (7) Extraction
ofplasmid DNA from the transformants ofinterest (see MATERIALS AND METHODS, section e). (8) Restriction mapping ofthe
Tn5-containing plasmids to determine the physical location of the Tn5 within the plasmid (see EXPERIMENTAL, section k).

TnS
IR IR
Km' obtained in the previous step (see MATERIALS AND
METHODS, section d).
cr,-c, :,:
-=-
"'"'X D
t

-
"' ;¡;
ce..
-C,)<
(4) Transformation o f competent cells o f E. coli
00

- =
) < ...,
::, u, "'
.-3, = o
Ql:::r I» ;;·
:. 5 .,
g, = 3
:,: = -
c.
carrying (a) mutation(s) in the gene(s) ofinterest, to

1kb
carry out complementation studies using the wild-
type and Tn5-mutagenized D N A segments, or a
Fig. 2. Partial restriction map of Tn5. In this figure the most
useful restriction sites within transposon Tn5 are indicated. Tn5 wild-type strain i f a screening method different from
does not contain any sites for the restriction endonucleases genetic complementation is employed.
EcoRI, Ball, Kpnl, Pvul, Clal, or Sst 1, and therefore these endo-
nucleases are useful to determine the approximate physical
location of a Tn5 with respect to target plasmid DNA fragments analysis of Tn5-target junction sequences (see EXPERIMEN-
generated by these restriction endonucleases. In addition, Tn5- TAL, section 1). Sa/1 and Smal can be employed to determine
containing DNA segments can be easily re-cloned using these the relative orientation of the Tn5, since these restriction endo-
endonucleases. Hindlll, Xhol, Pst I, Bg/II and Hpal cleave within nucleases cleave Tn5 asymmetrically. BamHI is convenient for
the in verted repeats (IRs) and are useful to determine the exact mapping the Tn5 insertion site, since it cleaves Tn5 approxi-
physical location ofthe Tn5 insertion, relative to (a) given site(s) mately in the middle. The cleavage sites shown are derived from
within the plasmid target sequences and their distance away from Jorgensen et al. (1979) and Auerswald et al. (1981). The nucleo-
the ends ofTn5 are 1195 bp, 485 bp, 680 bp, 1515 bp and 185 bp, tide sequence for the Km' gene encoding NPTII has also been
respectively. Hpal is also extremely useful for the DNA sequence determined by Beck et al. (1982).
138

(5) Isolation of plasmid DNA from single colonies the useful cleavage sites within Tn5 shown in Fig. 2
of the transformed cells using a small-scale plasmid (see EXPERIMENTAL, section k; for an example see
DNA preparation protocol (see MATERIALS AND Fig. 3).
METHODS, section e), followed by restriction endo- (6) Construction of a correlated physical and
nuclease mapping of the Tn.5 insertions employing genetic map of the cloned DNA segments by com-
bining the genetic complementation data obtained in
step 4 with the physical mapping data obtained in
step 5 (for an example see Fig. 4).
This protocol has been most effective in the
analysis of DNA segments carried by multicopy
plasmids, due to the relative ease with which large
numbers of independent Tn.5 insertions can be
generated and with which plasmid DNA can be pre-
pared from l-5-ml cultures for restriction endo-
nuclease analysis (see MATERIALS AND METHODS,
sections d and e). However, it can also be employed
in the analysis of DNA segments carried by low-
copy-number or single copy plasmids. In these cases,
the number of transpositions generated in a standard
H P H H P H experiment is reduced and the amount of cells used
for small scale plasmid DNA preparation has to be
W-1 _J
C B A
increased, but all other criteria remain the same.

Fig. 3. Example of an experiment to determine the location of


(c) Transposition frequency of Tn5
plasmid-borne Tn5 insertions. Hind111 cleavage pattern was
obtained for a series of plasmid::Tn5 isolates, run on a 0.75”,,
agarose gel and stained with ethidium bromide. The plasmid The transposition frequency of Tn.5 from the
used, pEGSI contains a single Hind111 site. TnS is cleaved twice /I : : Tn5 vector into the chromosome of its recipient
by HitzdIII, within the inverted repeats (IRS; see Fig. 2). There- cell is approximately 5 x 10 5 Km’ colonies per
fore cleavage of pEG8l::TnS plasmid with Hind111 generates
p.f.u. of the 2 : : Tn5 phage (Johnson et al., 1982)
three fragments, one fragment is a constant. internal 3400-bp
and from a site in the chromosomal fat operon (or
Tn5 fragment (B) and the other two reprcscnt Tn-target
function fragments (A and C). By measuring the size of the A an F’luc) into resident iL phage approx.
junction fragments and subtracting I I95 bp (distance between 5 x 10 ’ Km’ transductants per /1 phage produced
the Hind111 site in the inverted repeat and the end of Tn5), the (Egner and Berg, 1980).
exact position ofthe Tn5 insertion, relative to the unique Hind111 In our experiments we routinely infect 1 x 10”
site in pEG81, can be determined. The left-hand lane S shows the
cells with l-10 x 10” p.f.u. of iL: : Tn5 and obtain
DNA M, standards used, consisting of phage 1 DNA cleaved
with Hind111 + KpnI. and their sizes arc indicated in kb.
l-2 x 10” Km’ colonies, representing an approxi-
Additional M, markers used are shown in the right-hand lane S mate transposition frequency of 2 x 10 ’ to
and consist of M l3mp8 cleaved with HnelII; the size of these 1 x 10Ph Km’ colonies per p.f.u. of L : : Tn5 (De
fragments is 2527, 1623 and 849 bp, respectively. Lanes l-10 Bruijn et al., 1983). The transposition frequency of
show the Hind111 digestion pattern of ten pEG8 1::Tn5 plasmids
Tn5 into a supercoiled, multicopy plasmid may be
(Lupskiet al., 1983b)carryingTn5insertionsprogressivelyfarther
higher than the transposition frequency into a chro-
away from the closest unique Hind111 site of pEG8l. At the
bottom, the hatched boxes represent the pEG81 vector DNA mosomal DNA sequence, due to the possible prefer-
sequences, the open box represents TnS sequences (the wide ence of Tn elements to insert into actively replicating
open boxes represent the inverted repeats while the narrow open or supercoiled DNA molecules (Fitts and Taylor,
box represents the internal portion of Tn5 including the Km’ 1980). This would explain the relative ease with
gene), and the line represents the remainder of the cloned DNA
which 100-200 independent Tn5 insertion mutations
fragment carried by pEG81. The position of HirzdIII and Psi1
restriction sites is indicated by H and P. respectively. Restriction
into a plasmid can be generated from the infection of
fragment lengths A, B, and C correspond to those shown in the a 1 ml, mid-log culture of plasmid containing cells
above gel. (De Bruijn and Ausubel, 198 1; Ruvkun and Ausubel,
139

glnRlLGl glnA

pFB514

,
1
lkb ,
VGlnA-
VGlnA+R- Tn5 insertions
VGlnA+R+

Fig. 4. Example of a physical and genetic map of a cloned DNA segment, as generated by transposon Tn5 mutagenesis. The triangles
indicate the physical map position of independent Tn5 insertions within the cloned DNA fragment as determined by first analyzing
pFBS 14::Tn5 plasmid DNA with EcoRI, which does not cleave Tn5 (see Fig. 2), in order to determine into which pFBS 14 EcoRI fragment
the transposon had inserted. The precise physical location of each Tn5 insertion was determined relative to the Hitid sites flanking
the cloned DNA segment, by cleavage of the pFB514::TnS plasmids with Hind111 and determining the size of the TnS-target DNA
junction fragments (De Bruijn and Ausubel, 1981; see also Fig. 3). The phenotype of the pFB514::TnS plasmids was determined by
examining their ability to complement GlnA and/or GlnR - mutations of Klebsiellu pneumoniae. The solid, open and dotted triangles
represent GlnA _, GlnA + R- and GlnA + R + pFB514::TnS plasmids respectively. The boxed-in segments represent plasmid vector
DNA sequences and the horizontal line represents the cloned K. pnezmzoniue DNA fragment. The combination ofthe physical and genetic
data resulted in the construction of the correlated physical and genetic map of the cloned glnAR(glnALG) region of K. pneumoniue, as
shown (De Bruijn and Ausubel, 1981). This analysis enabled these authors to determine the location and physical boundaries of the
structural gene for glutamine synthetase (ghA) and the identification of a previously unidentified locus glnR(ghLG), immediately
adjacent to gInA, involved in positive activation of the nitrogen fixation (nif) genes in K. pneumoniae.

1982; Lupski et al., 1982; 1983b,c; De Bruijn et al., trophic mutations in E. coli. These authors con-
1983). cluded that even though a slight preference for
The transposition frequency of a given transposon insertion into certain genes appeared to exist, never-
may vary greatly depending on its host organism. theless a large variety of TnS-induced auxotrophic
For example, the transposition frequency of Tn5 in mutations could be readily identified. These results
K. pneumoniae (from which this transposon was were extended by Berg et al. (1980), who examined
originally isolated) appears to be even higher than in the distribution of Tn5 insertions into the E. coli lac
E. coli, and therefore difficulties have sometimes operon and concluded that less than 57; of the
been encountered generating and stably maintaining lac: : Tn5 insertions could not be separated by
a single Tn5 insertion in K. pneumoniae because of genetic recombination, further suggesting a low
secondary Tn5 transpositions (see EXPERIMENTAL, insertional specificity of Tn5. Independent results
section f). from Miller et al. (1980) support this conclusion.
In our experiments we examined this question by
(d) Insertional specificity of Tn5 determining the distribution of 1500 independent
Tn5 insertions into a multicopy plasmid carrying
A variety of Tn elements, such as Tn3, Tn5, Tn9, three genes whose expression could be monitored
TnlO and phage Mu, have been examined for their easily (pGR100; hisD, Tc’, Ap’; Riedel, 1980). By
insertional specificity or insertion site preference (for determining the number of Tn5 insertions leading to
a review see Kleckner, 1981). Tn5 has been ranked insertional inactivation of each of the three genes and
among the Tn elements with a relatively low inser- comparing these numbers with the percentage of
tional specificity based on a number of different plasmid DNA each individual gene occupies, an
experiments. Shaw and Berg (1979) examined the essentially random distribution pattern was observed
insertional specificity of Tn5 on a “macro-level” by (De Bruijn et al., 1983). In addition, by determining
determining the distribution of TnS-induced auxo- the physical location of 92 Tn5 insertions in the
140

cloned K. pneumoniae glnALG(glnAR) operon (De inverted repeat (Bossi and Ciampi, 1981; Berg,
Bruijn and Ausubel, 1981), 88 Tn5 insertions in the 1983). Thus, there may be some preferential sites for
cloned K. pneumoniae hisDG0 region (De Bruijn insertion of Tn5 at the nucleotide sequence level.
et al., 1983), 28 Tn5 insertions in a plasmid carrying This does not appear to affect the usefulness of Tn5
the K. pneumoniae glnF(ntrA) gene (De Bruijn and in the generation of correlated physical and genetic
Ausubel, 1983), 61 Tn5 insertions in the cloned maps.
R. meliloti glnA (GSI) region (De Bruijn, F.J. and
Ausubel, F.M., in preparation), 88 Tn.5 insertions in (e) Polarity of TnS-induced insertion mutations
the cloned E. coli dnaG region (Lupski et al., 1982)
and 57 insertions in a plasmid carrying a cDNA Tn-induced insertion mutations generally exert a
fragment of Plasmodium knowlesi (see EXPERIMEN- strong polar effect on genes located distally (down-
TAL, section j; Lupski et al., 1983b), we have further stream) to their insertion site within the same operon
analyzed insertional specificity of Tn5. We found an due to transcriptional termination signals carried by
essentially random distribution pattern of Tn5 inser- the Tn element (see Kleckner, 1981). Transposon
tions into the variety of cloned DNA sequences listed Tn5 has been no exception to this rule (Berg et al.,
above and no evidence for the existence of prominent 1980). Therefore, TnS-induced insertion mutations
insertional hotspots, and encountered no difficulty have been employed extensively to determine the
generating Tn5 insertions every 50-100 bp in any of genetic organization of a variety of operons such as
the cloned DNA sequences mutagenized to date. the S. typhimurium his operon (Kleckner et al., 1975;
Our results confirm earlier experiments regarding the 1977; 1979), the E. coli ilv operon, the E. coli lac
insertional specificity of Tn5 made in a large number operon (Berg et al., 1980) and the complex
of other laboratories (see Garfinkel et al., 1981; K. pneumoniae nifgene cluster (Merrick et al., 1978;
Corbin et al., 1982; 1983; Ruvkun et al., 1982; Laird 1980; Riedel et al., 1979).
and Young, 1980; Shanabruch and Walker, 1980; However, in some instances incomplete polarity
Hakkaart et al., 1981). has been observed; for example, when analyzing
Even though Tn5 appears to insert randomly, or certain chromosomal TnS-induced insertion muta-
at least randomly enough to make it useful for in- tions in the E. coli lac operon (Berg et al., 1980).
sertion mutagenesis of cloned DNA sequences, at These authors observed that approximately one third
the nucleotide sequence level Tn5 may have some of the Tn5 insertions in the 1acZ gene resulted in
increased frequency of insertion into DNA se- constitutive, low-level expression of the downstream
quences that have homology to the Tn5 ends. We located lacy gene, while the remaining two thirds of
previously isolated Tn5 insertions into a recom- the 1acZ : : Tn5 insertions exerted a strictly polar
binant plasmid containing a cDNA insert coding for effect of lacy. The observed non-polar or partially
a Plasmodium knowlesi surface antigen (Lupski et al., polar phenotype of these 1acZ : : Tn5 insertion muta-
1983b). This cDNA insert has subsequently been tions was ascribed to the presence of promoter-like
shown to consist of 36 bp repeated in tandem eight DNA sequences at the ends of Tn5, initiating low-
times (Godson et al., 1983). By sequencing the level transcription of downstream genes, regardless
cDNA .. : Tn5 junctions of six Tn5 insertions we of the relative orientation of the Tn5 (Berg et al.,
have determined the exact location of the Tn5 inserts 1980).
into the individual repeats. All of the Tn5 inserts This effect is observed even more readily when
have been located within 10 nucleotides of the indi- analyzing TnS-induced insertion mutations in ope-
vidual repeats. whose DNA sequence appeared to rons (or segments thereof) that are carried by multi-
have some homology to the ends of Tn5. Three Tn5 copy plasmids. Strong, polar effects of such TnS-
inserts have been located between the same nucleo- induced insertion have been observed in the case of
tides in different repeats and thus they occupied the the cloned ent gene cluster of E. coli (Laird and
same relative position (Lupski, et al., 1983a). It has Young, 1980), the uvrB locus of E. coli (Pannekoek
been shown previously that Tn5 appears to integrate et al., 1980), the cloned glnALG(glnAR) operon of
with increased frequency into sequences which show K. pneumoniae (De Bruijn and Ausubel, 1981; De
some degree of homology to the end of the Tn5 Bruijn, 1983), and a plasmid carrying the cloned
141

K. pneurnoniae glnF(ntrA) gene (De Bruijn and Ausu- et al., 1980), or insertion of Tn5 DNA sequences
bel, 1983). However, in the case of the cloned may result in some cases in the creation of a
hisDG0 region of K. pneumoniae, hisG : : Tn5 inser- fortuitous promoter at the Tn5 : : target junction
tion mutations were shown not to exert a polar effect site, which would result in expression of the down-
on the hisD gene located immediately downstream stream gene(s). The fact that transcription originat-
on the same plasmid (De Bruijn et al., 1983). This ing from “outward-reading” promoters carried by
result was unexpected in the light of previous find- the Tn element is usually not detectable, may be due
ings regarding the genetic organization of the his to termination of such messages by the rho factor;
operon in enteric bacteria, as determined by trans- only when the transposon is located so near to a
poson mutagenesis (Kleckner et al., 1975; 1977; ribosome-binding site that rho-dependent termi-
1979; Ciampi et al., 1982; Schmid and Roth, 1983). nation is prevented will expression of downstream
In addition, in the case of the Tn5 mutagenesis ana- genes be observed (Schmid and Roth, 1983).
lysis of the cloned dnaG region of E. coli (Lupski Regardless of the explanation for the lack of Tn5
et al., 1982), a number of Tn5 insertions that were polarity in some of the cases, caution ought to be
subsequently shown to map in the region between the exercised when using Tn5 mutagenesis of operons
dnaG promoter and its structural gene (Lupski et al., carried on multicopy plasmids to determine the
1983~) also failed to abolish dnaG expression, sug- genetic organization of such cloned regions. Tn5-
gesting lack of Tn5 polarity. In both of these cases, induced insertion mutations will generally exert a
the lack of polarity effect was shown to be indepen- strongly polar effect on downstream genes within the
dent of the relative orientation of the Tn5, unlike the same operon, but this effect may be masked by
orientation-dependent polarity effects observed with genetic “artifacts” introduced by the use of multicopy
IS2 (Saedler et al., 1974; Pilacinski et al., 1977; plasmids.
Sommer et al., 1979). Also, insertions of TnlO into
an E. coli ribosomal RNA operon (Morgan, 1980) (f) Stability of TnS-induced insertion mutations
and TnlO insertions in the his operon (Ciampi et al.,
1982; Schmid and Roth, 1983) have been shown to Tn elements frequently induce DNA rearrange-
be incompletely polar. ments, such as deletions, duplications and inver-
These examples of lack of Tn5 polarity, or expres- sions, usually with one endpoint at the site of inser-
sion of genes within an operon downstream of the tion (for a review see Kleckner, 1981). This pheno-
Tn5 insertion site, can be explained in a variety of menon would complicate any interpretation of re-
different ways. First, one can invoke the presence of sults obtained with Tn-induced insertion mutations.
a low-level promoter, responsible for the expression In addition, secondary transposition events, result-
of the gene(s) downstream of the Tn5 insertion site, ing in the insertion of a copy of the Tn element at a
whose action is not observed when the operon is new site in the genome of the cell or into any plasmid
present in single copy in the cell. Alternatively, the it may harbour, would also complicate the interpre-
supercoiled nature of multicopy plasmids carrying tation of results obtained from transposon muta-
cloned regions of interest may result in elevated, genesis experiments. It is therefore important to con-
randomly initiated transcription of plasmid DNA sider the stability of TnS-induced insertion mutations
sequences, allowing sufficient expression of a gene in our experimental system. Regarding TnS-induced
carried on the plasmid, to observe genetic comple- DNA rearrangements, either upon insertion or sub-
mentation. Third, the absence of polarity associated sequently, we have observed gross alterations of the
with TnS-induced insertion mutations in operons target plasmid DNA sequences in less than 1% of
carried on multicopy plasmids may be the result of the TnS-containing plasmids from any given Tn5-
a paucity of factors such as rho that are essential for mutagenesis experiment, as determined by restric-
Tn-mediated termination of transcription (see tion analysis. When strains harboring TnS-mutated
Kleckner, 198 1). Fourth, in some operons there may plasmids are stored in agar stabs in the presence of
exist recognition sequences for an endogenous anti- kanamycin, l-5% of the plasmid DNAs undergo
termination factor. Finally, Tn5 itself may carry DNA rearrangements. We therefore usually store
minor promoters within its inverted repeats (Berg our plasmid-containing strains as glycerinated cul-
tures at - 20” C or - 70” C and use the antibiotic ing out gene-replacement experiments (Ruvkun and
resistance marker carried by the plasmid vector for Ausubel, 1981) and even incorporating TnS-
selection when growing up fresh cultures for genetic containing DNA sequences into the genome of
complementation tests or plasmid DNA prepara- higher plants via the A. turnefaciens infection system
tions. This reduces the frequency of rearrangements (Gartinkel et al., 1981; Matzke and Chilton, 1981;
of plasmid DNA sequences to less than 1’;. Kahl and Schell, 1982).
Restriction analysis of TnS-containing plasmids of
course does not rule out the possibility of small DNA (g) Use of TnS-induced insertion mutations to identify
rearrangements at the site of insertion. However, proteins and to determine the direction of transcrip-
DNA sequencing of the TnS-target junction se- tion of cloned genes
quences (Lupski et al., 1983a) in nine independent
isolates has shown that not one single base was Tn5 contains translational stop signals in all three
deleted at the insertion and in each case 9 bp of reading frames within the terminal 30 bp of its
target sequence were duplicated as has been pre- inverted repeats (Auerswald et al., 1981; Giphart-
viously shown (Auerswald et al.. 1981; Bossi and Gassler et al., 1982). Therefore, Tn5 insertions can
Ciampi, 198 1). be used to identify gene products and to determine
Regarding the occurrence of secondary Tn5 trans- the direction of transcription. The polypeptides
position events, or the presence of two independent synthesized by plasmids can be examined for
Tn5 insertions into a single plasmid, we have rarely example in maxicells (Sankar et al., 1979) or mini-
encountered any such cases. However, when analys- cells (Reeve, 1979). Since TnS-mutated genes will
ing TnS-induced chromosomal insertion mutations direct the synthesis of truncated polypeptides, a cor-
in K. pneunzoniae, strains carrying two or more relation between the physical location of Tn5 inser-
copies of Tn5 have been encountered after con- tions and the observed IV,. of the truncated poly-
tinuing selection for Km’ (Meade, H., Brown, S., De peptides yields information regarding the direction of
Bruijn, F. and Ausubel, F., unpublished observa- transcription of the cloned gene and the approximate
tions). This phenomenon may be due to increased translational start site. This methodology has been
Tn5 transposition frequencies and/or altered regu- employed to identify the products of genes such as
lation of Tn5 transposition in this organism (see also the bacteriophage PI yes gene (Heilmann et al.,
Merrick et al., 1980). A slight tendency of Tn5 to 1980), the phage Mu S and Ugenes (Giphart-Gassler
transpose from its original site of insertion to a new et al., 1982) the nifgene clusters of K. pneumoniae.
locus has also been observed in E. coli (Harayama R. rneliloti and R. japonicum (Puhler and Klipp,
et al., 1979). However, generally Tn5 appears to reg- 1981; Pi.ihler et al., 1983; Fuhrmann and Hennecke,
ulate its own transposition with an efficient, trans- 1982) the CloDF13 Hgene (Hakkaart et al., 1981)
acting negative regulator of transposition of the resi- and the K. pneumoniae glnF(ntrA) gene (De Bruijn
dent, or an incoming Tn5 (Biek and Roth, 1980; and Ausubel, 1983). The only problem encountered
Reznikoff, 1982; Berg, 1983). appears to be the relative instability of truncated
Thus, even though some reports of instability of polypeptides, which is observed especially in maxi-
TnS-induced insertion mutations above the observed cells (De Bruijn, F., unpublished observations).
reversion frequency of lo’-10” (Berg, 1977) exist,
such mutations are generally stable, nonleaky, null (h) Tn.5 mutagenesis of cosmid derivatives
mutations and usually do not generate extensive
DNA rearrangements or secondary transpositions. We have observed some anomalous results while
The stability is sufficient to allow transductional mutagenizing derivatives of the pLAFR1 cosmid
mapping in a variety of bacterial species (see vector (Friedman et al., 1982) with Tn5 using the
Kleckner et al., 1977; Forrai et al., 1983; Shana- /1: : Tn5 phage. The pLAFR1 vector consists of the
bruch and Walker, 1981; Harayama et al., 1979; low-copy-number pRK290 replicon (Tc’; Ditta
Fouts and Barbour, 1981; Kuner and Kaiser, 1981) et al., 1980) and a DNA fragment carrying the A cos
re-cloning of Tn5 containing DNA sequences by site. When RecA- E. co/i cells harboring a
Km’ selection (see EXPERIMENTAL. section i), carry- pLAFRl-derivative plasmid were infected with
143

I : : Tn5, the resulting Km’ colonies were shown to clones should contain a portion of that gene. This
contain mostly “cointegrate” plasmids between the recombinant is then used as a probe to screen a
pLAFR1 derivative and the defective I : : Tn5 library that contains wild-type sequences and geno-
genome, as opposed to simple, Tn.5 containing, mic DNA fragments carrying the gene of interest are
pLAFR1 derivatives. Plasmid DNA prepared from isolated.
such colonies failed to transform competent cells of The absence of an EcoRI restriction site in Tn5
E. coli to Tc’ and Km’; moreover, extensive killing of facilitates this procedure. EcoRI restriction frag-
the recipient cells was observed (De Bruijn, F. and ments containing TnS-mutagenized genes are easily
Ausubel, F., unpublished observations). We postu- cloned either by insertional inactivation of the Cm’
lated that the apparent recombination event between gene of pBR325 or by using one of the more recently
the pLAFR1 derivative and the 1: : Tn5 phage developed direct-selection vectors. In particular, we
genome, taking place in a RecA - strain, might be have found pKY2700 (Ozaki et al., 1982) very useful.
due to ATer-mediated recombination at the I cos This plasmid contains the colicin El structural gene
sites of the pLAFR1 and the 1: : Tn5. The observed with a unique EcoRI site within the gene. The colicin
inability to obtain Tc’ Km’ transformants with the El gene is inducible by the SOS system (Ebina et al.,
resulting cointegrate plasmid DNA could be ex- 1982). Therefore, transformation into a LexA-
plained by zygotic induction of the 2 upon entry into strain kills all cells except those which contain a
the recipient cell, resulting in killing of such cells. We plasmid carrying a defective colicin El gene, due to
tested this hypothesis by doing the initial 2 : : Tn5 insertional inactivation by a cloned EcoRI restriction
infection experiment in an E. coli strain lysogenic for fragment (Ozaki, L., personal communication).
2, and observed a dramatic increase in genuine Tn5 The above approach has already been used
transpositions (resulting in a pLAFR1 : : Tn5 plas- successfully by a number of investigators. A Cuufo-
mid), presumably due to repression of ATer- batter crescentus gene cluster specifying flagellum
mediated cos-cos recombination. Thus when muta- production has been isolated by Purucker et al.
genizing cosmid derivatives using the ,? : : Tn5 phage (1982), using nonmotile TnS-insertion mutants. Iso-
vector, it may be necessary to carry out the initial lation of the E. coli tolC gene was performed by
infection experiment using a strain capable of sup- Morona and Reeves (1982) in a similar manner using
pression of A functions, and harboring the cosmid. a TnlO-insertion mutant and selecting for Tc’.

(i) Use of Tn5 mutagenesis to clone genes of interest (j) Tn5 mutagenesis analysis of eukaryotic genes
that do not have a readily selectable phenotype
In vivo transposition mapping with Tn5 is not
When a gene is difficult to clone, either because the limited to the study of prokaryotic gene structure and
only mutants available are conditional lethal mutants function. The only requirement for this methodology
that display leaky phenotypes or the phenotype is is that a piece of DNA expresses a measurable
one for which there is no easy screening procedure, phenotype in bacteria. With the advent of cDNA
then transposition mutagenesis followed by drug cloning and nucleotide synthesis many eukaryotic
marker selection may be useful. One limitation is that genes have been cloned and expressed in E. coli. One
it must be performed in bacteria in which Tn5 trans- way of detecting phenotypic expression involves the
position occurs. The organism is subjected to use of monoclonal antibodies directed against the
random mutagenesis with Tn5. After mutagenesis a protein product of these cDNA recombinant clones.
Km’ colony is isolated which has lost the phenotype Transposition mapping with Tn5 may be used to
of the gene of interest or in which the Km’ marker further localize on a cloned DNA fragment the
is found closely linked to the gene. The DNA from genetic information encoding an enzymatic activity
this strain is then shotgun-cloned into a plasmid or antibody reactivity. Thus, this may be a way of
vector, such as pBR322, competent cells are trans- rapidly determining an immunodominant region of a
formed, and Km’ transformants are selected. If the protein.
original mutagenized strain contained a Tn5 insert This is particularly applicable to synthetic vac-
into the gene of interest then Km’ recombinant cines, either bacterial, viral or parasitic. Here the
144

gene for a surface antigen protein involved in the their ability to complement the mammalian cell
infectious process is first cloned, sequenced, and marker. Theoretically, clusters of Tn5 insertions into
expressed in bacteria. Then, rather than synthesize a the cloned fragment should be found which are still
number of small peptides, the immunodominant able to complement the defective mammalian cells,
region of this surface antigen protein is identified and these would correspond to Tn5 insertions within
using transposition mapping and a monoclonal anti- intervening sequences that were spliced out during
body to the natural constituent as a probe. Cells RNA processing.
harboring plasmids with different Tn5 insertion
mutations are lysed and the lysates are screened for (k) Physical mapping of plasmid-borne transposon
their ability to bind monoclonal antibody in a RIA. Tn5 insertions
Insertion mutations which destroy the ability of the
monoclonal antibody to bind lysates identify a region A number of convenient restriction sites are avail-
of DNA that may encode antigenic portions of the able for the physical mapping of Tn5 insertions in
protein molecule. multicopy plasmids (see Fig. 2). When mapping Tn5
This approach has been applied to delineate an insertions in a large segment of cloned DNA, it is
immunodominant region of a surface antigen of the often convenient to first determine the approximate
malarial parasite Plasmodium knowlesi which is location of the insertions. For this purpose the
responsible for causing malaria in monkeys (Lupski restriction endonucleases which do not cleave Tn5
et al., 1983b). A cDNA clone, which expressed a are useful (see Fig. 2). For example, EcoRI was used
plactamase fusion polypeptide in E. coli that reacted to determine the approximate location of the Tn5
with a monoclonal antibody to the parasite surface insertions shown in Fig. 4. Since EcoRI does not
antigen (Ellis et al., 1983), was mutagenized with cleave Tn5, the presence ofTn5 in any of the pFB5 14
Tn5, and lysates were screened for their ability to EcoRI fragments leads to the disappearance of the
bind antibody. A few insertion mutations destroyed corresponding band on the gel (X bp) and the
the ability of the lysates to bind antibody, and appearance of a new band corresponding to a DNA
restriction analysis of these Tn.5-mutated plasmids size of X + 5700 bp (De Bruijn and Ausubel,
identified within the cDNA the approximate region 1981). For precise mapping, the restriction endo-
coding for the epitope. Thus, the above experiment nucleases that cleave within the IRS of Tn5 are most
demonstrates that any gene, prokaryotic or useful (see Fig. 2). These sites allow orientation-
eukaryotic, can be mapped by this method provided independent localization of Tn5. A restriction endo-
a phenotype is readily detectable in bacteria. nuclease from this group which cleaves the original
The insertion of Tn5 into cloned eukaryotic DNA plasmid once (or twice), provides a reference point
sequences has also been used to introduce mutations relative to which the Tn5 insertions will be mapped.
whose effects are observable as an altered phenotype The TnS-containing plasmids are cleaved with the
in mammalian cells. A cloned DNA fragment desired restriction endonuclease and the fragments
containing the human adenovirus type 5 transform- run on a gel. An example is shown in Fig. 3, where
ing gene was subjected to Tn5 mutagenesis in E. coli Hind111 was used to map the Tn5 insertions. It
(McKinnon et al., 1982). Individual insertion mu- cleaves the parental plasmid once and Tn5 twice,
tants were transfected into mammalian cells and within the IRS, 1195 bp away from the ends of the
assayed for their ability to transform. In this manner transposon (see Fig. 2; Jorgenson et al., 1979;
adenovirus sequences essential for DNA-mediated Auerswald et al., 1981). Thus, three fragments on the
transformation were identified. TnS-containing plasmids are generated by cleavage
One might also be able to map intervening sequen- with Hind111 (see Fig. 3). By measuring the sizes of
ces of eukaryotic genes by Tn5 mutagenesis. A the smallest junction fragments and subtracting
cloned genomic DNA fragment able to complement 1195 bp (Tn5 DNA sequence), the distance of that
some mutation in mammalian cells would be sub- Tn5 insertion to the reference site can be calculated,
jected to Tn5 mutagenesis in E. coli. The individual and a physical map constructed (see Fig. 4 for an
insertion mutations would then be transfected into example). To determine the relative orientation of
the appropriate mammalian cells and checked for Tn5 on the plasmid, the Sal1 and SmaI sites within
145

Tn5 can be used since these sites are located asym- inserted. Thus, since the physical location of the Tn.5
metrically within the transposon (see Fig. 2). is known, this locates within the Tn.5 mapped DNA
The physical location of a gene as determined by fragment, the nucleotide sequence obtained. The
transposon T&mutagenesis mapping correlates method can be further generalized with the synthesis
very well with the actual location as determined by of a synthetic primer consisting of the terminal 15-20
nucleotide sequence analysis. For example, the nucleotides of Tn5.
E. coli dnaG gene was physically mapped by (1) the
isolation of multicopy plasmids containing restric-
tion fr~ents that could complement a condi-
tionally lethal dnaGts chromosomal marker at non- CONCLUSIONS
permissive 4O”C, followed by (2) TnS-mutagenesis
mapping of these recombinant plasmids and (3) In this paper we have reviewed and examined a
genetic analysis of individual Tn5 inserts into dnaG- number of parameters of transposon Tn5 muta-
cont~~g plasmids harbored in a dvraGts genesis as applied to cloned DNA segments carried
background at 30°C and 40°C (Lupski et al., 1982). by multicopy plasmids, such as transposition fre-
This correlated physical and genetic map was sub- quency and insertional specificity of Tn5, stability
sequently shown to be within 50 bp of the actual and polarity of TnS-induced insertion mutations and
location for dnaG as determined by nucleotide Tn.5 carrying vectors.
sequence analysis (Smiley et al., 1982). Based on the physical mapping of at least 500
independent Tn5 insertions in seven different cloned
(I) Use of transposon Tn5 mutagenesis in directed DNA segments we have carried out, and similar
nucleotide sequencing experiments carried out in a number of other labora-
tories, we conclude that the insertional specificity of
Tn5 insertion mutations obtained in thegeneration Tn.5 is very low; no gross “insertional hot-spots”
of correlated physical and genetic maps can also be were found, and we did not encounter difficulties
very useful for determining the nucleotide sequence generating large collections of well-distributed Tn5
of a particular DNA segment. Having mapped an insertions in any of the genes and operons examined.
individual Tn5 insertion one can take a directed, as We have found the A: : Tn5 vector (Berg et al.,
opposed to a “shotgun”, approach to nucleotide 197.5; Berg, 1977; Kleckner, N., unpublished obser-
sequencing. The presence of a unique HpaI site vations) to be extremely useful in the generation of
185 bp from the end of Tn5 (Auerswald et al., 198 1) Tn5-induced insertion mutations in cloned DNA
is very helpful for this purpose. A plasmid containing segments carried by multicopy plasmids, due to the
Tn5 in a known position is digested with @aI and efficient recovery of cells in which Tn5 transposition
another enzyme, X (X = BarnHI, SalI, Pst I, IiindIII and concomitant loss of the 1: : Tn5 vector DNA
or any enzyme that leaves blunt ends), which cuts the sequences have occurred.
cloned DNA. This fragment mixture is ligated into We have found the physical mapping of Tn5-
M13mp9 (Messing et al., 1981), digested with SmaI induced insertion mutations in multicopy plasmids
plus enzyme X, competent cells are transformed and to be greatly facilitated by the occurrence of a number
transformants screened by hybridization with a of convenient restriction sites in Tn5, useful for the
plasmid probe that contains only the terminal 185 bp determination of the physical location of Tn5
of Tn5 to identify subclones of interest. DNA (F 100 bp), the determination of the relative orien-
sequence analysis using the Sanger et al. (1977) tation of the Tn5 and the determination of the
dideoxy technique is then carried out by priming of nucleotide sequence of the TnS-target junction sites.
the Ml3mp9 recombinants with synthetic Ml3 Moreover, the availability of rapid, small-scale plas-
primer. Extension of the primer with DNA poly- mid DNA preparation protocols makes possible the
merase I (Klenow fra~ent) yields DNA sequences physical mapping of large numbers of plasmid-bone
that extend through known Tn5 sequence into Tn5 insertions with relative ease.
unknown sequence. This “unknown sequence” cor- Lastly, we have found plasmid-borne TnS-induced
responds to the DNA into which the Tn5 originally insertion mutations to be essentially stable and there-
fore useful for a number of purposes other than leucine-valine auxotrophs in Escherichia coli K-12: Evidence

physical and genetic mapping, such as subcloning for an internal promoter in the ilvOGEDA operon. Genetics
93 (1979) 309-319.
and gene-replacement experiments, and eventually
Berg, D.E.: Insertion and excision of the transposable kanamycin
for directed DNA sequencing of cloned genes. determinant Tn5, in: Bukhari, AI., Shapiro, J.A. and Adhya,
Thus we conclude that the Tn5 mutagenesis proto- S.L. (Eds.), DNA Insertion Elements, Plasmids and Epi-
col, described in MATERIALS AND METHODS, somes, Cold Spring Harbor Laboratory, Cold Spring Harbor.
section d, and discussed above, is extremely useful in NY, 1977. pp. 205-212.
Berg. D.E.: Control of gene expression by a mobile recombi-
the rapid generation of correlated physical and
national switch. Proc. Natl. Acad. Sci. USA 77 (1980)
genetic maps of cloned DNA segments carried by
4880-4884.
multicopy plasmids. As such, it constitutes an Berg. D.E.: Structural requirement for ISSO-mediated gene
important step in the analysis of cloned DNA seg- transposition. Proc. Natl. Acad. Sci. USA 80 (1983) 792-796.
ments by allowing a rapid identification of the loca- Berg, D.E. and Berg. CM.: The prokaryotic transposable ele-

tion, approximate boundaries and genetic as well as ment Tn5. Biotechnology 1 (1983) 417-435.
Berg, D.E.. Davies, J., Allet, B. and Rochaix, J.D.: Transposition
transcriptional organization of cloned genes and
of R factor genes to bacteriophage lambda. Proc. Natl. Acad.
operons. Sci. USA 72 (1975) 3628-3632.
Berg, DE., Egner, C. and Lowe, J.B.: Mechanism of F factor-
enhanced excision of transposon Tn5. Gene 22 ( 1983) 1-7.
Berg, D.E., Jorgensen, R. and Davies. J.: Transposable kanamy-
tin-neomycin resistance determinants. in Schlessinger, 0.
ACKNOWLEDGEMENTS
(Ed.). Microbiology - 1978, American Society for Microbiol-
ogy. Washington, DC, 1978, pp. 13-15.
We would like to thank all those individuals who Berg, D.E., Weiss, A. and Crossland, L.: Polarity ofTnS insertion
have helped us by discussing and partaking in our mutations in Escherichiu coli. J. Bacterial. 142 (1980) 439346.
Tn.5 mutagenesis experiments. In particular we Beringer. J.E., Beynon, J.L.. Buchanan-Wollaston, A.V. and
Johnston, A.W.B.: Transfer of the drug-resistance trsns-
thank Drs. Fred Ausubel, Peter D’Eustachio, G.
poson Tn5 to Rhkobium. Nature 276 (1978) 633-634.
Nigel Godson and Luiz Ozaki for helpful dis- Bevan, M.W. and Chilton, M.-D.: T-DNA of the ilgwbac.kWm
cussions and Dr. Ahmad I. Bukhari for discussing Ti and Ri plasmids. Annu. Rev. Genct. 16 (1982) 357-384.
and reviewing the m~usc~pt. This work was sup- Biek, D. and Roth. J.R.: RegulatioI~ of TnS transpositi~~xl in
ported by NSF Grant PAM-8104193 to Fred M. ~u~~~o~~ell~ f~phiFrzii~iuf~7. Proc. Natl. Acad. Sci. USA 77

Ausubel and NIAID Grant 7-l 142-996 to G. Nigel (1080)6047-6051.


Birnboim. H.C. and Doiy, J.: A rapid extraction procedure for
Godson. JRL is a recipient of an N.I.H, Medical
screening recombinant plasmid DNA. Nucl. Acids Res. 7
Scientist in Training Grant. (1979) lS13-1523.
Bolivar, F.. Rodriguez, R.L.. Greene. P.J.. Betlach. M.C.,
Heynecker, H.L., Boyer, H.W., Cross, J.H. and Falkow, S.:
Construction and characterization of new cloning vehicles,
II. A multipurpose cloning system. Gene 2 ( 1977) 9%113.
REFERENCES Bossi, L. and Ciampi. M.S.: DNA sequence of the sites of three
insertions of the transposable element Tn5 into the histidine
Auerswald, E.A., Ludwig, G. and Schaller, H.: Structural ana- operon of ~alrn~~ze~lu. Mol. Gen. Genet. 183 (1981) 406-408.
lysis of Tn5. Cold Spring Harbor Symp. Quant. Biol. 45 Bricker. J., Mulks, M.H., Plaut, A.G., Moxon. E.R. and Wright,
(1981) 107-I 13. A.: IgAl proteases of ~ae~zo~~jl~s j~?~~e~~ffe: cloning and
Ausubel, F.M.: Molecular genetics ofsymbiotic nitrogen fixation. characterization in Escherichia coli K-12. Proc. Natl. iicad.
Cell 29 (1982) l-2. Sci. LISA 80 (1983) 2681-2685.
Beck. E., Ludwig, G., Auerswald, E.A., Reiss. B. and Schaller, Buchanan-Wollaston, A.V.. Beringer, J.E.. Brewin, N.J.. Hirsch.
H.: Nucleotide sequence and exact localization of the P.R. and Johnston, A.W.B.: Isolation of symbiotically dcfec-
neomycin phosphotransferase gene from transposon Tn5. tive mutants in Rhirobium leguminosanmz by insertion of the
Gene 19 (1982) 327-336. transposon Tn5 into a transmissable plasmid. Mol. Gcn.
Belas, R., Mileham, A.. Cohn, D., Hilmer, M., Simon, M. and Genet. 178 (1980) 185-190.
Silverman, M.: Bacterial bioluminescence: isolation and Ciampi. M.S., S&mid, M.B. and Roth. J.R.: Transposon Tnfo
expression of the luciferase genes from Vibrio Harvey?. Science provides a promoter for transcription of adjacent sequences.
218 (1982) 791-793. Proc. Natl. Acad. Soi. US4 79 (1982) 5016-5020.
Berg. C.M., Shaw, K.J., Vender, J. and Borucha-.M~kie~vicz. Cleweli. D.B. and Hehnski. D.R.: Supercoiled circular DNA-
M.: Physiological characterization of polar TnZ-induced iso- protein complex in ~~che~~~,hj~ coli: Purification and induced
147

conversion to an open circular form. Proc. Nat]. Acad. Sci. cloning vector and its use in the genetic analysis of Rhizobium
USA 62 (1969) 1159-1166. mutants. Gene 18 (1982) 289-296.
Corbin, D., Barran, L. and Ditta, G.: Organization and expres- Fuhrmann, M. and Hennecke, H.: Coding properties of cloned
sion of Rhizobium meliloti nitrogen fixation genes. Proc. Natl. nitrogenase structural genes from Rhizobium japonicum. Mol.
Acad. Sci. USA 80 (1983) 3005-3009. Gen. Genet. 187 (1982) 419-425.
Corbin, D., Ditta, G. and Helinski, D.R.: Clustering of nitrogen Gantotti, V.B., Kindle, K.L. and Beer, S.V.: Transfer of the
fixation (nif) genes in Rhizobium meliloti. J. Bacterial. 149 drug-resistance transposon Tn5 to Erwinia herbicola and the
(1982) 221-228. induction of insertion mutations. Curr. Microbial. 6 (1981)
De Bruijn, F.J.: Regulation of the Klebsiellupneumoniae nitrogen 377-381.
fixation (nr$) genes by the ntr genes. Ph.D. Thesis, Harvard Gartinkel, D.J. and Nester, E.W.: Agrobacterium tumefaciens
University, Cambridge, MA, 1983. mutants affected in crown gall tumorigenesis and octopine
De Bruijn, F.J. and Ausubel, F.M.: The cloning and transposon catabolism. J. Bacterial. 144 (1980) 732-743.
Tn5 mutagenesis of the gbtA region of Klebsiella pneumoniae: Gartinkel, D.J., Simpson, R.B., Ream, L.W., White, F.F.,
Identification ofglnR, a gene involved in the regulation of the Gordon, M.P. and Nester, E.W.: Genetic analysis of crown
nifand hut operons. Mol. Gen. Genet. 183 (1981) 289-297. gall: tine structure map of the T-DNA by site directed muta-
De Bruijn, F.J. and Ausubel, F.M.: The cloning and charac- genesis. Cell 27 (1981) 143-153.
terization of the glnF(ntrA) gene of Klebsiella pneumoniae: Giphart-Gassler, M., Plaskerk, R.H.A. and Van de Putte, P.:
Role of glnF[ntrA) in the regulation of nitrogen fixation (nil) G-inversion in bacteriophage Mu: a novel way of gene splic-
and other nitrogen assimilation genes. Mol. Gen. Genet. ing. Nature 297 (1982) 339-342.
192(1983)342-353. Godson, G.N.. Ellis, J., Svec, P., Schlessinger, D.H. and Nussen-
De Bruijn, F.J., Stroke, I.L., Marvel, D.J. and Ausubel, F.M.: zweig, V.: Identification and chemical synthesis of an epitope
Construction of a correlated physical and genetic map of the of Plusmodium knowlesi circumsporozoite protein. Evidence
Klebsiella pneumoniae hisDG0 region using transposon Tn5 for its tandemly repeated nature. Nature 305 (1983) 29-33.
mutagenesis. EMBO J. 2 (1983) 1831-1838. Guarente, L.P., Isberg, R.R., Sylvanen, M. and Silhavy, T.J.:
Ditta, G., Stantield, S., Corbin, D. and Helinski, D.R.: Broad Conferral of transposable properties to a chromosomal gene
host range DNA cloning system for gram-negative bacteria. in Escherichiu coli. J. Mol. Biol. 141 (1980) 235-248.
Construction of a gene bank of Rhizobium meliloti. Proc. Natl. Hakkaart, M.J.J., Veltkamp, E. and Nijkamp, H.J.J.: Protein H
Acad. Sci. USA 77 (1980) 7347-7351. encoded by plasmid CloDF 13 involved in lysis ofthe bacterial
Duncan, M.J.: Properties of T&induced carbohydrate mutants host, I. Localization of the gene and identification and sub-
in Rhizobium meliloti. J. Gen. Microbial. 122 (1981) 61-67. cellular localization of the gene H product. Mol. Gen. Genet.
Ebina, Y., Kishi, F. and Nakasawa, A.: Direct participation of 183 (1981) 318-325.
LexA protein in repression of colicin El synthesis. J. Bac- Harayama, S., Palva, E.T. and Hazelbauer, G.L.: Transposon
teriol. 150 (1982) 1479-1481. insertion mutants of Eschenchia coli K-12 defective in a com-
Egner, C. and Berg, D.E.: Excision of transposon Tn.5 is depen- ponent common to galactose and ribose chemotaxis. Mol.
dent on the inverted repeats but not on the transposase Gen. Genet. 171 (1979) 193-203.
function of Tn5. Proc. Nat]. Acad. Sci. USA 78 (1981) Heilmann, H., Burkardt, H.J., Puhler, A. and Reeve, J.N.: Trans-
459-463. poson mutagenesis of the gene encoding the bacteriophage
Ellis, J., Ozaki, L.S., Gwadz, R.W., Cochrane, A.H., Nussen- Pl restriction endonuclease. J. Mol. Biol. 144 (1980) 387-396.
zweig, V., Nussenzweig, R.S. and Godson, G.N.: Cloning and Holmes, D.S. and Quigley, M.: A rapid boiling method for the
expression in E. coliofthe malarial sporozoite surface antigen preparation of bacterial plasmids. Anal. Biochem. 114 (1981)
gene from Plasmodium knowlesi. Nature 302 (1983) 536-538. 193-197.
Ely, B. and Croft, R.H.: Transposon mutagenesis in Caulobacter Hooykaas, P.J.J., Van Brussel, A.A.N., Den Dulk-Ras, H., Van
crescentus. J. Bacterial. 149 (1982) 620-625. Slogteren, G.M.S. and Schilperoort, R.A.: Sym plasmid of
Engebrecht. J., Nealson, K. and Silverman, M.: Bacterial bio- Rhizobium trifoliiexpressed in different rhizobial species and
luminescence: isolation and genetic analysis of functions Agrobacterium tumefaciens. Nature 291 (1981) 35 l-353.
from VibrioJischeri. Cell 32 (1983) 773-781. Ish-Horowitz, D. and Burke, J.F.: Rapid and efficient cosmid
Fitts, R.A. and Taylor, A.L.: Integration of bacteriophage Mu at cloning. Nucl. Acids Res. 9 (1981) 2989-2998.
host chromosome replication forks during lytic development. Johnson, R.C., Yin, J.C.P. and Reznikoff, W.S.: Control of Tn5
Proc. Natl. Acad. Sci. USA 77 (1980) 2801-2805. transposition in Escherichiu coli is mediated by protein from
Forrai. T., Vincze, E., Banfalvi, Z., Kiss, G.B., Randhawa, G.S. the right repeat. Cell 30 (1982) 873-882.
and Kondorosi, A.: Location of symbiotic mutations in Rhizo- Johnston, A.W.B., Beynon, J.L., Buchanan-Wollaston, A.V.,
bium meliloti. J. Bacterial. 153 (1983) 635-643. Setchell, S.M., Hirsch, P.R. and Beringer, J.E.: High fre-
Fouts, K.E. and Barbour, S.D.: Transductional mapping of ksgB quency transfer of nodulating ability between strains and
and a new T&induced kasugamycin-resistance gene, ksgD, species of Rhizobium. Nature 276 (1978) 634-636.
in Escherichiu coh K-12. J. Bacterial. 145 (1981) 914-919. Jorgensen, R.A., Rothstein, S.J. and Reznikoff, W.S.: A restric-
Friedman, A.M., Long, S.R., Brown, S.E., Buikema, W.J. and tion enzyme cleavage map of Tn5 and location of a region
Ausubel, F.M.: Construction of a broad host range cosmid encoding neomycin resistance. Mol. Gen. Genet. 177 (1979)
65-72.
14x

Kahl, G. and &hell, J.S.: Molecular Biology of Plant Tumors, McKinnon, R.D., Bacchetti, S. and Graham, F.L.: Tn5 muta-
Academic Press. New York, 1982. genesis of the transforming genes of human adenovirus type
Kleckner, N.: Transposable elements in prokaryotes. Annu. Rev. 5. Gene 19 (1982) 33-42.
Genet. 15 (1981) 341-404. Meade, H.M., Long, S.R.. Ruvkun, G.B., Brown, S.E. and
Kleckner, N., Chan, R.K., Tye, B.K. and Botstein, D.: Muta- Ausubel, F.M.: Physical and genetic characterization of
genesis by insertion of a drug resistance element carrying an symbiotic and auxotrophic mutant of Rhizobium meliloti
inverted repetition. J. Mol. Biol. 97 (1975) 561-575. induced by transposon Tn5 mutagenesis. J. Bacterial. 149
Kleckner, N., Roth, J. and Botstein, D.: Genetic engineering in (1982) 114-122.
vivo using translocatable drug-resistance elements: New Merrick, M., Filser. M., Dixon, R.. Elmerich, C., Sibold. L. and
methods in bacterial genetics. J. Mol. Biol. 116 (1977) Houmard, J.: The use of translocatable genetic elements to
125-159. construct a line-structure map of the Klebsiellu pneumoniae
Kleckner, N., Steele, D.A., Reichardt, K. and Botstein, D.: nitrogen fixation (nif) gene cluster. J. Gen. Microbial. 117
Specificity of insertion by the translocatable tetracycline (1980) 509-520.
resistance element TnlO. Genetics 92 (1979) 1023-1040. Merrick, M., Filser, M., Kennedy, C. and Dixon, R.: Polarity of
Klee. H.J., White, F.F.. Iyer, V.N., Gordon, M.P. and Nester. mutations induced by insertion of transposons Tn5, Tn7. and
E.W.: Mutational analysis of the virulence region of an TnlO into the nifgene cluster of Klebsiella pneumoniue. Mol.
Agrobacterium fumefaciens Ti plasmid. J. Bacterial. 153 (1983) Gen. Genet. 165 (1978) 103-111.
878-883. Messing, J., Crea, R. and Seeburg, P.H.: A system for shotgun
Kuner, K.M. and Kaiser. D.: Introduction of transposon Tn.5 DNA sequencing. Nucl. Acids Res. 9 (1981) 309-321.
into M~.XOCOCCUS
for analysis of developmental and other Meyer, R., Both, G. and Shapiro, J.A.: Transposition of DNA
nonselectable mutants. Proc. Natl. Acad. Sci. USA 78 (1981) inserted into deletions of the Tn5 kanamycin resistance
425429. element. Mol. Gen. Genet. 171 (1979) 7-13.
Laird, A.J. and Young, LG.: Tn5 mutagenesis of the enterocholin Miller, J.H.: Experiments in Molecular Genetics. Cold Spring
gene cluster of Escherichia coli. Gene 11 (1980) 359-366. Harbor Laboratory, Cold Spring Harbor, NY. 1972.
Lamb, J.W., Hombrecher. G. and Johnson, A.W.B.: Plasmid- Miller, J.H., Calos, M.P., Galas. D., Hofer. M., Biichel. D.E. and
determined nodulation and nitrogen-fixation abilities in Rhi- Miiller-Hill. B.: Genetic analysis of transposition in the luc
zobium phaseoli. Mol. Gen. Genet. 186 (1982) 449-452. region of Escherichiu coli. J. Mol. Biol. 144 (1980) 1-18.
Leong, S.A., Ditta. G.S. and Helinski. D.: Heme biosynthesis in Morgan, E.A.: Insertions ofTnJ0 into an E. coli ribosomal RNA
Rhbobium identification of a cloned gene coding for &amino operon are incompletely polar. Cell 21 (1980) 257-265.
levulinic acid synthetase from Rhizobium meliloti. J. Biol. Morona, R. and Reeves, P.: Molecular cloning of the tolC locus
Chem. 257 (1982) 8724-8730. ofEscherichia coli K-l 2 with the use of transposon TnlO. Mol.
Lupski. J.R.. Gershon, P., Ozaki, L.S. and Godson, G.N.: Gen. Genet. 184 (1981) 430-434.
Specificity of Tn5 insertions into a 36 nucleotide DNA Ozaki, L.S.. Kimura. A.. Shimada, K. and Takagi. Y.: ColEl
sequence repeated in tandem eight times. (1983a), submitted. vectors for direct selection of cells carrying a hybrid plasmid.
Lupski, J.R., Ozaki, L.S.. Ellis, J. and Godson. G.N.: Locali- J. Biochem. 91 (1982) 1155-1162.
zation of the Plusmodium surface antigen epitope by Tn5 Palva. E.T., Liljestrom. P. and Harayama, S.: Cosmid cloning
mutagenesis mapping of a recombinant cDNA clone. Science and transposon mutagenesis in Salmonellu typhimurium using
220 (1983b) 1285-1288. phage i vehicles. Mol. Gen. Genet. 181 (1981) 153-157.
Lupski, J.R.. Smiley, B.L. and Godson, G.N.: Regulation of the Pannekoek. H.. Hille, J. and Noordermeer, I.: Relief of polarity
rpslJ-dmzG-rpoD macromolecular synthesis operon and the caused by transposon Tn5: application in mapping a cloned
initiation of DNA replication in Escherichiu coli K-12. Mol. region of the Escherichiu coli uvrB locus essential for UV
Gen. Genet. 189 (1983~) 48-57. resistance. Gene 12 (1980) 51-61.
Lupski, J.R., Smiley, B.L., Blattner. F.R. and Godson, G.N.: Phua. S.H., Bergquist, P.L. and Lane. H.E.D.: Effects of Tn.5
Cloning and characterization of the Escherichiu coli chromo- insertion in the incD region on mini-F maintenance and poly-
somal region surrounding the dnaC gene, with a correlated peptide synthesis. Mol. Gen. Genet. 188 (1982) 353-355.
physical and genetic map of dnaG generated via transposon Pilacinski, W., Mosharrafa. E., Edmundson. R.. Zissler, J..
Tn5 mutagenesis. Mol. Gen. Genet. 185 (1982) 120-128. Fiandt, M. and Szybalski. W.: Insertion sequence IS2 asso-
Ma, Q.S., Johnston, A.W.B.. Hombrecher. G. and Downie, J.A.: ciated with int-constitutive mutants ofbacteriophage lambda.
Molecular genetics of mutants of Rhkobium leguminosarum Gene 2 (1977) 61-74.
which fail to fix nitrogen, Mol. Gen. Genet. 187 (1982) Portier. C.: Isolation of a polynucleotide phosphorylase mutant
166-171. using a kanamycin resistant determinant. Mol. Gen. Genet.
Maniatis. T., Fritsch. E.F. and Sambrook, J.: Molecular Cloning, 178(1980)343-349.
A Laboratory Manual. Cold Spring Harbor Laboratory, Cold Piihler, A. and Klipp. W.: Fine structure analysis of the gene
Spring Harbor, NY, 1982. region for N,-fixation (nif ofKlebsiellupneumoniae. in Bothe.
Matzke, A.J.M. and Chilton, M.-D.: Site specific insertion of H. and Trebst, A. (Eds.). Biology of Inorganic Nitrogen and
genes into the T-DNA of the Agrobacterium tumor-inducing Sulfur. Springer Verlag, Heidelberg, 1981, pp. 276-286.
plasmid: An approach to genetic engineering of higher plant Ptihler. A.. Klipp, W. and Weber. G.: Mapping and regulation of
cells. J. Mol. Appl. Genet. 1 (1981) 39-49. the structural genes niJK. nifD and tlifH of Rhirobium meliloti.
149

in: Ptihler, A. (Ed.), Molecular Genetics ofthe Bacteria-Plant Schell, M.A.: Cloning and expression in Escherichiu coli of the
Interaction. Springer-Verlag, Heidelberg, in press. naphthalene degradation genes from plasmid NAH7. J.
Purcell, B.K. and Clegg, S.: Construction and expression of Bacterial. 153 (1983) 822-829.
recombinant plasmids encoding type 1 limbriae of a urinary Schmid, M.B. and Roth, J.R.: Internal promoters of the his
Klebsiellu pneumoniue isolate. Infect. Immun. 39 (1983) operon in Salmonella typhimurium. J. Bacterial. 153 (1983)
1122-l 127. 1114-1119.
Purucker, M., Bryan, R., Amemiya, K., Ely, B. and Shapiro, L.: Scott, K.F., Hughes, J.E., Gresshoff, P.M., Beringer, J.E., Rolfe,
Isolation of a Cuulobacter gene cluster specifying flagellum B.C. and Shine, J.: Molecular cloning of Rhizobium tn$lii
production by using nonmotile Tn5 insertion mutants. Proc. genes involved in symbiotic nitrogen fixation. J. Mol. Appl.
Natl. Acad. Sci. USA 79 (1982) 6797-6801. Genet. 1 (1982) 315-326.
Putnoky, P., Kiss, G.B., Ott, I. and Kondorosi, A.: Tn5 carries Shanabruch, W.G., Behlau, I. and Walker, G.C.: Spontaneous
a streptomycin-resistance determinant downstream from the mutations of Salmonella typhimurium LT2 generated by
kanamycin-resistance gene. Mol. Gen. Genet. 191 (1983) insertion of transposable elements. J. Bacterial. 147 (1981)
288-294. 827-835.
Reeve, J.: Use ofminicells for bacteriophage directed polypeptide Shanabruch, W.G. and Walker, G.C.: Localization of the plas-
synthesis, in Wu, R. (Ed.), Methods in Enzymology, Vol. 68, mid (pKMlO1) gene(s) involved in recA ‘1exA +-dependent
Academic Press, New York, 1979, pp. 493-503. mutagenesis. Mol. Gen. Genet. 179 (1980) 289-297.
Reznikoff, W.S.: Tn5 transposition and its regulation. Cell 34 Shaw, K.J. and Berg, C.M.: Escherichia coli K-12 auxotrophs
(1982) 362-368. induced by insertion of the transposable element Tn5.
Riedel, G.E.: The use of molecular cloning to study the Genetics 92 (1979) 741-744.
organization and expression of the nitrogen fixation (aif) Shaw, K.J., Berg, CM. and Sobol, T.: Salmonella typhimurium
genes of Klebsiella pneumoniae. Ph.D. Thesis, Harvard Uni- mutants defective in acetohydroxy acid synthetase I and II.
versity, Cambridge, MA, 1980. J. Bacterial. 141 (1980) 1258-1263.
Riedel, G.E., Ausubel, F.M. and Cannon, F.C.: Physical map of Shimkets, L.J., Gill, R.E. and Kaiser, D.: Developmental cell
chromosomal nitrogen fixation (nif genes of Klebsiellu interactions in ~Uyxococcusxunthus and the spoC locus. Proc.
pneumoniae. Proc. Natl. Acad. Sci. USA 76 (1979) 2866-2870. Natl. Acad. Sci. USA 80 (1983) 1406-1410.
Rolfe, B.C., Gresshoff, P.M. and Shine, J.: Rapid screening for Silverman. M. and Simon, M.: Phase variation: genetic analysis
symbiotic mutants of Rhizobium and white clover. Plant Sci. of switching mutants. Cell 19 (1980) 845-854.
Lett. 19 (1980) 277-284. Simon, R., Priefer, U. and Ptihler, A.: Vector plasmids for in vivo
Ruvkun, G.B. and Ausubel, F.M.: A general method for site- and in vitro manipulations of Gram-negative bacteria, in
directed mutagenesis in prokaryotes. Nature 289 (1981) Ptihler, A. (Ed.), Molecular Genetics of the Bacteria-Plant
85-88. Interaction. Springer-Verlag, Heidelberg, 1983, pp. 98-106.
Ruvkun, G.B., Sundaresan, V. and Ausubel, F.M.: Directed Smiley, B.L., Lupski, J.R., Svec, P.S., McMacken. R. and
transposon Tn5 mutagenesis and complementation analysis Godson, G.N.: Sequences of the Escherichia coli dnaG
ofRhizobium melilotisymbiotic nitrogen fixation genes. Cell 29 primase gene and regulation of its expression. Proc. Natl.
(1982) 551-559. Acad. Sci. USA 79 (1982) 4550-4554.
Saedler, H., Reif, H.J., Hu, S. and Davidson, N.: IS2, a genetic Sommer, H., Cullum, J. and Saedler, H.: ISZ-43 and ISZ-44: New
element for turn-off and turn-on ofgene activity in Escherichiu alleles of the insertion sequence IS2 which have promoter
coli. Mol. Gen. Genet. 132 (1974) 265-269. activity. Mol. Gen. Genet. 175 (1979) 53-56.
Sanger, F., Nicklen, S. and Coulson, A.R.: DNA sequencing with Sprent, J.I.: The Biology ofNitrogen Fixing Organisms. McGraw-
chain-terminating inhibitors. Proc. Natl. Acad. Sci. USA 74 Hill, London, 1979.
(1977) 5643-5647. Van Vliet, F., Silva, B., Van Montagu, M. and Schell, J.: Transfer
Sankar, A., Hack, A.M. and Rupp, W.D.: Simple method for of RP4 : : Mu plasmids to Agrobacterium tumefuciens. Plasmid
identification of plasmid-coded proteins. J. Bacterial. 137 1 (1978) 446-455.
(1979) 692-693. Walton, D.A. and Moseley,B.E.B.: Inducedmutagenesis inRhizo-
Sasakawa, C. and Yoshikawa, M.: Transposon (Tn_5)-mediated bium frifolii. J. Gen. Microbial. 124 (1981) 191-195.
suppressive integration of ColEl derivatives into the chromo-
some ofEscherichia coli K-12 (dnaA). Biochem. Biophys. Res.
Commun. 96 (1980) 1364-1370. Communicated by A.I. Bukhari.
ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, Apr. 2006, p. 1282–1286 Vol. 50, No. 4
0066-4804/06/$08.00⫹0 doi:10.1128/AAC.50.4.1282–1286.2006
Copyright © 2006, American Society for Microbiology. All Rights Reserved.

In Vitro Analysis of ISEcp1B-Mediated Mobilization of Naturally


Occurring ␤-Lactamase Gene blaCTX-M of Kluyvera ascorbata
Marie-Frédérique Lartigue, Laurent Poirel, Daniel Aubert, and Patrice Nordmann*
Service de Bactériologie-Virologie, Hôpital de Bicêtre, Assistance Publique/Hôpitaux de Paris, Faculté de Médecine Paris-Sud,
Université Paris XI, 94275 K-Bicêtre, France
Received 14 November 2005/Returned for modification 13 December 2005/Accepted 11 January 2006

ISEcp1B has been reported to be associated with and to mobilize the emerging expanded-spectrum ␤-lac-
tamase blaCTX-M genes in Enterobacteriaceae. Thus, the ability of this insertion sequence to mobilize the
blaCTX-M-2 gene was tested from its progenitor, Kluyvera ascorbata. Insertions of ISEcp1B upstream of the
blaCTX-M-2 gene in K. ascorbata reference strain CIP7953 were first selected with cefotaxime (0.5 and 2 ␮g/ml).
In those cases, ISEcp1B brought promoter sequences enhancing blaCTX-M-2 expression in K. ascorbata. Then,

Downloaded from http://aac.asm.org/ on June 5, 2015 by guest


ISEcp1B-mediated mobilization of the blaCTX-M-2 gene from K. ascorbata to Escherichia coli J53 was attempted.
The transposition frequency of ISEcp1B-blaCTX-M-2 occurred at (6.4 ⴞ 0.5) ⴛ 10ⴚ7 in E. coli. Cefotaxime,
ceftazidime, and piperacillin enhanced transposition, whereas amoxicillin, cefuroxime, and nalidixic acid did
not. Transposition was also enhanced when studied at 40°C.

Increasing worldwide reports of expanded-spectrum ␤-lac- of the blaCTX-M-14/-18, blaCTX-M-17, and blaCTX-M-19 ␤-lacta-
tamases of the CTX-M type in Enterobacteriaceae and mostly mase genes (4, 6, 24). Recently, we have shown that ISEcp1B
in Escherichia coli raise the question of their way of acquisition is able to mobilize the adjacent blaCTX-M-19 gene by a trans-
(4, 31). These enzymes are now widespread not only in noso- positional mechanism in Escherichia coli by recognizing a va-
comial but also in community-acquired pathogens (4, 23). The riety of DNA sequences as right inverted repeats (IRR) (26).
40 CTX-M-type ␤-lactamases may be grouped into five main Chromosome-encoded ␤-lactamases of several Kluyvera spe-
subgroups according to amino acid sequence identity (CTX- cies have been identified as progenitors of CTX-M-derived en-
M-1, -M-2, -M-8, -M-9, and -M-25) (1, 4, 13, 15, 27, 29). Most zymes. The CTX-M-1 and CTX-M-2 subgroups are derived from
CTX-M enzymes hydrolyze cefotaxime better than ceftazi- Kluyvera ascorbata (12, 28), whereas the CTX-M-8 and CTX-M-9
dime. However, the latest reported enzymes, including CTX- subgroups are derived from Kluyvera georgiana (21, 25).
M-15 (13), hydrolyze ceftazidime better than cefotaxime and The aim of this study was to experimentally evaluate the
are also widespread (32). It has been shown that different ability of ISEcp1B to mobilize a chromosome-encoded ␤-lac-
genetic elements are associated with blaCTX-M genes. ISEcp1- tamase gene from its reservoir, K. ascorbata, to a plasmid
like insertion sequences are most frequently reported (5, 7, 11, location in Escherichia coli. The effects of addition of different
13, 29). This insertion sequence element has been found to be antibiotics (mostly ␤-lactams) and of growth at various tem-
associated with four out of the five blaCTX-M gene clusters peratures were also tested.
(CTX-M-1, -M-2, -M-9, and -M-25 clusters) (1, 4, 13, 15, 27,
29). Nevertheless, the DNA sequence that separates the ␤-lac- MATERIALS AND METHODS
tamase gene from ISEcp1 varies within a given cluster of CTX-M
Bacterial strains. Clinical strain Klebsiella pneumoniae ILT-3 (expressing the
genes, indicating that different insertion events may have oc- blaCTX-M-19 gene associated with ISEcp1B) has been described previously (24).
curred (16). Moreover, several plasmid-encoded cephalospori- Kluyvera ascorbata CIP7953 reference strain, the recombination-deficient strain
nase genes, such as the blaCMY- or blaACC-type genes, may be E. coli DH5␣ (harboring pOX38-Gen, a self-conjugative, IS-free, and gentami-
associated also with the same ISEcp1-like element (2, 18). cin-resistant plasmid), and the azide-resistant E. coli J53 were used for transpo-
sition and conjugation experiments (10, 17). The low-copy-number cloning vec-
ISEcp1 is weakly related to other IS elements and belongs to
tor pBBR1MCS.3 was used for cloning experiments (14). Bacterial cells were
the IS1380 family (IS Database home page [http://www-is grown in Trypticase soy (TS) broth or onto TS agar plates (Sanofi Diagnostics
.biotoul.fr/page-is.html]) (8). Since ISEcp1-like elements are Pasteur, Marnes-La-Coquette, France) with antibiotics when required.
located upstream of several ␤-lactamase genes, analysis of the Antimicrobial agents and susceptibility testing. Routine antibiograms were
variable sequences separating these IS elements from initiation determined by the disk diffusion method on Mueller-Hinton agar (Sanofi Diagnos-
tics Pasteur). The antimicrobial agents and their sources have been referenced
codons of these genes allowed us to determine its boundaries. elsewhere (22). The antibiotic concentrations used for selection were as follows:
ISEcp1B possesses two imperfect inverted repeats (IR) likely cefotaxime (CTX; 0.5 and 2 ␮g/ml), amoxicillin (AMX; 100 ␮g/ml), tetracycline
made of 14 bp, with 12 of these 14 bp being complementary (TET; 15 ␮g/ml), kanamycin (KAN; 30 ␮g/ml), and gentamicin (GEN; 7 ␮g/ml).
(Table 1), and a gene encoding a 420-amino-acid transposase. Nucleic acid extraction. Recombinant plasmids and pOX38-Gen derivative
plasmids were extracted using QIAGEN Plasmid Midi kits and the very-low-copy
ISEcp1B brings promoter sequences for high-level expression
plasmid purification protocol, respectively (QIAGEN, Courtaboeuf, France).
Extraction of whole-cell DNA was done as described elsewhere (22).
PCR experiments. PCR experiments were performed as previously described
* Corresponding author. Mailing address: Service de Bactériologie- (30). The entire ISEcp1B gene was amplified using the primers preTnCTXM-1
Virologie, Hôpital de Bicêtre, 78 rue du Général Leclerc, 94275 K.- (5⬘-CTAACAGAGCTTAAGCTTCC-3⬘) and preISEcp1-2 (5⬘-CTCCCAATAC
Bicêtre, France. Phone: 33-1-45-21-36-32. Fax: 33-1-45-21-63-40. E-mail: GGTCAATCCG-3⬘) and subsequently cloned into the SmaI site of plasmid
nordmann.patrice@bct.ap-hop-paris.fr. pBBR1MCS.3.

1282
VOL. 50, 2006 ISEcp1B-MEDIATED MOBILIZATION OF THE blaCTX-M GENE 1283

TABLE 1. Sequences identified as IRR boundaries after Cloning experiments and sequencing. T4 DNA ligase and restriction endo-
ISEcp1B transposition nucleases were used according to the recommendations of the manufacturer
(Amersham Biosciences, Orsay, France). The recombinant plasmid pISE was
No. of base constructed by inserting the PCR product of the ISEcp1B gene into the SmaI site
Size of
pairs
Description of Nucleotide sequence transposed of plasmid pBBR1MCS.3, which was then electroporated into electrocompetent
identical to
sequence (5⬘33⬘)a fragment Kluyvera ascorbata CIP7953 cells, as previously described (22), and selection was
perfect
(bp) performed on TET (15 ␮g/ml)-containing plates (Fig. 1). In order to study the
IRR
transposition of ISEcp1B, an omega fragment (⍀Km) from plasmid pHP45⍀-
IRL of ISEcp1B GATTCTACGTCAGT Km, made of a kanamycin resistance gene [aph(3⬘)-IIa] flanked by transcrip-
Deduced perfect ACGTAGAATCTAGG tional and translational termination sequences, was introduced into ISEcp1B.
IRR of The recombinant plasmid pISE was digested by NsiI enzyme (into ISEcp1B
ISEcp1B between the stop codon of the transposase gene and the IRR). The digested
IRR of ISEcp1B ACGTGGAATTTAGG 12 plasmid was mixed with an EcoRI-restricted ⍀Km fragment (2.2 kb) in order to
IRR-1 CGTCATATAGCTGG 4 5,464
create the tagged insertion sequence ISEcp1B.Kan, yielding the plasmid
IRR-2 ATATGGATAAGGAG 5 3,450
pISEcp1B.Kan.
IRR-3 CTTTGTAAAGAACG 5 3,951
Sequencing of the insert was performed using laboratory-designed primers on
IRR-4 GAGAAGAAAATGGG 8 3,582
an ABI PRISM 3100 automated sequencer (Applied Biosystems, Les Ulis,
IRR-5 GCTCTTTTTTCTGG 4 2,667
France).
a
Underlined nucleotides correspond to those identified at the same positions Transposition experiments. Several transposition experiments were per-

Downloaded from http://aac.asm.org/ on June 5, 2015 by guest


in the deduced IRR of ISEcp1B. formed to determine (i) the mobility of ISEcp1B alone (i.e., without the ␤-lac-

FIG. 1. Schematic representation of ISEcp1B-mediated mobilization of the naturally occurring CTX-M-2 ␤-lactamase gene of Kluyvera
ascorbata CIP7953. 1. Introduction of ISEcp1B in K. ascorbata by electroporation. 2. Selection of K. ascorbata strains with ISEcp1B inserted
upstream of the blaCTX-M-2 gene. ISEcp1B enhances the gene expression by bringing promoter sequences. 3. Transfer of recipient plasmid in
cefotaxime-resistant K. ascorbata strains. 4. ISEcp1B mobilizes the blaCTX-M-2 gene on the plasmid under various conditions: with or without
amoxicillin, piperacillin, cefuroxime, cefotaxime, ceftazidime, or nalidixic acid at 22, 30, 37, or 40°C. 5. Plasmid-located events of transposition are
isolated. White arrow with black dots, ISEcp1B; black arrow with white dots, blaCTX-M.
1284 LARTIGUE ET AL. ANTIMICROB. AGENTS CHEMOTHER.

tamase gene), (ii) the ability of ISEcp1B to mobilize a chromosome-encoded sequences enhancing blaCTX-M expression. The transposition
␤-lactamase gene from K. ascorbata to a plasmid location in E. coli by transpo- of ISEcp1B generated a 5-bp duplication that was located at
sition, and (iii) the effects of antibiotics and of temperature on the transposition
events. The transposition of ISEcp1B.Kan onto the conjugative plasmid
various insertion sites in the chromosome of K. ascorbata
pOX38-Gen was investigated with a mating-out technique in liquid medium CIP7953 (TACTA, TAATA, and AATAC). Twenty transfor-
(22). The recombinant plasmid pISEcp1B.Kan was electroporated into E. coli mants were analyzed, including 11 obtained on agar plates
DH5␣(pOX38-Gen) for transposition experiments. Transfer of the recombinant containing 2 ␮g/ml of cefotaxime and 9 on plates with 0.5
plasmids with the pOX38 backbone into the E. coli J53 azide-resistant (AZr)
␮g/ml of cefotaxime. On one hand, detailed analysis of the
strain was then performed by conjugation. One colony obtained after 24 h of
growth was cultured under weak agitation in 1-ml of TS broth at 37°C for 3 h and target sites of transformants selected on CTX (2 ␮g/ml) re-
was used as a donor for the mating assay with E. coli J53 as recipient. Conjuga- vealed a preferential location 22 bp upstream of blaCTX-M-2
tion was done by incubating 800 ␮l of recipient and 200 ␮l of donor under low (64%), but other insertions were observed located 19 bp and 43
agitation at 37°C for an additional 3-h step. Mating was stopped by vigorous bp upstream of the blaCTX-M-2 gene. The insertion sites of most
vortexing and cooling on ice. The transconjugants were selected on agar plates
containing 7 ␮g per ml of GEN (plasmid marker), 30 ␮g per ml of KAN
of the transformants (five of nine) selected on CTX at 0.5
(transposon marker), and 100 ␮g per ml of azide (chromosomal marker). The ␮g/ml were located 19 bp upstream of the blaCTX-M-2 gene as
transposition frequency was calculated by dividing the number of transconju- the insertion site of ISEcp1B upstream of blaCTX-M-5, de-
gants by the number of donors. scribed on a natural plasmid (12). Moreover, ISEcp1 insertions
The MIC of cefotaxime for the wild-type K. ascorbata strain is 0.06 ␮g/ml. To
located 43 bp upstream of blaCTX-M have been identified up-

Downloaded from http://aac.asm.org/ on June 5, 2015 by guest


select for ISEcp1B upstream of blaCTX-M, K. ascorbata harboring the recombi-
nant plasmid pISE was screened on agar plates containing 0.5 or 2 ␮g of stream of blaCTX-M-9, blaCTX-M-14, blaCTX-M-17, and blaCTX-M-19
cefotaxime per ml after 24 h of growth in TS broth (Fig. 1). This strategy was (6, 7, 24, 29). Nevertheless, no ISEcp1 insertion has been
based on the previous observations demonstrating ISEcp1B mediated high-level identified 22 bp upstream of the blaCTX-M gene in clinical
expression of blaCTX-M ␤-lactamases genes, with the IS element providing pro- isolates.
moter sequences. The transposition frequency was calculated by dividing the
ISEcp1B-mediated transposition of blaCTX-M. A conjugation
number of CTXr transformants by the total number of bacteria. The second step
consisted of mobilization by ISEcp1B of the blaCTX-M-2 gene to the E. coli J53 was realized with the gentamicin-resistant plasmid pOX38-GEN
recipient strain. Plasmid pOX38-GEN was used as a target for transposition (GENr) (a transfer-proficient F plasmid derivative) as donor and
events. Transfer of pOX38-GEN into the K. ascorbata transformants was per- the K. ascorbata strain harboring ISEcp1B-blaKLUA as recipient
formed, and transconjugants were selected on agar plates containing 7 ␮g per ml (Fig. 1). Susceptibility to tetracycline was systematically observed
of GEN (plasmid marker) and 100 ␮g per ml of AMX (chromosomal marker)
(Fig. 1). Transposition events were searched for between the chromosomal
in transconjugant strains, ruling out full integration or cointegra-
blaCTX-M-2 gene and the recipient plasmid pOX38-GEN after overnight growth tion (TET resistance) of the recombinant plasmids derived from
in TS broth with and without antibiotics at different subinhibitory concentrations pBBR1MCS.3 into plasmid pOX38. The transposition ability of
and after 3 h of growth at various temperatures (22°C, 30°C, 37°C, and 40°C). ISEcp1B-blaKLUA was investigated by conjugation with the K.
Several structurally unrelated ␤-lactams were studied, since it is unknown
ascorbata strain, harboring ISEcp1B-blaKLUA and the plasmid
whether any ␤-lactam might enhance mobilization of the blaCTX-M-2 gene. Na-
lidixic acid was also studied, since quinolone resistance is frequently associated pOX38-GEN as donor, and E. coli J53 AZr as recipient (Fig. 1).
with extended-spectrum ␤-lactamase (ESBL)-mediated resistance in Entero- Transposition of the ISEcp1B-blaCTX-M-2 fragment occurred at a
bacteriaceae (23) and since quinolones are known to induce antibiotic resistance frequency of (6.4 ⫾ 0.5) ⫻ 10⫺7 in E. coli. Analysis of the genetic
through the SOS response (3). Transfer of the recombinant plasmids with the environment of several transposition events leading to insertions
pOX38-GEN backbone into the E. coli J53AZr strain was then performed by
conjugation, and transconjugants were selected on agar plates containing 7 ␮g
of ISEcp1B-blaCTX-M-2 revealed in all cases a 5-bp duplication
per ml of GEN (plasmid marker), 100 ␮g per ml of AMX (transposon marker), that very likely confirmed the acquisition of that structure through
and 100 ␮g per ml of azide (chromosomal marker) (Fig. 1). The transposition a transposition mechanism. These observations also indicated
frequency was calculated by dividing the number of transconjugants by the that ISEcp1B was able to mobilize by itself the blaCTX-M-2 gene;
number of donor bacteria. All the GENr AMXr AZr colonies were screened for
this was consistent with previous results (26).
tetracycline susceptibility to exclude those that may have resulted from nontrans-
position events. Target site preference of the ISEcp1B-blaCTX-M-2 transposon.
Insertion site determination. The regions where for insertions of ISEcp1B Several GENr AMXr AZr E. coli isolates from independent ex-
upstream of the blaCTX-M-2 gene were amplified by PCR and sequenced. Plasmid periments were analyzed. The insertion sites of ISEcp1B-blaKLUA
pOX38-GEN carrying various transposed structures was extracted and se- were determined by sequencing the neighboring regions of the
quenced in part.
inverted repeats. A 5-bp target site duplication, consistent with a
transposition event, was observed in all the studied insertion
events.
RESULTS AND DISCUSSION
To determine whether the ISEcp1B-blaKLUA transposon had
Transposition of ISEcp1B upstream of the chromosome- a target site preference, the locations of five insertion events
located blaCTX-M-2 gene. Selection of the K. ascorbata CIP7953 were mapped onto recombinant plasmid pOX38-GEN. Inser-
reference strain, in which ISEcp1B was inserted upstream of tions of ISEcp1B-blaKLUA sequences had occurred into five
the blaCTX-M-2 gene, was obtained at a frequency of (1 ⫾ 0.5) ⫻ different sites which were distantly located on the plasmid.
10⫺7 per donor, whereas the overall transposition frequency of Alignment of the insertion site sequences revealed variable se-
ISEcp1B.Kan was 10⫺6 (data not shown). Thus, transposition quences in recombinant plasmids (TATGA, TATCA, TACAT,
of ISEcp1B upstream of the chromosomal ␤-lactamase gene TATAC, and TTCAT). No consensus sequence was identified
occurred at a high frequency (10% of overall transposition among the 5-bp duplicated sites, whereas an AT-rich content
events of ISEcp1B.Kan). Among the K. ascorbata strains over- that may target ISEcp1B-mediated transposition was identified
expressing their blaCTX-M-2 gene, the ISEcp1B element was again (26).
inserted in such a manner that its transposase gene was tran- Characterization of ISEcp1B-blaCTX-M-2 transposons. Five
scribed in the same orientation as the ␤-lactamase gene. As different transposons were analyzed (Table 1). Their sizes var-
observed in clinical isolates, ISEcp1B brought promoter ied from 2,667 to 5,464 bp. ISEcp1B possesses two imperfect
VOL. 50, 2006 ISEcp1B-MEDIATED MOBILIZATION OF THE blaCTX-M GENE 1285

TABLE 2. IS transposition frequency with and without antibioticsa TABLE 3. IS transposition frequency at different temperaturesa
Transposition frequency Transposition frequency
Antibiotic (␮g/ml) Temp (°C)
(mean ⫾ SD) (mean ⫾ SD)

None ............................................................................ (4.3 ⫾ 2.3) ⫻ 10⫺7 22 ........................................................................... (4.9 ⫾ 2.8) ⫻ 10⫺7


Amoxicillin (256)........................................................ (3.4 ⫾ 2.3) ⫻ 10⫺7 30 ........................................................................... (6.8 ⫾ 5.8) ⫻ 10⫺7
Amoxicillin (128)........................................................ (4.2 ⫾ 3.8) ⫻ 10⫺7 37 ........................................................................... (6.4 ⫾ 0.5) ⫻ 10⫺7
Amoxicillin (52).......................................................... (5.2 ⫾ 2.5) ⫻ 10⫺7 40 ........................................................................... (2.6 ⫾ 2.2) ⫻ 10⫺6
Cefuroxime (128) ....................................................... (6.5 ⫾ 8.2) ⫻ 10⫺7 a
Cefuroxime (64) ......................................................... (4.6 ⫾ 3.9) ⫻ 10⫺7 In each case, three independent experiments were performed and the aver-
age and standard deviation were calculated.
Cefuroxime (26) ......................................................... (2.0 ⫾ 0.4) ⫻ 10⫺7
Cefotaxime (8)............................................................ (6.6 ⫾ 3.8) ⫻ 10⫺6
Cefotaxime (4)............................................................ (1.9 ⫾ 1.4) ⫻ 10⫺6
Cefotaxime (1.5)......................................................... (5.4 ⫾ 1.4) ⫻ 10⫺7
mented conditions, ceftazidime seemed to enhance the transpo-
Ceftazidime (0.5) ....................................................... (5.2 ⫾ 2.7) ⫻ 10⫺5
Ceftazidime (0.25) ..................................................... (5.6 ⫾ 3.2) ⫻ 10⫺6 sition of ISEcp1B at a higher level than cefotaxime. No difference
Ceftazidime (0.1) ....................................................... (6.2 ⫾ 2.7) ⫻ 10⫺7 of transposition frequency was found with ceftazidime (0.1 ␮g/
Piperacillin (64).......................................................... (2.9 ⫾ 2.8) ⫻ 10⫺6 ml), cefotaxime (1.5 ␮g/ml), or piperacillin (32 and 13 ␮g/ml).
Piperacillin (32).......................................................... (6.5 ⫾ 7.4) ⫻ 10⫺7
Although amoxicillin and cefuroxime are widely prescribed for

Downloaded from http://aac.asm.org/ on June 5, 2015 by guest


Piperacillin (16).......................................................... (2.8 ⫾ 2.8) ⫻ 10⫺7
Nalidixic acid (2)........................................................ (7.4 ⫾ 1.3) ⫻ 10⫺7 community-acquired infections and cefuroxime has been de-
Nalidixic acid (1)........................................................ (2.9 ⫾ 1.4) ⫻ 10⫺7 scribed as a risk factor in selection of ESBLs in the community
Nalidixic acid (0.4)..................................................... (2.9 ⫾ 1.4) ⫻ 10⫺7 (9), our results indicated that, under the experimental conditions
a
In each case, three independent experiments were performed and the mean described, those ␤-lactams might not select for those transposi-
and standard deviation were calculated. tion events. By contrast, cefotaxime and ceftazidime may induce
those transposition events, but they are not given frequently for
treating community-acquired gram-negative infections.
IRs likely made of 14 bp, with 12 of these 14 bp being com- Nalidixic acid, which may induce the SOS repair system and
plementary (Table 1) (26). A detailed analysis of the bound- recombination events (3), did not play a role in ISEcp1B-
aries of the transposed fragments identified five different IRR- related transposition events. Thus, although there is somewhat
like sequences downstream of the ␤-lactamase gene (Table 1). of a strong association between quinolone resistance and
These IRR-like sequences had been recognized by the trans- ESBL phenotype in Enterobacteriaceae (23), this result would
posase of ISEcp1B during the mobilization process. The num- indicate that quinolone might not enhance the transposition of
ber of identical base pairs among the sequences defined as the blaCTX-M gene. The association between quinolone resis-
IRR boundaries varied from 4 to 8 bp, corresponding to less
tance and ESBLs may rather result from clonal selection of
than 50% identity with the IRR of ISEcp1B. These results
specific strains by quinolone or expanded-spectrum cephalo-
indicated that ISEcp1B might use different sequences as IRRs
sporins.
downstream of the ␤-lactamase gene of the K. ascorbata chro-
The transposition frequency was increased at 40°C com-
mosome. No consensus sequence could be determined by com-
pared to that determined at 37°C, whereas no difference was
paring the 14-bp-long IRR sequences identified in recombi-
observed at 22°C and 30°C (Table 3). These results may em-
nant plasmids (Table 1). Nevertheless, a guanosine residue
phasize the induction of ISEcp1B transposition under high-
located at the 3⬘ end of these IRRs was always found, likely
temperature conditions, as recently observed for four IS ele-
indicating that this nucleotide was necessary in the transposi-
ments, IS401, IS402, IS406, and ISBmu3 in Burkholderia
tion process, as already reported (26).
Transposition frequencies under different growth condi- multivorans ATCC 17616 (20). This is the first report demon-
tions. Since changes of growth conditions may affect transpo- strating increased transposition frequencies at a high temper-
sition efficiency of several mobile elements (19), the transpo- ature for an IS belonging to the IS1380 family. This result
sition of ISEcp1B-blaKLUA was examined in the presence of contrasts with the decreased transposition activities of several
several concentrations of different antibiotics at different sub- mobile elements (e.g., Tn3, IS1, IS30, and IS911) in E. coli at
inhibitory concentrations and under different temperatures. this temperature (8, 19). The temperature sensitivity of trans-
Studied antibiotic concentrations were 1/2, 1/4, and 1/10 of the position in E. coli has been considered to be a feature of each
MICs. For K. ascorbata strain CIP7953, harboring ISEcp1B- transposase (8, 19). It is tempting to speculate that global
blaKLUA, the MICs were as follows: amoxicillin, 512 ␮g/ml; piperacil- warming may increase transposition of ISEcp1B. In addition,
lin, 128 ␮g/ml; cefuroxime, 512 ␮g/ml; cefotaxime, 16 ␮g/ml; although growth curves did not differ significantly for E. coli
ceftazidime, 1 ␮g/ml; nalidixic acid, 4 ␮g/ml. No significant dif- and K. ascorbata, a slight increase of growth of both donor
ference of transposition frequency was found with or without (Kluyvera sp.) and recipient (E. coli) at temperatures ranging
amoxicillin, cefuroxime, and nalidixic acid at the studied concen- from 22°C to 37°C may account also for dissemination of
trations (Table 2). In contrast, the transposition frequency of the blaCTX-M genes.
ISEcp1B-blaKLUA element was 100-fold higher when ceftazidime The reservoir of most blaCTX-M genes has been identified in
was added at 0.5 ␮g/ml (half of the MIC), 10-fold higher with Kluyvera spp. (12, 21, 25, 28). Their translocation process has been
ceftazidime at 0.25 ␮g/ml (one-quarter of the MIC) or cefotaxime studied here, which represents the first evidence of in vivo mobi-
at 8 ␮g/ml (half of the MIC), 7-fold higher with piperacillin at 64 lization of a clinically important antibiotic resistance gene from its
␮g/ml (half of the MIC), and 4-fold higher with cefotaxime at 4 natural reservoir. Thus, there is now an urgent need for identifi-
␮g/ml (one-quarter of the MIC) (Table 2). Under these experi- cation of the environmental location (if any) of Kluyvera spp.
1286 LARTIGUE ET AL. ANTIMICROB. AGENTS CHEMOTHER.

ACKNOWLEDGMENTS First description of CTX-M-15-producing Klebsiella pneumoniae in Turkey.


J. Antimicrob. Chemother. 52:315–316.
This work was financed by a grant from the Ministère de l’Education 16. Lartigue, M. F., L. Poirel, and P. Nordmann. 2004. Diversity of genetic
Nationale et de la Recherche (UPRES-EA3539), Université Paris XI, environment of the blaCTX-M genes. FEMS Microbiol. Lett. 234:201–207.
Paris, France, and mostly by the European Community (6th PCRD, 17. Martinez-Martinez, L., A. Pascual, and G. A. Jacoby. 1998. Quinolone re-
LSHM-CT-2003-503-335). L.P. is a researcher from the INSERM, sistance from a transferable plasmid. Lancet 351:797–799.
Paris, France. 18. Nadjar, D., M. Rouveau, C. Verdet, L. Donay, J. Herrmann, P. H. Lagrange,
A. Philippon, and G. Arlet. 2000. Outbreak of Klebsiella pneumoniae pro-
We thank T. Naas for precious advice.
ducing transferable AmpC-type ␤-lactamase (ACC-1) originating from
Hafnia alvei. FEMS Microbiol. Lett. 187:35–40.
REFERENCES 19. Nagy, Z., and M. Chandler. 2004. Regulation of transposition in bacteria.
Res. Microbiol. 155:387–398.
1. Baraniak, A., J. Fiett, W. Hryniewicz, P. Nordmann, and M. Gniadkowski. 20. Ohtsubo, Y., H. Genka, H. Komatsu, Y. Nagata, and M. Tsuda. 2005. High-
2002. Ceftazidime-hydrolysing CTX-M-15 extended-spectrum ␤-lactamase temperature-induced transposition of insertion elements in Burkholderia
(ESBL) in Poland. J. Antimicrob. Chemother. 50:393–396. multivorans ATCC 17616. Appl. Environ. Microbiol. 71:1822–1828.
2. Bauernfeind, A., I. Schneider, R. Jungwirth, H. Sahly, and U. Ullmann. 21. Olson, A. B., M. Silverman, D. A. Boyd, A. McGeer, B. M. Willey, V. Pong-
1999. A novel type of AmpC ␤-lactamase, ACC-1, produced by a Klebsiella Porter, V. Daneman, and M. R. Mulvey. 2005. Identification of a progenitor
pneumoniae strain causing nosocomial pneumonia. Antimicrob. Agents Che- of the CTX-M-9 group of extended-spectrum ␤-lactamases from Kluyvera
mother. 43:1924–1931. georgiana isolated in Guyana. Antimicrob. Agents Chemother. 49:2112–2115.
3. Beaber, J. W., B. Hochhut, and M. K. Waldor. 2004. SOS response promotes 22. Philippon, L. N., T. Naas, A. T. Bouthors, V. Barakett, and P. Nordmann.
horizontal dissemination of antibiotic resistance genes. Nature 427:72–74. 1997. OXA-18, a class D clavulanic acid-inhibited extended-spectrum ␤-lac-
4. Bonnet, R. 2004. Growing group of extended-spectrum ␤-lactamases: the tamase from Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 41:

Downloaded from http://aac.asm.org/ on June 5, 2015 by guest


CTX-M enzymes. Antimicrob. Agents Chemother. 48:1–14. 2188–2195.
5. Bou, G., M. Cartelle, M. Tomas, D. Canle, F. Molina, R. Moure, J. M. Eiros, 23. Pitout, J. D., P. Nordmann, K. B. Laupland, and L. Poirel. 2005. Emergence
and A. Guerrero. 2002. Identification and broad dissemination of the CTX- of Enterobacteriaceae producing extended-spectrum ␤-lactamases (ESBLs)
M-14 ␤-lactamase in different Escherichia coli strains in the northwest area in the community. J. Antimicrob. Chemother. 56:52–59.
of Spain. J. Clin. Microbiol. 40:4030–4036. 24. Poirel, L., J. W. Decousser, and P. Nordmann. 2003. Insertion sequence
6. Cao, V., T. Lambert, and P. Courvalin. 2002. ColE1-like plasmid pIP843 of ISEcp1B is involved in the expression and mobilization of a blaCTX-M ␤-lac-
Klebsiella pneumoniae encoding extended-spectrum ␤-lactamase CTX-M-17. tamase gene. Antimicrob. Agents Chemother. 47:2938–2945.
Antimicrob. Agents Chemother. 46:1212–1217. 25. Poirel, L., P. Kämpfer, and P. Nordmann. 2002. Chromosome-encoded
7. Chanawong, A., F. H. M’Zali, J. Heritage, J. H. Xiong, and P. M. Hawkey. Ambler class A ␤-lactamase of Kluyvera georgiana, a probable progenitor of
2002. Three cefotaximases, CTX-M-9, CTX-M-13, and CTX-M-14, among a subgroup of CTX-M extended-spectrum ␤-lactamases. Antimicrob. Agents
Enterobacteriaceae in the People’s Republic of China. Antimicrob. Agents Chemother. 46:4038–4040.
Chemother. 46:630–637. 26. Poirel, L., M. F. Lartigue, J. W. Decousser, and P. Nordmann. 2005.
8. Chandler, M., and J. Mahillon. 2002. Insertion sequences revisited, p. 305– ISEcp1B-mediated transposition of blaCTX-M in Escherichia coli. Antimicrob.
366. In N. L. Craig, R. Craigie, M. Gellert, and A. M. Lambowitz (ed.), Agents Chemother. 49:447–450.
Mobile DNA II. ASM Press, Washington, D.C. 27. Poirel, L., T. Naas, I. Le Thomas, A. Karim, E. Bingen, and P. Nordmann.
9. Colodner, R., W. Rock, B. Chazan, N. Keller, N. Guy, W. Sakran, and R. Raz. 2001. CTX-M-type extended-spectrum ␤-lactamase that hydrolyzes ceftazi-
2004. Risk factors for the development of extended-spectrum ␤-lactamase- dime through a single amino acid substitution in the omega loop. Antimi-
producing bacteria in nonhospitalized patients. Eur. J. Clin. Microbiol. In- crob. Agents Chemother. 45:3355–3361.
fect. Dis. 23:163–167. 28. Rodriguez, M. M., P. Power, M. Radice, C. Vay, A. Famiglietti, M. Galleni,
10. Derbyshire, K. M., L. Hwang, and N. D. Grindley. 1987. Genetic analysis of J. A. Ayala, and G. Gutkind. 2004. Chromosome-encoded CTX-M-3 from
the interaction of the insertion sequence IS903 transposase with its terminal Kluyvera ascorbata: a possible origin of plasmid-borne CTX-M-1-derived
inverted repeats. Proc. Natl. Acad. Sci. USA 84:8048–8053. cefotaximases. Antimicrob. Agents Chemother. 48:4895–4897.
11. Dutour, C., R. Bonnet, H. Marchandin, M. Boyer, C. Chanal, D. Sirot, and 29. Saladin, M., V. T. Cao, T. Lambert, J. L. Donay, J. L. Herrmann, Z. Ould-
J. Sirot. 2002. CTX-M-1, CTX-M-3, and CTX-M-14 ␤-lactamases from Hocine, C. Verdet, F. Delisle, A. Philippon, and G. Arlet. 2002. Diversity of
Enterobacteriaceae isolated in France. Antimicrob. Agents Chemother. 46: CTX-M ␤-lactamases and their promoter regions from Enterobacteriaceae
534–537. isolated in three Parisian hospitals. FEMS Microbiol. Lett. 209:161–168.
12. Humeniuk, C., G. Arlet, V. Gautier, P. Grimont, R. Labia, and A. Philippon. 30. Sambrook, J., and D. W. Russell. 2001. Molecular cloning: a laboratory
2002. ␤-Lactamases of Kluyvera ascorbata, probable progenitors of some manual, 3rd ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor,
plasmid-encoded CTX-M types. Antimicrob. Agents Chemother. 46:3045– N.Y.
3049. 31. Tzouvelekis, L. S., E. Tzelepi, P. T. Tassios, and N. J. Legakis. 2000. CTX-
13. Karim, A., L. Poirel, S. Nagarajan, and P. Nordmann. 2001. Plasmid-medi- M-type ␤-lactamases: an emerging group of extended-spectrum enzymes.
ated extended-spectrum ␤-lactamase (CTX-M-3-like) from India and gene Int. J. Antimicrob. Agents 14:137–140.
association with insertion sequence ISEcp1. FEMS Microbiol. Lett. 201:237– 32. Woodford, N., M. E. Ward, M. E. Kaufmann, J. Turton, E. J. Fagan, D.
241. James, A. P. Johnson, R. Pike, M. Warner, T. Cheasty, A. Pearson, S. Harry,
14. Kovach, M. E., R. W. Phillips, P. H. Elzer, R. M. Roop II, and K. M. J. B. Leach, A. Loughrey, J. A. Lowes, R. E. Warren, and D. M. Livermore.
Peterson. 1994. pBBR1MCS: a broad-host-range cloning vector. BioTech- 2004. Community and hospital spread of Escherichia coli producing CTX-M
niques 16:800–802. extended-spectrum ␤-lactamases in the UK. J. Antimicrob. Chemother. 54:
15. Lartigue, M. F., L. Poirel, C. Héritier, V. Tolün, and P. Nordmann. 2003. 735–743.
Appl Microbiol Biotechnol (1999) 51: 1±12 Ó Springer-Verlag 1999

MINI-REVIEW

H.-M. Tan

Bacterial catabolic transposons

Received: 13 July 1998 / Received revision: 22 September 1998 / Accepted: 26 September 1998

Abstract The introduction of foreign organic hydro- However, the degradation of xenobiotic chemicals by
carbons into the environment in recent years, as in the microorganisms has attracted more recent attention. A
widespread use of antibiotics, has resulted in the evo- wide variety of microorganisms in the environment can
lution of novel adaptive mechanisms by bacteria for the adapt to use xenobiotic chemicals as new sources of
biodegradation of the organic pollutants. Plasmids have growth and energy. Despite being isolated from geo-
been implicated in the catabolism of many of these graphically separated areas of the world, these microor-
complex xenobiotics. The catabolic genes are prone to ganisms are often found to possess similar specialized
undergo genetic rearrangement and this is due to their enzyme systems and metabolic pathways for the degra-
presence on transposons or their association with dation of synthetic organic compounds such as toluene,
transposable elements. Most of the catabolic transpo- naphthalene and chlorinated biphenyls. At the molecular
sons have structural features of the class I (composite) level, similarities in genetic organisation and nucleotide
elements. These include transposons for chlorobenzoate sequence suggest that discrete modules of catabolic genes
(Tn5271), chlorobenzene (Tn5280), the newly discovered may have been combined in di€erent fashions to generate
benzene catabolic transposon (Tn5542), and transpo- new catabolic pathways in bacteria. The tendency for
sons encoding halogenated alkanoates and nylon-oligo- catabolic genes to undergo genetic rearrangements, such
mer-degradative genes. Transposons for the catabolism as insertions, deletions, duplications and inversions, is
of toluene (Tn4651, Tn4653, Tn4656) and naphthalene attributable to the presence of elements that possess the
(Tn4655) belong to class II (Tn3 family) elements. Many ability to mobilise the catabolic genes.
catabolic genes have been associated with insertion se- Although many of the well-characterized bacterial
quences, which suggests that these gene clusters could be transposons were originally isolated from clinical sam-
rapidly disseminated among the bacterial populations. ples (Galas and Chandler 1989; Sheratt 1989), new ele-
This greatly expands the substrate range of the micro- ments and transposons associated with catabolic
organisms in the environment and aids the evolution of operons are being characterized from environmental
new and novel degradative pathways. This enhanced isolates, suggesting that bacterial catabolic transposons
metabolic versatility can be exploited for and is believed (Nakatsu et al. 1991; Springael et al. 1993; Tsuda et al.
to play a major part in the bioremediation of polluted 1989; van der Meer et al. 1991b) may be widespread and
environments. diverse in nature. It is clear that transposons are in-
volved in the rapid adaptation of bacterial communities
to aromatic compounds, just as is the case for antibi-
otics, and their frequent location on transmissible plas-
Introduction mids may facilitate their dissemination (Dahlberg and
Hermansson 1995). Such plasmids are usually very large
The role microorganisms play in the recycling of ele- (70±500 kb) and, where investigated, are often trans-
ments in the biosphere is a crucial one, albeit not new. missible (Table 1).
Transposons are discrete DNA segments that are able
H.-M. Tan to move in the absence of genetic homology from one
Department of Microbiology, genetic location (donor site) to another (target site)
National University of Singapore, (Berg and Howe 1989). This process requires a trans-
Lower Kent Ridge Road, 119260, Singapore
e-mail: mictanhm@nus.edu.sg
posase that is encoded by the genetic element itself. The
Tel.: +65-874-6407 transposase interacts with the ends of the transposon in
Fax: +65-776-6872 a site-speci®c manner, cuts the DNA at both ends of the
2

Table 1 A selection of catabolic plasmids from bacteria

Primary substrate Plasmid Size (kb) Organism Conjugative Reference

Benzene pHMT112 112 P. putida ML2 Tan and Mason 1990


Camphor PpG1 (CAM) 500 P. putida + Rheinwald et al. 1973; Frantz and Chakrabarty 1986
Chlorobenzene pP51 110 Pseudomonas sp. P51 + van der Meer et al. 1991a
3-Chlorobenzoate pAC25 117 P. putida AC858 + Chatterjee et al. 1981
pB13 (pWR1) 111 Pseudomonas sp. B13 + Chatterjee and Chakrabarty 1983
pBRC60 88 Alcaligenes sp. BR60 + Fulthorpe and Wyndham 1991
p-Cresol pRA4000 85 P. putida NCIB 9886 + Dean et al. 1989
Haloacetates pUO1 65 Moraxella sp. B + Kawasaki et al. 1981
Isopropylbenzene pRE4 105 P. putida RE204 + Eaton and Timmis 1986
pBD2a 210 R. erythropolis BD2 + Dabrock et al. 1994; Kesseler et al. 1996
Naphthalene NAH7 83 P. putida PpG7 + Dunn and Gunsalus 1973; Gunsalus et al. 1982
pNPL-41 P. putida + Boronin et al. 1980
pND 140 P. putida UB1-1 + Dunn et al. 1980
Nylon pOAD2 44 Flavobacterium sp. K172 Kato et al. 1994
2,4-Dichlorophenoxyacetate pJP4 80 A. eutrophus JMP134 + Don et al. 1985
Phenol pPGH1 200 P. putida H ) Hermann et al. 1987
Salicylate SAL 1 90 P. putida R1 + Gunsalus et al. 1982; Chakrabarty 1972
p-Sulphobenzoate pPSB 85 C. testosteroni PSB-4 + Junker and Cook 1997
p-Toluene sulphonate pTSA 85 C. testosteroni T-2 + Junker and Cook 1997
Toluene pWW0 (TOL) 117 P. putida mt-2 + Williams and Worsey 1976; Duggleby et al. 1977
pDKI 180 P. putida HS1 (PpCl) + Kunz and Chapman 1981
pTKO 150 P. putida PPK1 ) Keshavarz et al. 1985
pKJ1 225 Pseudomonas sp. TA8 + Yano and Nishi 1980;
1,2,4-Trimethylbenzene pGB 85 P. putida TMB Besteti and Galli 1987
3,5-Xylenol pRA500 500 P. putida NCIB9869 + Hooper and Kemp 1980; Jain et al. 1984
a
This is a linear plasmid that also encodes trichloroethane catabolism
3

element and proceeds with the strand-transfer reaction matic hydrocarbons and other organic compounds.
(Hallet and Sherratt 1997; Mizuuchi 1992). Newly isolated and other potential catabolic transpo-
Most transposable elements can be grouped into sons are also presented. The ecological potential of these
three classes depending on the genetic organisation, elements and their in¯uence on bioremediation are
DNA sequence homologies and mechanistic properties brie¯y discussed. There have been a number of recent
(Grindley and Reed 1995). related reviews of the structure and function of trans-
Class I elements include the insertion sequences (IS) posable elements (van der Meer et al. 1992; Wyndham
containing the genetic determinants for transposition et al. 1994a; Tsuda 1996).
only, and composite transposons that have an inter-
vening sequence with ¯anking IS constituents (Syvanen
1988). IS elements, which are normal constituents of Class I catabolic transposons
many bacterial chromosomes and plasmids, are distin-
guished from transposons in that they do not carry any Several examples of class I transposable elements that
accessory genes such as drug or heavy-metal resistance, encode enzymes for catabolic pathways involved in the
or catabolic markers. IS elements from both gram-neg- biodegradation of organic compounds are known.
ative and gram-positive bacteria have been described Transposition of class I elements is via the conservative
and their genetic properties have been extensively re- model, in which the transposing element translocates as
viewed (Syvanen 1988; Galas and Chandler 1989). These a physical entity from one site to another. These ele-
include the duplication of 2±20 bases at the target site of ments exhibit great diversity at the sequence level in both
insertion of the IS element (Syvanen1988). These direct their inverted repeats and their transposase genes.
repeats are believed to have arisen during the transpo- The chlorobenzoate catabolic genes on the plasmid
sition process when the transposase, encoded by the IS pBRC60 from Alcaligenes sp. strain BR60 have been
element itself and acting in cis, makes a staggered cut in reported to be localised on a composite transposon,
the DNA molecule, which is eventually ®lled-in by a designated Tn5271 (Nakatsu et al. 1991). Tn5271 is
DNA-repair system of the host cell (Grindley and Reed 17 kb in length and is ¯anked by two copies of IS1071,
1985). The length of the target duplication is charac- which are 3.2-kb direct repeats. Tn5271 resembles a class
teristic for each IS element (Craig 1996; Galas and I composite transposon but the ¯anking IS1071 element
Chandler 1989). IS elements are generally ¯anked by is a class II transposon (see below).
inverted repeats, which are believed to be the recognition The catabolic operon of Tn5271 carries cbaABC,
and binding site for the transposase during transposition which encodes a 3-chlorobenzoate 3,4-(4,5)-dioxygenase
(Gamas et al. 1987; Syvanen 1988). Class II groups the and the corresponding reductase and dehydrogenase in-
set of ancestrally related elements that are known as volved in the conversion of 3-chloro- and 3,4-dichloro-
the Tn3 family of transposons, while class III covers the benzoate to protocatechuate and chloroprotocatechuate
transposing bacteriophage Mu and related phages. respectively (Nakatsu and Wyndham 1993; Wyndham
There are, however, a number of transposable elements et al. 1994b; Nakatsu et al. 1997). Tn5271 was observed
that do not ®t into any of these classes. Nevertheless, re- to be able to transpose to the chromosomes of Coma-
gardless of their classi®cation, all transposable elements monas acidovorans (ATCC 15668) and Comamonas test-
consist of a unique DNA sequence ¯anked by short (8± osteroni (ATCC 11996), always as a portion of a larger
40 bp) inverted nucleotide sequence repeats. Almost all encompassing element, which suggests that Tn5271 may
the bacterial catabolic elements reported to date are be part of a larger transposon (Wyndham et al. 1994a).
classi®ed as either class I or class II transposons (Table 2). The ®rst two dioxygenase/dehydrogenase enzymes of
Transposition of catabolic transposons can either be the chlorobenzene degradation pathway of Pseudomonas
replicative or non-replicative depending on whether the sp. strain P51 are encoded on a plasmid-located trans-
mobile genetic element is duplicated or not during the poson Tn5280 (van der Meer et al. 1991b). The trans-
transposition process. In general, replicative transposi- poson comprises the 5.1-kb tcbAB catabolic gene cluster
tion proceeds through the formation of a cointegrate ¯anked by 1142-bp insertion sequences, IS1066 and
and involves the enzymatic activities of a transposase IS1067, which di€er at only 1 base pair within the 13-bp
that acts on the ends of the original transposon, and a inverted repeat on the pP51 plasmid. The 1068-bp open
resolvase that acts on the duplicated copies. Class-II- reading frame within the insertion elements could en-
type elements transpose in this way. On the other hand, code a putative transposase, with features similar to
non-replicative transposition requires only a transposase other IS-encoded transposases, such as a basic isoelectric
and allows the transposon to move as a physical entity point and a helix-turn-helix motif (Galas and Chandler
directly from a donor to a recipient site, as seen in the 1989). The end of IS1066 also contained an outwardly
transposition of Tn5 and Tn10. Other mobile elements, facing putative )35 promoter sequence, which is char-
such as IS1, use both non-replicative and replicative acteristic of other bacterial insertion elements. The in-
pathways (Biel and Berg 1984; Bender and Kleckner sertion elements IS1066 and IS1067 were found to be
1986; Berg and Howe 1989). homologous to a class of repetitive elements of Brady-
This review describes bacterial catabolic transposons rhizobium japonicum and distantly related to IS630 of
that encode enzymes involved in the catabolism of aro- Shigella sonnei (van der Meer et al. 1991b). When
4

Table 2 Catabolic transposons and transposon-like elements from bacteria

Transposon Size (kb) Catabolic marker (genes) IS Location Organism Reference

Class I transposons
Tn5271 17 Chlorobenzoate (cbaABC) IS1071 pBRC60 Alcaligenes sp. BR60 Nakatsu et al. 1991
Tn5280 8.5 Chlorobenzene (tcbAB) IS1066 pP51 Pseudomonas sp. P51 van der Meer et al. 1991b
IS1067
Tn5542 12 Benzene (bedDC1C2BA) IS1489 pHMT112 P. putida ML2 Fong et al. 1997
Chlorinated aliphatic acids (dehH2) IS1071 pUO1 Moraxellai sp. B Kawasaki et al. 1992
Nylon oligomers (nylABC) IS6100 pOAD2 Flavobacterium sp. K172 Kato et al. 1994
Transposons of unknown class and transposon-like elements
Tn4371 59 Chlorobiphenyl (bph/cbpABCD) ND Chromosome A. eutrophus A5 Springael et al. 1993
90 Biphenyl/salicylate (bph/sal) ND Chromosome P. putida KF715 Nishi et al. 1998
26 Aniline (tdnQTA1A2BR) IS1071 pTDN1 P. putida UCC22 Fukumori and Saint 1997
p-Toluenesulphonate/sulphobenzoate IS1071 pTSA C. testosteroni T2 Junker and Cook 1997
(tsaMBCDR/psbAC)
4-Carboxydiphenyl ether (pobAB) IS1071 pPOB P. pseudoalcaligenes POB310 Dehmel et al. 1995
Monobromoacetate (dhlB) IS1247 Chromosome X. autotrophicus GJ10 van der Ploeg et al. 1995
Class II transposons
Tn4651 56 Toluene (xyl) 46a pWW0 P. putida mt-2 Tsuda and Iino 1987
Tn4652 17 None Chromosome P. putida PaW85 Horak and Kivisaar 1998
a
Tn4653 70 Toluene (xyl) 38 pWW0 P. putida mt-2 Tsuda and Iino 1988
Tn4655 38 Naphthalene (nah) 38a NAH7 P. putida G7 Tsuda and Iino 1990
Tn4656 39 Toluene (xyl) 38a pWW53 P. putida MT53 Williams et al. 1992
a
Size of inverted repeats in bp
ND not determined
5

marked with a kanamycin-resistance gene and intro-


duced into Pseudomonas putida KT2442, Tn5280 was IS1071
found to be functional, transposing into the genome at
random and in a single copy. The IS1071 element deserves special mention. Although
The structure of a new catabolic transposon in a class-II-type insertion element it has been associated
P. putida ML2 carrying the ``upper'' pathway genes for with class-I transposons. Increasingly a number of
benzene degradation has been described (Fong et al. transposable catabolic gene clusters that are ¯anked by
1997). Two directly repeated sequences of the IS element IS1071 have been found. Involvement of IS1071 is
IS1489 are found to ¯ank the benzene dioxygenase and thought to be responsible for transferring degradative
dehydrogenase genes on the catabolic plasmid phenotypes into the environment (Fulthorpe and
pHMT112 in PpML2 (Tan and Mason 1990; Tan et al. Wyndham 1992; Fukumori and Saint 1997).
1993; Tan and Fong 1993; Fong et al. 1996) forming a IS1071, part of the chlorobenzoate catabolic trans-
composite transposon Tn5542. Only the left-hand IS is poson Tn5271, is 3201 bp long and contains inverted
bound by a pair of 40-bp imperfect, inverted repeats 110-bp repeat sequences. The outer 38 bp of these re-
while the right-hand copy lacks the right-hand inverted peats are related to the class II inverted repeats that
repeat. Both IS elements, termed variants 1 and 2 re- comprise the transposase recognition sequence. There is
spectively, contain an identical 1371-bp open reading an integration host factor (IHF) consensus binding se-
frame, tnpA, which was shown to encode a 53-kDa quence within the IR sequences. However, the 5-bp di-
polypeptide. The deduced amino acid sequence and pI rect repeats of DNA following transposition of all class
are typical of other transposases. In addition, seven dam II elements (Sherratt 1989) are missing in Tn5271
methylation and four possible chi sites, involved in (Nakatsu et al. 1997). Between the inverted repeats is a
regulating transposition and homologous recombination tnpA open reading frame. There are no tnpR gene or res
respectively, were identi®ed throughout the sequence of sites within IS1071. Intramolecular transposition/inver-
tnpA. An outwardly facing )10 and )35 promoter was sion derivatives of the right-hand IS1071 end of Tn5271
detected at the right-hand end of IS1489 v1 and overlaps have been detected in an actual groundwater bioreme-
with the bedD gene encoding cis-benzene dehydrodiol diation system and indicate the functionality of IS1071
dehydrogenase (Fong et al. 1996, 1997). Tn5542, how- (Wyndham et al. 1994b).
ever, was not shown to be functional. Besides ¯anking the cba genes in Alcaligenes sp. strain
The presence of a catabolic transposon is also im- BR60, isolated from the Niagara River watershed,
plicated in the nylon-oligomer-degrading Flavobacterium IS1071 has been found to ¯ank the dehH2 dehalogenase
sp. strain K172 (Kato et al. 1994). Three of the degra- gene of plasmid pU01 from Moraxella sp. strain B,
dative genes (nylABC) are borne on the 43.6-kb plasmid which has been isolated from industrial wastewater in
pOAD2 ¯anked by two types of repeated sequences. The Japan (Wyndham et al. 1994a; Kawasaki et al. 1992). In
nucleotide sequence of the type of repeated sequences addition, partial sequence information reveals a copy of
¯anking ny1A revealed a 765-bp open reading frame IS1071 upstream of the pobA gene that is involved in the
encoding a putative transposon and 14-bp IR that metabolism of carboxydiphenyl ethers in Pseudomonas
showed identity to IS6100, which is part of Tn610 in pseudoalcaligenes POB310 (Dehmel et al. 1995), while
Mycobacterium fortuitum encoding sulphonamide resis- two copies of the IS are present on plasmid pTSA car-
tance (Martin et al. 1990). rying the sulphobenzoate- and p-toluenesulphonate-
There are indications that the haloacid dehalogenase degradative genes in C. testosteroni strain T-2 (Nakatsu
genes may be located on a transposon. In Morexella sp. et al. 1997; Junker et al. 1996; Junker and Cook 1997).
strain B, the dehH2 gene encoding one of the two ha-
loacetate dehalogenases, which acts on monochloro-,
monobromo- and monoiodoacetate was found to be Class II transposons
sandwiched between two repeated sequences of 1.8 kb
on a 65-kb conjugative plasmid, pUO1 (Kawasaki et al. The class II or Tn3-like transposons are characterised by
1981). The repeat sequences are IS1071-like IS, encoding having short (fewer than 50 bp) terminal inverted repeat
the tnpA transposase, and are thought to play a part in (IR) sequences and the involvement of a transposase
the frequent spontaneous deletion of a 5.4-kb fragment (TnpA) and resolvase (TnpR) in a two-step transposi-
containing dehH2 from pUO1 (Kawasaki 1992; Wynd- tion process that leads to a 5-bp duplication of the target
ham et al. 1994a). sequence (Sherratt 1989; Grinsted et al. 1990). In the
P. putida strain PP3 contains the gene for dehaloge- ®rst step of transposition, TnpA recognises the IR and
nase (deh1) involved in the hydrolytic dehalogenation of catalyses the formation of a cointegrate of the donor and
2-chloro- and 2,3,-dichloroacetic, propionic and but- target DNA molecules connected by two directly re-
anoic acids on a mobile genetic element designated peated copies of the transposon. In the second step, the
DEH. Integration of the DEH element into a number of cointegrate between the two sites of resolution (res) is
plasmids and the chromosome of the host has been re- resolved in a site-speci®c manner by the resolvase.
ported (Beeching et al. 1983; Slater et al. 1979; Thomas The ®rst report of a transposable catabolic operon
et al. 1992a, b). was that of the xyl genes for toluene catabolism from
6

P. putida mt-2 (Jacoby et al. 1978; Nakazawa et al. 1978; Both Tn4651 and Tn4653 are responsible for the
Chakrabarty et al. 1978). The genetic organization and many reported transpositions of the xyl gene cluster into
regulation of the xyl operon is perhaps the most well- the chromosome and various resistance and catabolic
characterized one among aromatic catabolic pathways plasmids of bacteria reported (Sinclair et al. 1986; Sin-
in bacteria. The xyl genes are encoded on the pWW0 clair and Holloway 1991; Assinder and Williams 1990;
plasmid and located on a 56-kb transposon, Tn4651, Jahnke et al. 1993).
which is contained within another 70-kb transposon, Another toluene-catabolic transposon, Tn4656, has
Tn4653. Transposition of both toluene-catabolic trans- been described which contains the xyl gene cluster and a
posons has been shown to occur via cointegrate forma- res-tnpR-tnpA region (Tsuda 1996). This transposon is
tion and resolution, and both have properties present in P. putida strain MT53 (Assinder and Williams
characteristic of class II elements (Tsuda and Iino 1987; 1990; Williams et al. 1992) and is believed to have arisen
1988; Tsuda et al. 1989). Tn4651 carries a complete set through DNA site-speci®c recombination. It is likely
of transposition functions not interchangeable with that more such catabolic transposons will be uncovered
those of other class II transposons. These include the cis- in other bacteria capable of degrading toluene.
acting res region and two uncharacterised divergently Similar to the xyl operon of pWW0, the nah gene
transcribed trans-acting tnpS and tnpT gene products. cluster for naphthalene catabolism was found to be part
These are present on a 2.2-kb DNA fragment located of a 38-kb class II transposon, Tn4655, on the 83-kb
some 48 kb away from the tnpA gene. Located within plasmid NAH7 from P. putida PpG7 (Yen and Serdar
the 48-kb intervening region is a 39-kb fragment con- 1988; Tsuda and Iino 1990). Tn4655 contains the tnpR
taining the xyl operon ¯anked by two directly repeated gene and res region but is defective in the cointegration
1275-bp sequences, designated IS1246. Reciprocal re- step requiring a complementing transposase from other
combination between the IS elements is believed to be Tn1722-type transposons in order to transpose. The 38-
responsible for the high frequency of the 39-kb deletion bp IR of Tn4655 are almost identical to those of Tn4653
observed (Assinder and Williams 1990; Reddy et al. and Tn1722. The resolution function of Tn4655 is un-
1994). Tn4651 is considered a novel subgroup of class II ique as it cannot complement other class II transposons
transposons (Grinsted et al. 1990). The 46-bp IR of nor be replaced by their resolvases. The 1.8-kb tnpR
Tn4651 contain sequences associated with TnpA binding region of Tn4655 is three times longer than the typical
and nuclease activity (Ichikawa et al. 1990). class II resolvase gene sequence. Analysis and experi-
A 17-kb derivative of the 56-kb Tn4651, designated mental observation suggest the Tn4655 TnpR protein to
Tn4652, has been reported in the chromosome of P. putida be a site-speci®c integrase (Tsuda 1996) able to catalyse
PaW85 and shown to belong to the Tn3 family on the basis both integration and resolution reactions (Berg and
of its transposition properties (Tsuda and Iino 1987). Howe 1989; Abremski and Hoess 1992).
Genetic analysis has localised the tnpA gene to the right
arm of Tn4652 (Tsuda and Iino 1987). The transposase
was most homologous to Tn5041, a mercury-resistance Transposons of unknown class
transposon. Functional analysis showed the presence of and transposon-like elements
an IHF-binding site upstream of the tnpA promoter,
which is only active in P. putida. Expression of the trans- C. testosteroni T-2 degrades p-toluenesulphonate (TSA)
posase gene was found to be enhanced by IHF, which via p-sulphobenzoate (PSB) and protocatechuate. The
binds to the terminal sequences of Tn4652 just adjacent to TSA operon (tsaMBCD) with its regulatory gene (tsaR),
the inverted repeats (HoÄrak and Kivisaar 1998). the PSB genes (psbAC) and possibly the transport genes
Tn4653, found on pWW0 in P. putida mt-2, encom- for both substrates are carried on an 85-kb plasmid pTSA
passes Tn4651 and two other transposition genes, tnpR in strain T-2 (Junker and Cook 1997). In another strain
and tnpA (Tsuda and Iino 1988). The transposase of C. testosteroni PSB-4, which catabolises PBS, another
Tn4653 ensures the independent cointegration of the plasmid pPSB was shown to carry the psb genes. Both
two toluene transposons. It is interchangeable with those pTSA and pPSB contain two copies of IS1071. Conju-
of Tn1722-type transposons, encoding resistance to tet- gation experiments that yielded PSB+ transconjugants
racycline, but not the Tn4651 transposase. The IR of that carried two copies of IS1071 in the chromosome
Tn4653 are also closely related to the those of Tn1722- suggest the presence of a composite PSB transposon
type transposons. Furthermore, the resolvases of analogous to Tn5271 (Junker and Cook 1997).
Tn4653 and Tn1722 are identical in primary sequence Xanthobacter autotrophicus GJ10 was found to be
(Grinsted et al. 1990) with the tnpR gene from Tn4653 able to adapt to monobromoacetate by overexpressing a
being able to complement the tnpR mutation in Tn1722. haloacid dehalogenase encoded by the dhlB gene, similar
Resolution of the Tn4653-mediated cointegrate, how- to the enzyme encoded by deh2. This overexpression is
ever, requires the resolution system of Tn4651. This is due to the insertion of IS1247 34 bp upstream of dhlB.
due to the fact that the Tn4653 res region, located up- IS1247 was found to encode a putative 464-amino-acid
stream of tnpR, is defective, lacking a 30-bp sequence transposase and to have terminal IR of 17 bp and 16 bp
that contains the essential crossover site for resolution of with three mismatches (van der Ploeg et al. 1995). In-
the cointegrate (Tsuda 1996; Allmeier et al. 1992). sertion of the IS generated a 4 bp duplication of the
7

target site. One copy of IS1247 together with dhlB is able In P. putida TMB, which is involved in the catabolism
to transpose to another replicon in the pPJ20. This of methyl-substituted aromatic hydrocarbons, a region
phenomenon has been termed one-ended transposition of the chromosome was found to contain the tmb ope-
and occurs in Tn3, Tn21 and Tn1721, which lack one ron, which is functionally and genetically homolgous to
end of the inverted repeats (Arthur et al. 1984; Avila the xyl genes of pWW0. Located within the tmb operon
et al. 1984; Motsch and Schmitt 1984). is a DNA fragment that has homology to IS1001 (Fa-
The aromatic-amine-catabolising plasmid pTDN1 varo et al. 1996).
was discovered in a derivative of P. putida mt-2 fol- There are other examples of catabolic genes associ-
lowing adaptation to growth on aromatic amines. The ating with mobile elements but many of these genes have
catabolic genes are contained within a 26-kb region not been observed to transpose as discrete elements. The
bounded by 1.8-kb direct-repeat sequences which are (2,4,5-trichlorophenoxy)acetic-acid-catabolic genes of
readily deleted when the host cells are cultured on non- Burkholderia cepacia AC1100 are ¯anked by the 1477-bp
selective carbon sources (Saint et al. 1990; McClure and IS931 (Tomasek et al. 1989; Haugland et al. 1990),
Venables 1987). Six of the aniline-degradative genes which is self-transposable.
(tdnQTA1A2BR) are located downstream of a copy of
IS1071, which di€ers in a single base from that involved
in the transposition of the chlorobenzoate genes (Nak-
atsu et al. 1991). The partial sequence of the other direct- Non-aromatic catabolic transposons
repeat sequence on pTDN1 is reported to be identical to
IS1071 (Fukumori and Saint 1997). In addition to the aromatic catabolic transposons,
Alcaligenes eutrophus A5 can degrade chlorobiphenyl there are other catabolic transposable elements that
and biphenyl. It contains a 75-kb plasmid that carries carry genes encoding the metabolism of sugars and
4-chlorobenzoate-catabolic genes (cbp). Upon repeated citric acid. The lactose operon in Yersinia enterocolitica
subculturing, A. eutrophus A5 lost the ability to min- is carried on a 16.6-kb transposon, Tn951, encoded on
eralize 4-chlorobenzoate to CO2 as a result of a loss of the plasmid pGC1 (Cornelis et al. 1976). The lacIZY
dehalogenase activity, and consequently had a 51-kb genes of Tn951 are homologous to the E. coli K12
plasmid pSS50 that did not contain any chlorobiphenyl- lactose operon (Cornelis et al. 1978) as well as to those
or chlorobenzoate-catabolic genes. However, in conju- of 11 other plasmids of di€erent origins, although none
gation experiments, pSS50 was found to acquire a of these was found to contain the entire Tn951
59-kb transposon Tn4371 from the A. eutrophus A5 (Cornelis 1981). Tn951 is ¯anked by two perfect 41-bp
chromosomal DNA. The transposon carries the genes IR containing within them the 38-bp IR of Tn3. Tn951
involved in the degradation of biphenyl and 4-chlor- also contains a single copy of the insertion sequence,
obiphenyl to benzoate and 4-chlorobenzoate (bphA- IS1, upstream of lacI. This IS has been shown to
BCD/cbpABCD) respectively, and is able to transpose generate deletions and inversions within the transposon
to pSS50 and other plasmids. Tn4371 awaits further resulting in Lac) phenotypes (Cornelis and Saedler
characterization. 1980). Internal to the transposon and downstream of
The biphenyl- and salicylate-catabolic pathways of P. lacY is another transposon, Tn2501, which is related to
putida KF715 are encoded by bph and sal gene clusters the Tn21 family of class II transposable elements.
arranged 10 kb apart on the chromosome. These gene Tn2501 is ¯anked by 48-bp imperfect IR and does not
clusters were found to be highly prone to deletion when carry any transposon marker. The right-hand IR of
KF715 was grown in rich medium and could be trans- Tn2501 is found to lie next to the end of the defective
ferred by conjugation to P. putida AC30 at a frequency tnpA gene of Tn951 (Michels and Cornelis 1984).
of 10)6/cell. The integrated 90-kb DNA fragment con- Tn951 does not transpose on its own. However, its
taining bph and sal genes in AC30 could undergo further transposition can be complemented by the tnpA gene of
transposition and deletion and is believed to be located other class II or Tn3-type transposons such as Tn801
on a large catabolic transposon (Nishi et al. 1997, 1998). or Tn3, but not Tn501, Tn1721 and c-d, suggesting
The presence of IS and IS-like elements on catabolic that the complementation is a speci®c process (Cornelis
plasmids or in association with chromosomally encoded et al. 1981). Tn951 is able to transpose into multiple
catabolic genes suggests that these elements may be sites on the broad-host-range plasmid RP1 in Pseudo-
precursors to the development of catabolic transposons. monas aeruginosa as well as into other replicons
By recruiting genes from a variety of host bacteria, IS (Cornelis et al. 1978, 1979, 1981). As is the case for
elements play an important role in the evolution and Tn3, the transposition of Tn951, in the presence of
patchwork assembly of novel catabolic genes and ope- Tn801 or Tn3, is a thermosensitive process occurring
rons. readily at 30 °C, but not at 37 °C (Cornelis 1980).
IS1412, a 1656-bp IS element, contains terminally Transposition also results in a 5-bp duplication of the
partially matched 17-bp and 18-bp inverted repeats and target site. However, unlike Tn3, Tn951 has not been
a putative 457-amino-acid transposase. IS1412 has been shown to contain any tnpR gene (Cornelis et al. 1981),
cloned from plasmid DNA in the carbofuran-degrading but is thought to rely on the resolution system of
Sphingomonas sp. CF06 (Feng et al. 1997a, b). Tn2501 (Michels and Cornelis 1984).
8

The genes for the catabolism of sucrose (sacA, en- sulted in the appearance of a bacterial population able
coding sucrose-6-phosphate hydrolase) and the produc- to degrade 3-chlorobenzoate (Pertsova et al. 1984). The
tion of the peptide antibiotic nisin (nisA) are located on conjugative transfer of the catabolic transposon Tn5271
conjugative transposons, Tn5276 and Tn5301, in Lac- allows the utilization of 3-chlorobenzoate in a freshwa-
tococcus lactis (Dodd et al. 1990; Rauch and de Vos ter microcosm, and its constituent IS1071 plays a sig-
1992). The better-characterised Tn5276 is a 70-kb con- ni®cant role in the adaptation to 4-chloroaniline
jugative transposon in the chromosome of L. lactis. It (Fulthorpe and Wyndham 1992).
contains (A + T)-rich termini ¯anked by a direct Soil having indigenous biphenyl degraders but not
hexanucleotide repeat without any clear IR. The inte- chlorobenzoate degraders and inoculated with P. aeru-
gration of Tn5276 into the chromosome of a plasmid- ginosa JB2, a chlorobenzoate degrader, was found to
free donor strain displays orientation and site speci®city produce two recombinant strains, P. aeruginosa JB2-3
(Rauch and de Vos 1992). and Pseudomonas sp. JB2-M, which could metabolise
In Escherichia coli the plasmid-borne genes encoding the polychlorinated biphenyls of Aroclor 1242. Results
citrate metabolism are carried on a 7.4-kb transposon, from repetitive extragenic palindromic polymerase chain
Tn3411 (Ishiguro et al. 1982). Tn3411, a class I com- reaction analysis suggest that the recombinants arose
posite element, contains two directly repeated copies of from the transfer of bph genes from indigenous biphenyl
an insertion sequence, IS3411, and generates a 3-bp re- degraders to the inoculant JB2, probably via a trans-
peat upon integration. IS3411 is 1309 bp long, has 27-bp poson (Focht et al. 1996). The recombinant strains iso-
imperfect IR and a putative 240-amino-acid transposase lated were, however, unstable. This instability presents
(Ishiguro and Sato 1988). Intramolecular recombination an interesting paradox, for while it may be dicult to
between the two copies of IS3411 occurs frequently in a maintain stable inoculants for bioremediation applica-
recA strain resulting in Cit) deletion mutants (Ishiguro tion, this phenomenon may be advantageous in mini-
and Sato 1984). Tn3411 has been shown to transpose to mizing the perceived risks of recombinant persistance in
k bb phage and then from the Cit+-transducing k bb the environment (Focht et al. 1996).
phage to pBR3222 in recA-de®cient strains (Ishiguro Plasmid-encoded pathways, such as those for
et al. 1982). Other plasmids specifying citrate utilization chlorocatechol degradation (e.g. on pAC27, pJP4, pP51),
have also been reported (Ishiguro et al. 1981), but none are expected to evolve more rapidly than chromosomal
of these could transpose its citrate-utilization genes genes (Eberhard 1990). It is believed that the ancient
(Ishiguro et al. 1982). chlorocatechol-degradation operon, which evolved long
ago to give rise to the clc, tfd and tcb variants, probably
existed originally for the catabolism of naturally occur-
Ecological signi®cance ring halogenated compounds such as chlorosubstituted
and bromosubstituted aromatics (SchloÈmann 1994). Al-
The nature of waste being generated has changed dras- though gene transfers may occur within a very limited
tically over the years. In recent decades a large number taxonomic range of hosts (Williams and Sayers 1994), the
of xenobiotics have been released into the environment. genetic characteristics of the bedDC1C2BA operon in
While many of these chemicals are rapidly degraded by PpML2 carried on Tn5542 (Tan et al. 1993; Fong et al.
microorganisms in the environment, some resist attack 1996) point to a juxtaposition of catabolic genes on the
and remain recalcitrant. Given time, however, most catabolic plasmid pHMT112 from hosts of di€erent ge-
microorganisms, in particular bacteria, are able to adapt nera. This would also have been true of other catabolic
to using these compounds as energy and carbon sources. operons (Shanley et al. 1994; van der Meer et al. 1991b;
This biochemical versatility is largely due to the plas- Werlen et al. 1996). Furthermore, horizontal transfer of
ticity of the microbial genomes. By modifying the ex- genetic material encoding toluene or biphenyl-degrada-
isting genes, a novel metabolic capacity can be tion enzymes from gram-negative species (most probably
developed that allows xenobiotics to be metabolised. Pseudomonas) to gram-positive Rhodococcus globerulus
This requires the alteration and exchange of genetic in- P6 is thought to be an example of how microorganisms
formation, and recombination processes such as gene gain novel catabolic activities for xenobiotics (Asturias
conversion, duplication and transposition play crucial et al. 1995). The co-location of these di€erent catabolic
roles in the reassortment of discrete genetic modules and gene clusters on a single plasmid or in a single organism
their expression (van der Meer et al. 1992). ensures the proper regulation of the pathways involved
Catabolic plasmids and transposons allow the hori- (van der Meer et al. 1991b). Indeed, the transposition of
zontal spreading of degradative genes among microbial mobile genetic elements can e€ect the expression, by
communities and this is important for genetic ¯exibility activation, of adjacent catabolic genes (Kivisaar et al.
and adaptation. It has been reported that greater num- 1990; Haugland et al. 1990; van der Meer et al. 1992; van
bers of plasmid-containing bacteria are often isolated der Ploeg et al. 1995; Fong et al. 1996).
from polluted areas than are found in unpolluted areas The combination of transposons with plasmids plays
(Hardman et al. 1986; Focht et al. 1996). Genetic ex- a major role in the rapid dissemination and evolution of
change between strains of P. aeruginosa and P. putida antibiotic and heavy-metal-resistance markers in the
containing plasmids and indigenous microbiota has re- chemical environment following widespread use of an-
9

tibiotics (Amabile-Cuevas and Chicuvel 1992). The same bacteria would be employed in the natural environment
is true for the ability of microbial communities to adapt on a large scale. On the other hand, an increasing
to arthropogenic xenobiotic compounds. This genetic number of natural isolates have been reported that
adaptability is aided by an accumulation of single-site possess unique metabolic genes and new catabolic
mutations followed by gene conversion or slipped-strand plasmids and transposons. Some of these, selected under
mispairing and by gene duplication followed by muta- speci®c substrates during enrichment, have been shown
tions in one of the gene copies as well as DNA rear- to possess a combination of catabolic genes encoding
rangements and transposition (van der Meer et al. 1992; metabolic functions as versatile if not more so, than
Williams and Sayers 1994). These mechanisms greatly those designed using molecular techniques in the labo-
expand the substrate range of the microorganisms and ratory (Mars et al. 1997; Pettigrew et al. 1991; Eaton
contribute to the evolution of catabolic pathways in 1996; Williams and Sayers 1994). These microorganisms
general. Currently there is a paucity of ®eld data for the that have evolved naturally tend to be more competitive
rate of evolution in the natural environment (Liu and (van der Meer et al. 1992). The extensive bioremediation
Su¯ita 1993; van der Meer et al. 1992; Williams and of the Alaskan oil spill represents the most successful use
Sayers 1994). Nevertheless, the involvement of catabolic of indigenous microorganisms to date (Pritchard and
transposons in such genetic processes would inevitably Costa 1991) although numerous other successful appli-
enhance evolutionary changes in metabolic pathways, cations for the remediation of sites polluted by chemical
rendering microorganisms the ability to evolve novel spills, leaking underground storage tanks and industrial-
and productive pathways. process wastes also exist (Caplan 1993). Clearly, optimal
physical and chemical conditions, superimposed on the
genetic exchange among the microbial community, me-
Biotechnological implications diated by catabolic transposons, among other mecha-
nisms, will allow the eventual evolution of the most
Bioremediation, which exploits the catabolic versatility adapted microbial population, the growth and activity
of microorganisms to accelerate the degradation of en- of which ensures the recycling of aromatic compounds in
vironmental pollutants, is an important industry in the biosphere and a balanced global ecosystem.
mitigating environmental contamination. Microorgan-
isms that are able to utilize the polluting chemicals as Acknowledgements The author appreciates the helpful comments
potential energy sources survive in the hostile environ- and suggestions of M. Tsuda. Some of the results presented here
come from the contributions of K. Fong, C. Goh and other
ment. In a sense, bioremediation of pollutants is an ex- members of the laboratory. The support of the National University
tension of the metabolism that occurs within the of Singapore is gratefully acknowledged.
microorganism and its e€ectiveness depends largely on
the extent to which the desired microbial community can
be selected and maintained in the contaminated envi- References
ronment. In a way, catabolic transposons serve as units
of genetic ¯uidity in the environment. Although the rates Abremski KE, Hoess RH (1992) Evidence for a second conserved
of genetic rearrangement may not be high under normal arginine residue in the integrase family of recombination pro-
conditions, it can be envisaged that, upon the intro- teins. Protein Eng 5: 87±91
duction of xenobiotic substrates into the environment, Allmeier H, Cresnar B, Greck M, Schmitt R (1992) Complete nu-
cleotide sequence of Tn1721: gene organization and a novel
selective pressure would increase to the point where high gene product with features of a chemotaxis protein. Gene 111:
growth rates, resulting in suciently high microbial 11±20
populations that favour such genetic interactions, can be Amabile-Cuevas CF, Chicurel ME (1992) Bacterial plasmids and
established. The location of catabolic genes on trans- gene ¯ux. Cell 70: 189±199
Arthur A, Nimmo E, Hettle S, Sherratt D (1984) Transposition and
posons and plasmids increases the possibilities of re- transposition immunity of transposon Tn3 derivatives having
combination events and accelerates the metabolic di€erent ends. EMBO J 3: 1723±1729
potential of the microbial population best adapted to Assinder SJ, Williams PA (1990) The TOL plasmids: determinants
biodegrading the xenobiotic compound. of the catabolism of toluene and the xylenes. Adv Microb
Many bacterial strains have been engineered to pos- Physiol 31: 1±69
Asturias JA, Diaz E, Timmis KN (1995) The evolutionary rela-
sess additional catabolic genes for added metabolic tionship of biphenyl dioxygenase from gram-positive Rho-
prowess (Rojo et al. 1987; Lehrbach et al. 1984; Ramos dococcus globerulus P6 to multicomponent dioxygenases from
et al. 1987; reviewed by Timmis et al. 1994). While such gram-negative bacteria. Gene 156: 11±18
engineered strains may perform under laboratory con- Avila P, Cruz F de la, Ward E, Grinsted J (1984) Plasmids con-
taining one inverted repeat of Tn21 can fuse with other plas-
ditions and be considered for application in bioremedi- mids in the presence of Tn21 transposase. Mol Gen Genet 195:
ation, these bacteria in general are too fastidious to 288±293
survive in a complex multisubstrate, multiorganism-in- Beeching JR, Weightman AJ, Slater JH (1983) The formation of an
teracting environment without speci®c selection pres- R prime carrying the fraction I dehalogenase from Pseudo-
monas putida PP3 using the IncP plasmid R68.44. J Gen Mi-
sure. Coupled with the need for physical containment, crobiol 129: 2071±2078
genetic tracking and attendant biosafety issues and Bender J, Kleckner N (1986) Genetic evidence that Tn10 transposes
public pressure, it is debatable whether such engineered by a nonreplicative mechanism. Cell 45: 801±815
10

Berg DE, Howe MM (eds) (1989) Mobile DNA. American Society chlorobenzoate in Alcaligenes eutrophus JMP134 (pJP4).
for Microbiology, Washington DC J Bacteriol 161: 85±90
Besteti G, Galli E (1987) Characterization of a novel TOL-like Duggleby CJ, Bayley SA, Worsey MJ, Williams PA, Broda P
plasmid from Pseudomonas putida involved in 1,2,4-trimethyl- (1977) Molecular sizes and relationships of TOL plasmids in
benzene degradation. J Bacteriol 169: 1780±1783 Pseudomonas. J Bacteriol 130: 1274±1284
Biel SW, Berg DE (1984) Mechanism of IS1 transposition in E. coli: Dunn NW, Dunn HM, Austen RA (1980) Evidence for the exis-
choice between simple insertion and cointegration. Genetics tence of two catabolic plasmids coding for the degradation of
108: 319±330 naphthalene. J Gen Microbiol 117: 529±533
Boronin AM, Kochetkov VV, Skryabin GK (1980) Incompatibility Dunn NW, Gunsalus IC (1973) Transmissible plasmid coding early
groups of naphthalene degradative plasmids in Pseudomonas. enzymes of naphthalene oxidation in Pseudomonas putida.
FEMS Microbiol Lett 7: 249±252 J Bacteriol 114: 974±979
Caplan JA (1993) The worldwide bioremediation industry: pros- Eaton RW (1996) p-Cumate catabolic pathway in Pseudomonas
pects for pro®t. Trends Biotechnol 11: 320±323 putida F1: cloning and characterization of DNA carrying the
Chakrabarty AM (1972) Genetic basis of the biodegradation of cmt operon. J Bacteriol 178: 1351±1362
salicylate in Pseudomonas. J Bacteriol 112: 815±823 Eaton RW, Timmis KN (1986) Characterization of a plasmid-
Chakrabarty AM, Friello DA, Bopp LH (1978) Transposition of speci®ed pathway for catabolism of isopropylbenzene in Pseu-
plasmid DNA segments specifying hydrocarbon degradation domonas putida RE204. J Bacteriol 168: 123±131
and their expression in various microorganisms. Proc Natl Eberhard WG (1990) Evolution in bacterial plasmids and levels of
Acad Sci USA 75: 3109±3112 selection. Q Rev Biol 65: 3±22
Chatterjee DK, Kellogg ST, Hamada S, Chakrabarty AM (1981) Favaro R, Bernasconi C, Passini N, Bertoni G, Bestetti G, Galli E,
Plasmid specifying total degradation of 3-chlorobenzoate by a Deho G (1996) Organisation of the tmb catabolic operons of
modi®ed ortho pathway. J Bacteriol 146: 639±646 Pseudomonas putida TMB and evolutionary relationship with
Chatterjee DK, Chakrabarty AM (1983) Genetic homology be- the xyl operons of the TOL plasmid pWWO. Gene 182:
tween independently isolated chlorobenzene-degradative plas- 189±193
mids. J Bacteriol 153: 532±534 Feng X, Ou LT, Ogram A (1997a) Plasmid mediated mineraliza-
Cornelis G (1980) Transposition of Tn951 (Tnlac) and cointegrate tion of carbofuran by Sphingomonas sp. CF06. Appl Environ
formation are thermosensitive processes. J Gen Microbiol 117: Microbiol 63: 1332±1337
243±247 Feng X, Ou LT, Ogram A (1997b) Cloning and sequence analysis
Cornelis G (1981) Sequence relationships between plasmids carry- of a novel insertion element from plasmids harbored by the
ing genes for lactose utilization. J Gen Microbiol 124: 91±97 carbonfuran-degrading bacterium, Sphingomonas sp. CF06.
Cornelis G, Saedler H (1980) Deletions and an inversion induced Plasmid 37: 169±179
by a resident IS1 of the lactose transposon Tn951. Mol Gen Focht DD, Searles DB, Koh SC (1996) Genetic exchange in soil
Genet 178: 367±374 between introduced chlorobenzoate degraders and indigenous
Cornelis G, Bennett PM, Grinsted J (1976) Properties of pGL1, a biphenyl degraders. Appl Environ Microbiol 62: 3910±3913
lac plasmid originating in Yersinia enterocolitica 842. J Bacteriol Fong KPY, Goh BH, Tan HM (1996) Characterization and ex-
127: 1058±1062 pression of the plasmid-borne bedD gene from Pseudomonas
Cornelis G, Ghosal D, Saedler H (1978) Tn951: a new transposon putida ML2, which codes for a NAD+-dependent cis-benzene
carrying a lactose operon. Mol Gen Genet 160: 215±224 dihydrodiol dehydrogenase. J Bacteriol 178: 5592±5601
Cornelis G, Ghosal D, Saedler H (1979) Multiple integration sites Fong PY, Goh C, Tan G, Tan HM (1997) Identi®cation and ge-
for the lactose transposon Tn951 on plasmid RP1 and estab- netic analysis of Tn5542, a transposable element carrying the
lishment of a coordinate system for Tn951. Mol Gen Genet 168: bedD and bedC1C2BA genes in Pseudomonas putida ML2. In:
61±67 Abstracts of the Sixth International Congress on Pseudomonas:
Cornelis G, Sommer H, Saedler H (1981) Transposon Tn951 molecular biology and biotechnology 1997. Madrid, Spain, p 53
(Tnlac) is defective and related to Tn3. Mol Gen Genet 184: Frantz B, Chakrabarty AM (1986) Degradative plasmids in Pseu-
241±248 domonas. In: Sokatch JR (ed) The Bacteria, vol X. Academic
Craig NL (1996) Transposition. In: Neidhart FC (ed) Escherichia Press, London, pp 295±323
coli and Salmonella: cellular and molecular biology. American Fukumori F, Saint CP (1997) Nucleotide sequence and regulational
Society for Microbiology, Washington DC, pp 2239±2362 analysis of genes involved in conversion of aniline to catechol in
Dabrock B, Kesseler M, Averho€ B, Gottschalk RG (1994) Iden- Pseudomonas putida UCC22 (pTDN1). J Bacteriol 179: 399±408
ti®cation and characterization of a transmissible linear plasmid Fulthorpe RR, Wyndham RC (1991) Transfer and expression of
from Rhodococcus erythropolis BD2 that encodes isopropyl- the catabolic plasmid pBRC60 in wild bacterial recipients in a
benzene and trichoroethane catabolism. Appl Environ Micro- fresh water ecosystem. Appl Environ Microbiol 57: 1546±1553
biol 60: 853±860 Fulthorpe RR, Wyndham RC (1992) Involvement of a chloro-
Dahlberg C, Hermansson M (1995) Abundance of Tn3, Tn21, and benzoate-catabolic transposon, Tn5271, in a community ad-
Tn501 transposase (tnpA) sequences in bacterial community aptation to chlorobiphenyl, chloroaniline and 2,4-
DNA from marine environments. Appl Environ Microbiol 61: dichlorphenoxyacetic acid in a freshwater ecosystem. Appl
3051±3056 Environ Microbiol 58: 314±325
Dean HF, Cheevadhanarak S, Skurray Ra, Bayly RC (1989) Galas DJ, Chandler (1989) Bacterial insertion sequences. In: Berg
Characterization of a degradative plasmid in Pseudomonas DE, Howe MM (eds) Mobile DNA. American Society for
putida that controls the expression of 2,3-xylenol degradative Microbiology. Washington, DC, pp 109±62
genes. FEMS Microbiol Lett 61: 153±158 Gamas P, Chandler MG, Prentki P, Galas DJ (1987) Escherichia
Dehmel U, Engesser KH, Timmis KN, Dwyer DF (1995) Cloning, coli integration host factor binds speci®cally to the ends of the
nucleotide sequence, and expression of the gene encoding a insertion sequence IS1 and to its major insertion hot-spot in
novel dioxygenase involved in metabolism of carboxydiphenyl pBR322. J Mol Biol 195: 261±272
ethers in Pseudomonas pseudoalcaligenes POB310. Arch Mi- Grindley NDF, Reed PR (1985) Transpositional recombination in
crobiol 163: 35±41 prokaryotes. Annu Rev Biochem 54: 863±896
Dodd HM, Horn N, Gasson MJ (1990) Analysis of the genetic Grinsted J, De La Cruz F, Schmitt R (1990) The Tn21 subgroup of
determinant for production of the peptide antibiotic nisin. J Gen bacterial transposable elements. Plasmid 24: 163±189
Microbiol 136: 555±566 Gunsalus IC, Grund A, Yen KM (1982) Aromatic oxygenation,
Don RH, Weightman AJ, Knackmuss HJ, Timmis KN (1985) genetics and regulation. In: Nozaki M, Yamamoto S, Ishimura
Transposon mutagenesis and cloning analysis of the pathways Y, Coon MJ, Ernster L, Eastbrook RW (eds) Oxygenases and
for degradation of 2,3-dichlorophenoxy acetic acid and 3- oxygen metabolism. Academic Press, London, pp 79±91
11

Hallet B, Sherratt DJ (1997) Transposition and site-speci®c re- Kivisaar M, Horak R, Kasak L, Heinaru A, Habicht J (1990) Se-
combination: adapting DNA cut-and-paste mechanisms to a lection of independent plasmids determining phenol degrada-
variety of genetic rearrangements. FEMS Microbiol Rev 21: tion in Pseudomonas putida and expression of genes encoding
157±178 phenol monoxygenase and catechol-1,2-dioxygenase. Plasmid
Hardman D, Gowland PC, Slater JH (1986) Large plasmids from 24: 25±36
soil bacteria enriched on halogenated alkanoic acids. Appl Kunz DA, Chapman PJ (1981) Isolation and characterization of
Environ Microbiol 51: 41±51 spontaneously occurring TOL plasmid mutants of Pseudomonas
Haugland RA, Sangodkar UMX, Chakrabarty AM (1990) Re- putida HS1. J Bacteriol 146: 952±964
peated sequences including RS1100 from Pseudomonas cepacia Lehrbach PR, Zeyer J, Reineke W, Knackmuss HJ, Timmis KN
AC1100 function as IS elements. Mol Gen Genet 220: (1984) Enzyme recruitment in vitro: use of cloned genes to ex-
222±228 tend the range of haloaromatics degraded by Pseudomonas sp.
Hermann H, Janke D, Krejsa S, Kunze I (1987) Involvement of the strain B13. J Bacteriol 158: 1025±1032
plasmid pPGH1 in the phenol degradation of Pseudomonas Liu S, Su¯ita JM (1993) Ecology and evolution of microbial pop-
putida strain H. FEMS Microbiol Lett 43: 133±137 ulations for bioremediation. Trends Biotechnol 11: 344±352
Hooper DJ, Kemp PD (1980) Regulation of enzymes of the 3,5- Mars AE, Kasberg T, Kaschabek SR, Agtenen MH van, Janssen
xylenol degradative pathway in Pseudomonas putida: evidence DB, Reineke W (1997) Microbial degradation of chloroaro-
for a plasmid. J Bacteriol 142: 21±26 matics: Use of the meta-cleavage pathway for mineralization of
HoÄrak R, Kivisaar M (1998) Expression of the transposase gene chlorobenzene. J Bacteriol 179: 4530±4537
tnpA of Tn4652 is positively a€ected by integration host factor. Martin C, Timm J, Rauzier J, Gomez-Lus R, Davies J, Gicquel B
J Bacteriol 180: 2822±2829 (1990) Transposition of an antibiotic resistance element in
Ichikawa H, Ikeda K, Amemura J, Ohtsubo E (1990) Two domains mycobacteria. Nature 345: 739±743
in the terminal inverted-repeat sequence of transposon Tn3. Meer JR van der, Zehnder AJB, Vos WM de (1991b) Identi®cation
Gene 86: 11±17 of a novel composite transposable element, Tn5280, carrying
Ishiguro N, Sato G (1984) Spontaneous deletion of citrate-utilizing chlorobenzene dioxygenase genes of Pseudomonas sp. strain
ability promoted by insertion sequences. J Bacteriol 160: P51. J Bacteriol 173: 7077±7083
642±652 Meer JR van der, Vos WM de, Harayama S, Zehnder AJB (1992)
Ishiguro N, Sato G (1988) Nucleotide sequence of IS3411, which Molecular mechanisms of genetic adaptation to xenobiotic
¯anks the citrate utilization determinant of transposon Tn3411. compounds. Microbiol Rev 56: 677±694
J Bacteriol 170: 1902±1906 Meer JR van der, Neerven ARW van, Vries EJ de, Vos WM de,
Ishiguro N, Hirose K, Asagi M, Sato G (1981) Incompatibility of Zehnder AJB (1991a) Cloning and characterization of plasmid-
citrate utilization plasmids isolated from Escherichia coli. J Gen encoded genes for the degradation of 1,2-dichloro-,1,4-dichlo-
Microbiol 123: 193±196 ro-, and 1,2,4-trichlorobenzene of Pseudomonas sp. strain P51.
Ishiguro N, Sato G, Sasakawa C, Danbara H, Yoshikawa M J Bacteriol 173: 6±15
(1982) Identi®cation of citrate utilization transposon Tn3411 McClure NC, Venables WA (1987) pTDN1, a catabolic plasmid
from a naturally occurring citrate utilization plasmid. J Bac- involved in aromatic amine catabolism in Pseudomonas putida
teriol 149: 961±968 mt-2. J Gen Microbiol 133: 2073±2077
Jacoby GA, Rogers JE, Jacob AE, Hedges RW (1978) Transposi- Michels T, Cornelis G (1984) Detection and characterization of
tion of Pseudomonas toluene-degrading genes and expression in Tn2501, a transposon included within the lactose transposon
Escherichia coli. Nature 274: 179±180 Tn951. J Bacteriol 158: 866±871
Jahnke M, Lehmann F, Schoebel A, Auling G (1993) Transposi- Mizuuchi K (1992) Transpositional recombination: mechanistic
tion of the TOL catabolic genes (Tn4651) into the degradative insights from studies of Mu and other elements. Annu Rev
plasmid pSAH of Alcaligenes sp. 0-1 ensures simultaneous Biochem 61: 1011±1051
mineralisation of sulpho- and methyl-substituted aromatics. Motsch S, Schmitt R (1984) Replicon fusion mediated by a single-
J Gen Microbiol 139: 1959±1966 ended derivative of transposon Tn1721. Mol Gen Genet 195:
Jain RK, Bayly RC, Skurray Ra (1984) Characterization and 281±287
physical analysis of a 3,5-xylenol degradative plasmid in Pseu- Nakatsu CH, Wyndham RC (1993) Cloning and expression of the
domonas putida. J Gen Microbiol 130: 3019±3028 transposable chlorobenzoate-3,4-dioxygenase genes of Alcali-
Junker F, Cook AM (1997) Conjugative plasmids and the degra- genes sp. strain BR60. Appl Environ Microbiol 59: 3625±3633
dation of arylsulfonates in Comamonas testosteroni. Appl En- Nakatsu C, Ng J, Singh R, Straus N, Wyndham C (1991)
viron Microbiol 63: 2403±2410 Chlorobenzoate catabolic transposon Tn5271 is a composite
Junker F, Saller E, Schla® Oppenberg HR, Kroneck PHM, Lei- class I element with ¯anking class II insertion sequences. Proc
singer T, Cook AM (1996) Degradative pathways for p-toluate Natl Acad Sci USA 88: 8312±8316
and p-toluenesulfonate and their multicomponent oxygenases in Nakatsu CH, Providenti M, Wyndham RC (1997) The cis-diol de-
Comamonas testosteroni strains PSB-4 and T-2. Microbiology hydrogenase cbaC gene of Tn5271 is required for growth on 3-
142: 2419±2427 chlorobenzoate but not 3,4-dichlorobenzoate. Gene 196: 209±218
Kato K, Ohtsuki K, Mitsuda H, Yomo T, Negoro S, Urabe I Nakazawa T, Hayashi E, Yokota T, Ebina Y, Nakazawa A (1978)
(1994) Insertion sequence IS6100 on plasmid pOAD2, which Isolation of TOL and RP4 recombinants by integrative sup-
degrades nylon oligomers. J Bacteriol 176: 1197±1200 pression. J Bacteriol 134: 270±277
Kawasaki H, Yahara N, Tonomura K (1981) Isolation and char- Nishi A, Tominaga K, Furukawa K (1997) Deletion and self-mo-
acterization of plasmid pU01 mediating dehalogenation of ha- bilization of the chromosomal gene clusters coding for biphenyl
loacetate and mercury resistance in Moraxella sp. B. Agric Biol and salicylate metabolism in Pseudomonas putida KF715. In:
Chem 45: 1477±1481 Abstracts of Sixth International Congress on Pseudomonas:
Kawasaki H, Tsuda K, Matsushita I, Tunomura K (1992) Lack of molecular biology and biotechnology 1997. Madrid, Spain,
homology between two haloacetate dehalogenase genes encod- p 125
ed on a plasmid from Moraxella sp. strain B. J Gen Microbiol Nishi A, Tominaga K, Furukawa K (1998) Horizontal transfer of
138: 1317±1323 the chromosomal gene clusters coding for biphenyl and salicy-
Keshavarz T, Lilly MD, Clarke PH (1985) Stability of a catabolic late metabolism in Pseudomonas putida KF715. In: Abstracts of
plasmid in continuous culture. J Gen Microbiol 131: 1193±1203 First International Conference of the Federation of Asia-Paci®c
Kesseler M, Dabbs ER, Averho€ B, Gottschalk G (1996) Studies Microbiology Societies 1998. Singapore, p 92
on the isopropylbenzene 2,3-dixoygenase and 3-isopropyl cat- Pertsova RN, Kunc F, Golovleva LA (1984) Degradation of 3-
echol 2,3-dioxygenase genes encoded by the linear plasmid of chlorobenzoate in soil by pseudomonads carrying biodegrada-
Rhodococcus erythropolis BD2. Microbiology 142: 3241±3251 tive plasmids. Folia Microbiol 29: 242±247
12

Pettigrew CA, Haigler BE, Spain JC (1991) Simultaneous biodeg- cloning of the cis-benzene dihydrodiol dehydrogenase gene.
radation of chlorobenzene and toluene by a Pseudomonas Can J Microbiol 39: 357±362
strain. Appl Environ Microbiol 57: 157±162 Tan HM, Tang HY, Joannou CL, Abdel Wahab NH, Mason JR
Ploeg J van der, Willemsen M, van Hall G, Janssen DB (1995) (1993) The Pseudomonas putida ML2 plasmid-encoded genes
Adaptation of Xanthobacter autotrophicus GJ10 to bromoace- for benzene dioxygenase are unusual in codon usage and low in
tate due to activation and mobilization of the haloacetate de- G + C content. Gene 130: 32±39
halogenase gene by insertion element IS1247. J Bacteriol 177: Thomas AW, Slater JH, Weightman AJ (1992a) The dehalogenase
1348±1356 gene dehI from Pseudomonas putida PP3 is carried on an un-
Pritchard PH, Costa CF (1991) EPA's Alaska oil spill bioremedi- usual mobile genetic element designated DEH. J Bacteriol 174:
ation project. Environ Sci Technol 25: 372±379 1932±1940
Ramos JL, Wasserfallen A, Rose K, Timmis KN (1987) Rede- Thomas AW, Topping AW, Slater JH, Weightman AJ (1992b)
signing metabolic routes: manipulation of TOL plasmid path- Localization and functional analysis of structural and regula-
way for catabolism of alkylbenzoates. Science 235: 593±596 tory dehalogenase genes carried on DEH from Pseudomonas
Rauch PJG, Beerthuyzen MM, Vos VM de (1990) Nucleotide se- putida PP3. J Bacteriol 174: 1941±1947
quence of IS904 from Lactococcus lactis subsp. lactis strain Timmis KN, Ste€an RJ, Unterman R (1994) Designing microor-
NIZO R5. Nucleic Acids Res 18: 4253±4254 ganisms for the treatment of toxic wastes. Annu Rev Microbiol
Rauch PJG, Vos VM de (1992) Characterization of the novel nisin- 48: 525±557
sucrose conjugative transposon Tn5276 and its insertion in Tomasek PH, Frantz B, Sangodkar UMX, Haugland RA,
Lactococcus lactis. J Bacteriol 174: 1280±1287 Chakrabarty AM (1989) Characterization and nucelotide se-
Reddy BR, Shaw LE, Sayers JR, Williams PA (1994) Two identical quence determination of a repeat element isolated from a 2,4,5-
copies of IS1246, a 1275 base pair sequence related to other T degrading strain of Pseudomonas cepacia. Gene 76: 227±238
bacterial insertion sequences, enclose the xyl genes on TOL Tsuda M (1996) Catabolic transposons in pseudomonads. In:
plasmid pWWO. Microbiology 140: 2305±2307 Nakazawa T, Furukawa K, Haas D, Silver S (eds) Molecular
Rheinwald JG, Chakarabarty AM, Gunsalus IC (1973) A trans- biology of pseudomonads. American Society for Microbiology,
missable plasmid controlling camphor oxidation in Pseudo- Washington DC, pp 219±228
monas putida. Proc Natl Acad Sci USA 70: 885±889 Tsuda M, Iino T (1987) Genetic analysis of a transposon carrying
Rojo F, Pieper DH, Engesser KH, Knackmuss HJ, Timmis KN toluene degrading genes on a TOL plasmid pWWO. Mol Gen
(1987) Assemblage of ortho cleavage route for simultaneous Genet 210: 270±276
degradation of chloro- and methylaromatics. Science 238: Tsuda M, Iino T (1988) Identi®cation and characterization of
1395±1398 Tn4653, a transposon convering the toluene transposon Tn4651
Saint CP, McClure NC, Venables WA (1990) Physical map of the on TOL plasmid pWWO. Mol Gen Genet 213: 72±77
aromatic amine and m-toluate catabolic plasmid pTDN1 in Tsuda M, Iino T (1990) Naphthalene degrading genes on plasmid
Pseudomonas putida: location of a unique meta-cleavage path- NAH7 are on a defective transposon. Mol Gen Genet 223:
way. J Gen Microbiol 136: 615±625 33±39
SchloÈmann M (1994) Evolution of chlorocatechol catabolic path- Tsuda M, Minegishi KI, Iino T (1989) Toluene transposons
ways. Biodegradation 5: 301±321 Tn4651 and Tn4653 are class II transposons. J Bacteriol 171:
Shanley MS, Harrison A, Parales RE, Kowalchuk G, Mitchell DJ, 1386±1393
Ornston LN (1994) Unusual G + C content and codon usage Werlen C, Kohler HPE, Meer JR van der (1996) The broad sub-
in catIJF, a segment of the ben-cat supra-operonic cluster in the strate chlorobenzene dioxygenase and cis-chlorobenzene dihy-
Acinetobacter calcoaceticus chromosome. Gene 138: 59±65 drodiol dehydrogenase of Pseudomonas sp. strain P51 are linked
Sherratt D (1989) Tn3 and related transposable elements: site- evolutionarily to the enzymes for benzene and toluene degra-
speci®c recombination and transposition. In: Berg DE, Howe dation. J Biol Chem 271: 4009±4016
MM (eds) Mobile DNA. American Society for Microbiology, Williams PA, Sayers JR (1994) The evolution of pathways for ar-
Washington DC, pp 163±184 omatic hydrocarbon oxidation in Pseudomonas. Biodegradation
Sinclair MI, Holloway BW (1991) Chromosomal insertion of TOL 5: 195±217
transposons in Pseudomonas aeruginosa PAO. J Gen Microbiol Williams PA, Worsey MJ (1976) Ubiquity of plasmids in coding for
137: 1111±1120 toluene and xylene metabolism in soil bacteria: evidence for the
Sinclair MI, Maxwell PC, Lyon BR, Holloway BW (1986) Chro- existence of new TOL plasmids. J Bacteriol 125: 818±828
mosomal location of TOL plasmid DNA in Pseudomonas put- Williams PA, Assinder SJ, Marco P, O'Donnell KJ, Poh CL, Shaw
ida. J Bacteriol 168: 1302±1308 LE, Winson MK (1992) Catabolic gene duplications in TOL
Slater JH, Lovatt D, Weightman AJ, Senior E, Bull AT (1979) The plasmids. In: Galli E, Silver S, Witholt B (eds) Pseudomonas:
growth of Pseudomonas putida on chlorinated aliphatic acids molecular biology and biotechnology. American Society for
and its dehalogenase activity. J Gen Microbiol 114: 125±136 Microbiology, Washington DC, pp 341±352
Springael D, Kreps S, Mergeay M (1993) Identi®cation of a cata- Wyndham RC, Cashore AE, Nakatsu CH, Peel MC (1994a) Cat-
bolic transposon, Tn4371, carrying biphenyl and 4-chlorobi- abolic transposons. Biodegradation 5: 323±342
phenyl degradation genes in Alcaligenes eutrophus A5. Wyndham RC, Nakatsu C, Peel M, Cashore A, Ng J, Szilagyi F
J Bacteriol 175: 1674±1681 (1994b) Distribution of the catabolic transposon Tn5271 in a
Syvanen M (1988) Bacterial insertion sequences. In: Kucherlapati groundwater bioremediation system. Appl Environ Microbiol
R, Smith GR (eds) Genetic recombination. American Society 60: 86±93
for Microbiology, Washington DC, pp 331±356 Yano K, Nishi T (1980) pKJ1, a naturally occurring conjugative
Tan HM, Mason JR (1990) Cloning and expression of the plasmid- plasmid coding for toluene degradation and resistance to
encoded benzene dioxygenase genes from Pseudomonas putida streptomycin and sulphonamides. J Bacteriol 143: 552±560
ML2. FEMS Microbiol Lett 72: 259±264 Yen KM, Serdar CM (1988) Genetics of naphthalene catabolism in
Tan HM, Fong KPY (1993) Molecular analysis of the plasmid- pseudomonads. Crit Rev Microbiol 15: 247±268
borne bed gene cluster from Pseudomonas putida ML2 and
Virology 434 (2012) 210–221

Contents lists available at SciVerse ScienceDirect

Virology
journal homepage: www.elsevier.com/locate/yviro

Review

Pirates of the Caudovirales


Gail E. Christie a,n, Terje Dokland b
a
Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, P.O. Box 980678, 1011 E Marshall St., Richmond, VA 23298-0678, USA
b
Department of Microbiology, University of Alabama at Birmingham, 845 19th Street South, BBRB 311, Birmingham, AL 35294, USA

a r t i c l e i n f o a b s t r a c t

Available online 3 November 2012 Molecular piracy is a biological phenomenon in which one replicon (the pirate) uses the structural
Keywords: proteins encoded by another replicon (the helper) to package its own genome and thus allow its
Molecular piracy propagation and spread. Such piracy is dependent on a complex web of interactions between the helper
Bacteriophage 80a and the pirate that occur at several levels, from transcriptional control to macromolecular assembly.
Staphylococcus aureus pathogenicity island The best characterized examples of molecular piracy are from the E. coli P2/P4 system and the S. aureus
mobilization SaPI pathogenicity island/helper system. In both of these cases, the pirate element is mobilized and
Bacteriophage P2 packaged into phage-like transducing particles assembled from proteins supplied by a helper phage
Satellite phage P4 that belongs to the Caudovirales order of viruses (tailed, dsDNA bacteriophages). In this review we will
Capsid assembly
summarize and compare the processes that are involved in molecular piracy in these two systems.
Size determination
& 2012 Elsevier Inc. All rights reserved.
Derepression
Transactivation
Interference
DNA packaging
SaPI

Contents

Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 210
Overview of the P2/P4 system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 211
Mobilization of S. aureus pathogenicity islands (SaPIs) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 212
Derepression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 212
P4 immunity and derepression of P4 by P2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 212
P2 immunity and reciprocal derepression of P2 by P4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 213
Derepression of SaPIs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 213
Transactivation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 213
Assembly and capsid size determination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 214
DNA packaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 217
Interference . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 218
Conclusion and perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 218
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 219
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 219

Introduction describe the P2/P4 system of bacteriophages, where the ‘‘pirate’’


phage/plasmid replicon P4 usurps the structural gene products
Inspired by stories of piracy in the South China Sea (Rosenberg, of an unrelated ‘‘helper’’ bacteriophage for its own propagation
2009), the term ‘‘molecular piracy’’ was coined by Bjørn Lindqvist (Christie and Calendar, 1990; Lindqvist et al., 1993). In fact, the
while he was on sabbatical in the Dokland lab in Singapore, to term ‘‘molecular piracy’’ was not entirely new, but has been
used previously to refer, variously, to the way by which viruses
take control of their hosts’ biosynthetic machinery (Flaitz and
n Hicks, 1998; Fujimuro et al., 2007), and to the acquisition of host
Corresponding address: Fax: þ1 804 828 9946.
E-mail addresses: christie@vcu.edu (G.E. Christie), genes during viral evolution (Ahuja and Murphy, 1993; Choi et al.,
dokland@uab.edu (T. Dokland). 2001; Sinkovics et al., 1998). Of course, by the first definition, all

0042-6822/$ - see front matter & 2012 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.virol.2012.10.028
G.E. Christie, T. Dokland / Virology 434 (2012) 210–221 211

Table 1
Comparison of the steps involved in molecular piracy by SaPIs and P4.

SaPI/80a P4/P2

Derepression 80a Sri, Dut, gp15 bind and inactivate different SaPI Stl repressors P2 Cox activates immunity-insensitive transcription from P4 PLL promoter
Reciprocal No known mechanism P4 Epsilon binds and inactivates the P2 master repressor C
derepression
Mutual transactivation No known mechanism P2 Ogr and P4 Delta each activate both P2 and P4 late promoters
Excision SaPI encoded. Derepression by helper required Independent of helper; P4 encoded. Derepression by helper enhances
Replication SaPI encoded Derepression by helper required Independent of helper, P4 encoded Derepression by helper enhances
Capsid size redirection Internal scaffold; SaPI CpmA and CpmB External scaffold; P4 Sid P4 Psu provides additional stability
DNA packaging Headful packaging. SaPI-encoded TerS redirects specificity Cos-site packaging. No known P4-encoded functions
SaPI Ppi blocks phage DNA packaging

viruses are pirates, since all viruses require functions supplied by transcriptional activation, excision and replication of the pirate
the host cell for their own propagation, and exchange of genetic DNA, and finally assembly and packaging of pirate DNA into
material is a fundamental mechanism in the evolution of viruses virus-like particles made from helper proteins. A variety of
and other organisms. Some viruses, such as HIV, vaccinia or interactions between the pirates and their helpers modulate
herpesviruses even incorporate host proteins into their virions these processes, ranging from gene regulation to morpho-
(Maxwell and Frappier, 2007; Ott, 2008), but such incorporation genetic control (Table 1). In the following sections, we will
tends to be incidental or play an auxiliary role, rather than serving discuss each of these interactions separately and also outline
as an integral part of the viral structure. where the systems differ.
In our definition, molecular piracy refers specifically to the
case in which one infectious genetic element (the ‘‘pirate’’) uses
the structural proteins encoded by a viral replicon (the ‘‘helper’’)
for assembly of its own virion. This characteristic distinguishes Overview of the P2/P4 system
the pirate/helper systems from the satellite viruses commonly
found in eukaryotes (Hu et al., 2009), or the recently described Bacteriophage P2 was originally isolated from the Lisbonne &
‘‘virophage’’, which depends on and interferes with the replica- Carre re strain of Escherichia coli by Bertani in 1951 and is a
tion of mimivirus (La Scola et al., 2008). Although these satellites member of the Myoviridae family of viruses, having an icosahedral
depend upon the helpers for their propagation, they encode their head (capsid) and a contractile tail, and a 33.6 kb double-stranded
own capsid proteins. Even hepatitis delta virus, which packages DNA genome (Bertani, 1951; Bertani and Six, 1988; Nilsson
its genome-containing nucleocapsids within a viral envelope and Haggard Ljungquist, 2005). P2 is a so-called ‘‘non-inducible’’
formed by glycoproteins encoded by a Hepatitis B virus helper, phage; unlike l and many other prophages P2 is not mobilized by
encodes its own nucleocapsid protein (Sureau, 2006). UV light. Several other P2-related phages have also been shown to
In the P2/P4 system, not only does the pirate depend on the function as helpers for P4, including PK (the original helper in the
helper for structural proteins, but has the ability to redirect the strain from which P4 was isolated) (Six, 1963; Six and Klug,
capsid assembly process to suit its own needs. As it turns out, 1973), P3 (Lin, 1984) and coliphage 186 (Sauer et al., 1982). P2-
the P2/P4 system is not the only example of such a phenomenon. like prophages are common in the environment (Breitbart et al.,
More recently, a similar system was discovered in Staphylococcus 2002) and are present in about 30% of strains in the E. coli
aureus, where genetic elements called pathogenicity islands reference collection (Nilsson et al., 2004), in enterohemorrhagic
(SaPIs) are mobilized by specific helper phages (Lindsay et al., E. coli, and in a variety of other g-proteobacteria, including strains
1998; Novick et al., 2010) and are packaged into phage-like of Salmonella, Pseudomonas, Serratia, Haemophilus, Vibrio, Yersinia
transducing particles using structural proteins supplied by the and the Burkholderia cepacia complex (Garcia et al., 2008;
helper phage (Poliakov et al., 2008; Tallent et al., 2007; Tormo Lynch et al., 2010; Nilsson and Haggard Ljungquist, 2005).
et al., 2008). These two molecular pirates are not degenerate Most of the characterization of helper exploitation by P4 has
versions of their helpers, but rather independent replicons that been carried out using P2, however, which will be the focus of
have evolved a highly specialized machinery to exploit helper the discussion here.
bacteriophages for their own benefit. An additional example of P4 is an 11.6 kb replicon that can exist either as a plasmid or
molecular piracy has been described in Sulfolobus, where two integrated into the host genome like a prophage (Briani et al.,
nonconjugative plasmids have been shown to exploit archaeal 2001; Deho and Ghisotti, 2005; Lindqvist et al., 1993). P4 is
fuselloviruses for packaging and spread. However, little is known genetically unrelated to P2, and while it has been described as a
about the underlying molecular mechanisms in this system satellite phage it is probably more appropriate to consider it as an
(Arnold et al., 1999; Wang et al., 2007). integrative plasmid that has acquired functions for helper phage
The focus of this review will be on the mechanisms used by piracy. P4 lacks genes encoding major structural proteins and
the P4-related elements and the SaPIs to manipulate their respec- requires all of the morphogenetic genes of its helper phage
tive helper phages, which are members of the order Caudovirales— (Six, 1975). A second P4-like element found in E. coli, retronphage
tailed, double-stranded DNA (dsDNA) bacteriophages. The biology fR73, can also exploit P2 as a helper (Inouye et al., 1991). The
of the P2/P4 system has been described in great detail in the exploitation of P2 by P4 can take place under a variety of circum-
decades since its discovery (Christie and Calendar, 1990; Deho stances, including P2 infection of a strain carrying P4 in either the
and Ghisotti, 2005; Lindqvist et al., 1993). Reports elucidating immune-integrated or multicopy plasmid state, P4 infection of
SaPI biology have a much briefer history, but there have been a P2 lysogen, and coinfection by both phages. In each of these
significant recent advances in our understanding of the interac- scenarios, P4 responds to the presence of the helper phage by
tions between these pathogenicity islands and their helpers in interacting with certain phage-encoded functions and by activat-
S. aureus (Novick et al., 2010). The molecular piracy that takes ing P4 functions that allow it to manipulate the helper phage
place in these systems involves several steps, typically including appropriately. The nature and timing of the regulatory crosstalk
212 G.E. Christie, T. Dokland / Virology 434 (2012) 210–221

between P4 and its P2 helper depends on the infection conditions,


and appears to be designed to optimize P4 reproduction.

Mobilization of S. aureus pathogenicity islands (SaPIs)

SaPIs are a family of 14–27 kb genetic elements that are


integrated into the S. aureus host genome and contain phage-
like repressor, integrase and terminase genes. Different SaPIs also
express a variety of superantigen toxins and other virulence
and antibiotic resistance factors. Ten of the 17 identified to date
in staphylococcal genomes have been shown to be inducible by
either known or endogenous prophages (Novick et al., 2010). The
two best characterized SaPIs are SaPI1 and SaPIbov1, found in S.
aureus strains RN4282 and RF122, respectively (Novick et al.,
2010). SaPI1 carries genes for the toxic shock syndrome toxin 1
(tst) and enterotoxins K (sek) and Q (seq) (Ruzin et al., 2001).
SaPIbov1 carries tst as well as genes encoding enterotoxins C (sec)
and L (sel), and is associated with bovine pathogenic S. aureus
(Fitzgerald et al., 2001). Some SaPIs (SaPI2, SaPI5) are found in
clinically important MRSA strains, including USA200 and USA300
(Highlander et al., 2007). Related elements have also been
Fig. 1. Mutual derepression in P2 and P4. Maps depict the portions of the P2 (top)
identified in other staphylococcal species (Kuroda et al., 2005, and P4 (bottom) genomes encoding the transcripts involved in lysogeny control
Takeuchi et al., 2005) and in streptococci (Scott et al., 2012). and DNA replication. Transcripts and their promoters are indicated by arrows
While normally repressed and stably integrated in the host below the map; the dotted line shows the part of the early leftward transcript
genome, SaPIs become activated and mobilized when a compa- that is no longer synthesized when P4 establishes lysogeny. The repressors and
their targets in both genomes are shown by the red lines. Genes encoding the
tible helper bacteriophage enters the lytic cycle, whether through
derepression functions are designated with a yellow circle, and the targets on
infection or by induction of an endogenous prophage. Upon which they act are denoted by green lines to indicate activation of transcription.
mobilization, the SaPI genomes are packaged into transducing The arrowhead indicates that Cox acts at the PLL promoter to stimulate transcrip-
particles formed by structural proteins encoded by the helper tion, while the perpendicular line indicates that Epsilon blocks the action of P2 C,
phage (Tallent et al., 2007, Tormo et al., 2008). Helper phages for thereby turning on Pe transcription.

SaPI mobilization belong to a large family of temperate transdu-


cing phages found in S. aureus that are members of the family
Siphoviridae, with dsDNA genomes ranging from 39.6 to 45.9 kb (Deho et al., 1988). One of these is constitutive and the other is
(Kwan et al., 2005). Several helper phages for different SaPIs have subject to complex regulatory control. In a P4 lysogen, transcription
been described, including f11, 53, 80, 80a, and fNM1 (Christie from the constitutive early leftward promoter, PLE, yields a transcript
et al., 2010; Dearborn and Dokland, 2012; Lindsay et al., 1998). of  300 nucleotides that terminates upstream of the P4 replication
The phage-induced mobilization of SaPIs is specific: thus, SaPI1 genes (Briani et al., 2000; Deho et al., 1992) and is subsequently
can be mobilized by phage 80a but not by the closely related f11, processed to generate the CI RNA (Forti et al., 2002). Termination
nor the more distantly related phage 80; SaPI2 can be mobilized depends upon RNA–RNA interactions between the CI RNA and two
both by phage 80 and by 80a. SaPIbov1 can be mobilized by f11 specific target sequences in the untranslated leader region of the
and 80a, but not by 80 (Christie et al., 2010). f13 is able to induce nascent transcript. (Sabbattini et al., 1995). In order to overcome P4
SaPI1 excision and replication, but not to package the genome immunity, this RNA-mediated termination must be circumvented.
into phage particles (Lindsay et al., 1998). The helper/SaPI This is accomplished by initiation of transcription about 400 bp
specificity is manifested at several levels, which will be discussed upstream of PLE from a second promoter, PLL. Translation of two
in greater detail in the following sections. nested genes in this longer transcript leads to translational suppres-
sion of the CI RNA-mediated transcription termination and therefore
expression of the P4 replication functions (Forti et al., 1999).
Derepression Derepression of immune-integrated P4 by a P2 helper phage
requires activation of transcription from PLL, which bypasses the
SaPIs are normally integrated stably in their host genome in a P4 immunity system and leads to transcription of the P4 replica-
repressed state, as is the case with P4 in the immune-integrated tion genes (Saha et al., 1989). This requires the product of the
state. For both of these elements, an essential first step in P2 cox gene (Saha et al., 1989). Activation of PLL by Cox also leads
mobilization is relief of repression by a helper phage-encoded to the induction of P4 prophage excision that is observed upon
function. Each has evolved a mechanism that exploits helper P2 infection of a P4 lysogenic strain (Six and Lindqvist, 1978).
phage genes which also perform other roles in the phage life Excision requires the P4 Vis protein, which is encoded by the first
cycle. In addition, P4 has the ability to derepress a resident helper reading frame in the PLL transcript (Cali et al., 2004). Derepression
prophage (mutual derepression). of P4 following P2 infection appears to be primarily a mechanism
for allowing P4 to survive P2- mediated host cell death rather
P4 immunity and derepression of P4 by P2 than a mode for efficient horizontal transfer, since the yield of P4
when P2 infects a P4 lysogen is normally quite low, less than 1 P4
P4 immunity involves a unique mechanism in which a short, per cell (Six and Lindqvist, 1978). However, if P2 replication is
stable RNA (CI RNA) regulates transcription termination through blocked (by infection of a bacterial host that lacks the rep gene, or
sequence-specific binding. Leftward transcription of the P4 a by mutation in a phage replication function) the yield of P4
operon (Fig. 1), which encodes functions required for both increases to a level comparable to that seen during P4 infection of
plasmid and lytic growth, initiates from two different promoters a P2 lysogen (Six and Lindqvist, 1978).
G.E. Christie, T. Dokland / Virology 434 (2012) 210–221 213

Fig. 2. SaPI derepression by multiple helper phage genes. The known derepression functions are identified by yellow circles on a map of the 80a genome. Shown below are
the non-homologous immunity regions from three different SaPIs, with the rightward str promoter repressed by the product of the stl gene, as indicated by the red lines.
Each of the phage derepression functions activates str expression from a different SaPI by interfering with Stl repression, as indicated by the green lines.

P2 immunity and reciprocal derepression of P2 by P4 Three different derepression proteins encoded by phage 80a have
been identified, and each targets a different SaPI (Fig. 2). All of
When the P2 helper is present as a prophage, P4 is able to these proteins share a common mechanism; they act as anti-
derepress it to activate expression of required helper functions. repressors by direct binding and inhibition of their respective
P4 infection of P2 lysogens gives rise to about 100 P4 and about Stl proteins (Harwich, 2009; Tormo-Mas et al., 2010). SaPI1 is
10  3 P2 per infected cell (Six and Klug, 1973). As in other temperate derepressed by Sri, the product of 80a ORF22, SaPIbov1 is
phages, P2 early transcription initiates from a pair of divergent derepressed by Dut, the product of 80a ORF32, and SaPIbov2 is
promoters encoding competing repressors that regulate the lysogeny derepressed by the product of 80a ORF15. Each of these genes is
functions (Fig. 1). The leftward transcript encodes the P2 immunity nonessential for phage growth but required for mobilization of
repressor, C, and the phage integrase, while the rightward transcript the respective SaPI (Tormo-Mas et al., 2010). Two of these phage-
includes genes encoding the repressor of the lysogenic promoter encoded antirepressors have other known functions. Sri was
(Cox), as well as the replication functions. C regulates its own previously identified in the related phage 77 as a protein that
promoter and blocks expression of Cox, while Cox blocks expression inhibited host DNA replication by binding to DnaI (Liu et al.,
of C (Saha et al., 1987). P2 Cox is a remarkable protein with multiple 2004), while Dut is a dUTPase (Tormo-Mas et al., 2010). Like P4,
roles; it functions not only as the repressor of the lysogenic promoter SaPIs have apparently evolved to sense the presence of a helper
but also as the recombination directionality factor for prophage phage by exploiting genes that play another role in the biology of
excision (Yu and Haggard-Ljungquist, 1993) and, as discussed above, the phage.
positively regulates the P4 PLL promoter to derepress P4.
The derepression of P2 by P4 requires the P4 e gene product
(Geisselsoder et al., 1981; Liu et al., 1997). Epsilon binds to the P2 Transactivation
immunity repressor and interferes directly with binding of the
repressor to its operator (Liu et al., 1998). This leads to expression Both P4 and the SaPIs depend upon their helper phages for
of the helper early genes and to in situ replication of the P2 gene products needed for virion assembly, DNA packaging, and
prophage, which does not excise (Six and Lindqvist, 1978). The e cell lysis. During lytic growth of the helper phages, these func-
gene is essential for P4 growth in a P2 lysogen, but not during a tions are expressed late in infection as part of the normal
P2þP4 co-infection of a nonlysogenic cell. However, Epsilon does temporal regulation of the phage morphogenetic genes. In the
appear to contribute to interference with growth of the helper P2/P4 system, at least, there is a second set of reciprocal interac-
phage during a coinfection (Diana et al., 1978). The interaction tions that regulate late gene transcription, allowing P4 to opti-
between Epsilon and the phage repressor determines whether P4 mize exploitation of the helper phage under the different
can use a lysogenic helper phage. The P2-related phage 186, conditions it might encounter. This is accomplished by a pair of
which has morphogenetic genes similar to those of P2 but an related transcriptional activators encoded by P2 and P4 that
unrelated repressor (Kalionis et al., 1986), cannot be derepressed recognize the same promoters on both genomes but differ in
by P4 and can only serve as a P4 helper if it is growing lytically the efficiencies with which they activate gene expression.
(Sauer et al., 1982). The P2 morphogenetic genes, encoding the head, tail, packa-
ging and lysis functions, lie in four operons expressed late in
Derepression of SaPIs infection (Fig. 3). P2 late gene transcription requires the product
of the phage ogr gene, a transcriptional activator that binds to a
In the absence of helper phage, SaPIs are maintained in a site about 55 bp upstream of the initiation sites for the four P2
stable repressed state by a master repressor, Stl. Like prophage late promoters (Christie and Calendar, 1985; Christie et al., 2003)
repressors, Stl binds to a region between two divergent promoters and interacts with the C-terminal domain of the a subunit(s) of E.
where it inhibits most SaPI gene expression. Inactivation of stl by coli RNA polymerase (Ayers et al., 1994; Sunshine and Sauer,
mutation leads to SaPI excision and replication (Ubeda et al., 1975; Wood et al., 1997). Ogr belongs to a family of zinc-binding
2008). Thus, derepression by the helper phage is a key regulatory transcription factors found almost exclusively among P2-related
step in SaPI mobilization. Remarkably, the Stl proteins of different phages and their satellites, with a C2C2 motif essential for metal
SaPIs are widely divergent, and the ability of a particular helper binding and activity (Julien et al., 1998; Pountney et al., 1997).
phage to derepress a given SaPI appears to be a primary determi- P4 has two operons that are expressed during lytic growth
nant of helper phage-SaPI specificity (Tormo-Mas et al., 2010). (Fig. 3), and the two P4 late promoters have the same conserved
214 G.E. Christie, T. Dokland / Virology 434 (2012) 210–221

Fig. 3. Mutual transactivation in P2 and P4. The four P2 and two P4 late transcription units are indicated by black arrows below their respective genetic maps. The genes
for the late transcription factors ogr (P2) and d (P4) are identified by yellow circles on the maps. Both proteins activate transcription from the same P2 and P4 late
promoters, as indicated by the green arrows.

sequence element found upstream of the P2 late promoters. and packaging specificity (discussed below) lie in a six-gene
The leftward PLL promoter is the same promoter that is dere- operon designated as operon 1, which is preceded by a LexA-
pressed by P2 Cox to initiate P4 excision and replication from regulated promoter (Ubeda et al., 2007). During 80a infection,
the prophage state. The second late promoter, Psid, regulates transcription of these genes in SaPI1 requires derepression of the
rightward transcription of three genes involved in helper exploi- SaPI and initiates farther upstream, at a promoter that has not yet
tation: sid, d and psu. Sid and Psu play roles in P4 capsid assembly been identified (Harwich, 2009). Transcription from the Lex-A
(see below). The third gene product, Delta, is an Ogr homolog that regulated promoter would lead to a burst of operon 1 expression
activates transcription from the two P4 late promoters and the during SOS induction of a resident helper prophage, which might
four P2 late promoters. Likewise, Ogr activates transcription from improve SaPI yield but is not essential for mobilization. A f11DrinA
the two P4 late promoters as well as the four P2 late promoters mutant did not show any impairment in transcription of SaPIbov1
(Dale et al., 1986; Deho et al., 1988; Halling and Calendar, 1990). operon 1, even under conditions where transcription from the
Although there is extensive similarity among proteins in the LexA-regulated promoter was blocked by mutation (Ferrer et al.,
P2 Ogr family, they fall into two functionally discrete classes. 2011). This indicates that the helper phage RinA transcription
Members of the ‘‘helper’’ class, exemplified by Ogr, activate the P4 factor does not play a direct role in controlling SaPI operon
late promoters better than the P2 late promoters. Members of the 1 expression.
‘‘satellite class,’’ exemplified by the Delta proteins of P4 and fR73,
activate the P2 late promoters better than the P4 late promoters
and are able to cause transcription in the absence of replicating P2
DNA (Julien and Calendar, 1996; McAlister et al., 2003). These Assembly and capsid size determination
differences contribute to earlier expression of P4 late genes in the
presence of a P2 helper and maximize expression of the P2 late Tailed, dsDNA bacteriophages of the Caudovirales assemble
genes in the presence of P4. They also allow P4 to activate directly their capsids (or heads) as empty precursors—procapsids—from
the transcription of the P2 morphogenetic genes required for the major capsid protein (CP), typically requiring a scaffolding
packaging and lysis, bypassing their normal requirement for P2 protein (SP) that acts as a chaperone for the assembly process
DNA replication. (Fig. 4) (Dokland, 1999; Fane and Prevelige, 2003). The main
There is at this point no evidence to suggest a similar set of exception to the requirement for SP is the HK97-like phages, in
complex, reciprocal interactions as a general mechanism regulat- which an N-terminal sequence in CP appears to serve this role
ing helper phage exploitation by SaPIs. In contrast to the P2/P4 (Conway et al., 1995; Duda et al., 1995). During DNA packaging,
system, infection of a helper phage lysogen by a SaPI-containing the capsid undergoes expansion accompanied by major confor-
particle has not been reported to lead to a burst of progeny SaPI mational changes in CP (Johnson, 2010). Tail structures (and
virions. Helper phage late transcription, studied in most detail for sometimes ‘‘decoration’’ proteins) are added to the finished
80a and f11, appears to initiate from a single late promoter that capsid. Capsids are either icosahedral or elongated with icosahe-
is activated by the RinA transcription factor, which is encoded by dral caps, and—in spite of weak or undetectable sequence
a gene that lies immediately upstream of the late operon. Deletion homology—all members of the Caudovirales studied to date share
of rinA eliminates phage production and essentially eliminates a characteristic and unique CP fold, called the HK97-like fold
SaPI1 transduction by 80a (Ferrer et al., 2011). This argues that (Johnson and Chiu, 2007, Wikoff et al., 2000).
SaPI1 does not encode a function that can replace rinA in helper One of the most striking features about the piracy both in the
phage late gene transcription. Consistent with this, no increase in P2/P4 system and in the mobilization of SaPIs is the redirection
80a late transcription was detected following prophage induction of the helper phage assembly pathway to form capsids that are
in the presence of SaPI1 (Harwich, 2009). However, there is still about 1/3 the size (45 nm, T¼ 4) of those normally made by the
significant residual transduction of SaPIbov1 by both 80aDrinA phage itself (60 nm, T¼7), commensurate with the difference in
and f11DrinA (Ferrer et al., 2011), suggesting that unlike SaPI1, size of the genomes (Figs. 4 and 5A) (Dearborn et al., 2011;
SaPIbov1 may encode a function that can activate helper phage Dearborn et al., 2012; Dokland et al., 1992; Ruzin et al., 2001;
late transcription to some extent. Spilman et al., 2011). The small capsids are unable to package
RinA does not appear to have a reciprocal influence on SaPI1 complete phage P2 genomes, thus this redirection of the assembly
transcription. The SaPI genes involved in capsid size determination pathway strongly interferes with P2 multiplication.
G.E. Christie, T. Dokland / Virology 434 (2012) 210–221 215

Fig. 4. Comparison of assembly pathways for P2/P4 (A) and 80a/SaPI1 (B). In each panel the top pathway is the helper phage and the bottom pathway is the pirate. Phage
procapsids are assembled from the major capsid protein (gpN in P2, gp47 in 80a; yellow), scaffolding protein (P2 gpO, 80a gp46; red), and portal protein (P2 gpQ, 80a
gp42; green). 80a also incorporates a minor capsid protein (gp44; cyan) that may play a possible role in stabilizing the DNA in the capsid. The presence of the P4 Sid (panel
A) or SaPI CpmA and CpmB (panel B) proteins (orange) lead to the formation of small procapsids. Sid forms an external scaffold while CpmB forms an internal one; the
location of CpmA in the small SaPI1 procapsids is currently unknown. DNA is packaged into procapsids by terminase complexes (gpP and gpM for P2, TerS and TerL for
80a), concomitant with removal of the scaffolding proteins and expansion of the capsid. In P2, only the C-terminal half of gpO (DO) is removed, leaving the O* protease
domain inside the capsid. P4 Psu is added to small capsids as a decoration protein.

How do the pirate elements carry out this size change? In P4, domain, On, which remains inside the mature capsids (Fig. 4A)
the size redirection depends on a P4 size determination gene, (Chang et al., 2008, 2009; Dokland, 2012). Indeed, the mutant
sid (Barrett et al., 1976), which encodes an external scaffolding Oam279, which lacks the C-terminal 47 amino acids and is
protein that forms an external dodecahedral cage around the defective in scaffolding activity, retains protease activity and is
procapsid (Fig. 4A) (Marvik et al., 1995). Sid is an elongated viable in the presence of Sid (Agarwal et al., 1990).
protein made up of bundles of a-helices (Fig. 5B) (Dearborn et al., The P4-encoded psu (polarity suppression) gene product,
2012). Trimers of Sid connect hexamers of the gpN capsid protein which acts as a suppressor of rho-dependent transcription termi-
across the threefold axes, forcing the shell into a T¼ 4 architecture nation (Pani et al., 2009; Sauer et al., 1981), also serves a role as a
(Fig. 6A). P4 sid mutants fail to form small capsids, and while P4 decoration protein that is added to the outside of the completed
DNA can still get packaged as dimers or trimers into large capsids, capsid (Fig. 4A) (Dokland et al., 1993). Psu apparently stabilizes
the efficiency is low (Shore et al., 1978). Mutants in gpN, called sir the inherently less stable P4 capsids against environmental stress
(sid responsiveness) render the capsid protein resistant to the Sid- (Isaksen et al., 1993).
induced size redirection and thus do not form small capsids (Six Size determination by SaPIs works differently. In the most well
et al., 1991). These mutations are clustered in an external loop in described system—SaPI1 mobilized by phage 80a—two SaPI1
the gpN CP, where they presumably interfere with gpN–Sid proteins, gp6 and gp7, are both required for efficient small capsid
interactions (Fig. 5B) (Dearborn et al., 2012). Conversely, muta- formation (Fig. 4B) (Damle et al., 2012; Poliakov et al., 2008).
tions in Sid, named super-sid or nms (N mutation sensitive) (Kim Homologous proteins are found in most, but not all, SaPIs, and the
et al., 2001), which are clustered in a C-terminal a-helix, corresponding capsid morphogenesis genes have been named
(Dearborn et al, 2012) recover the ability of Sid to form small cpmA (gp7) and cpmB (gp6) (Damle et al., 2012; Dearborn and
capsids even on a P2 Nsir background. Dokland, 2012; Ram et al., 2012). These two proteins, CpmA and
Expression of gpN and Sid alone is sufficient to efficiently form CpmB, are sufficient to induce small capsid formation when
small procapsids (Dokland et al., 2002), although the gpO SP is expressed during phage infection or upon co-expression with just
incorporated when both proteins are present (Fig. 6A) (Wang CP and SP in a S. aureus co-expression system (Damle et al., 2012;
et al., 2006). However, gpO is required for the formation of viable Ram et al., 2012; Spilman et al., in press).
P4 phage (Six, 1975), presumably due to other functions of gpO, in Unlike the P2/P4 system, there is no Sid-like external scaffold-
particular the protease activity that resides in its N-terminal ing protein. Instead, SaPI1 procapsids contain internal fingerlike
216 G.E. Christie, T. Dokland / Virology 434 (2012) 210–221

Fig. 5. (A) Isosurface representations of cryo-EM reconstructions of P2, P4, 80a and SaPI1 procapsids, and radially colored from red (inside) to blue (outside) (Spilman et al,
2011; Dearborn et al, 2011; Dearborn et al, 2012). For 80a and SaPI1, the right halves show a cutaway view of the interior of the procapsids, revealing the internal
protrusions in SaPI1 corresponding to CpmB. (B) Closeup view of the P4 hexamer (ivory isosurface with three copies of gpN fitted in, shown as blue, red, and yellow
ribbons) with the density corresponding to Sid shown in red. The Nsir mutations are indicated as purple balls on one gpN monomer. (C) Ribbon representation of the NMR
structure of two SaPI1 gp6 (CpmB) dimers (orange and yellow; Dearborn et al., 2011) fitted into the internal protrusions in the SaPI1 procapsid reconstruction, shown as a
solid isosurface. The predicted C-terminal a-helices are shown in pink for one subunit of each CpmB dimer. The gp47 capsid protein model is shown in green. The 3D
reconstructions were generated using AUTO3DEM (Yan et al, 2007). Rendering and fitting of the maps was done in UCSF Chimera (Pettersen et al, 2004).

Fig. 6. Models for capsid size determination. (A) The P2 internal scaffolding protein gpO (red) promotes assembly of the gpN capsid protein (yellow) through dimerization
and specific interactions with gpN (top panel). The cylinders represent the C-terminal a-helical domain, while the bullets indicate the N-terminal protease domain of gpO.
In the presence of P4 Sid (orange), gpN is tethered at the threefold (triangle) and twofold (ovals) symmetry axes, forcing the formation of a smaller capsid. (B) In the 80a/
SaPI1 system, the gp46 scaffolding protein, which forms an internal core, is also believed to interact with the capsid protein (gp47) through a predicted C-terminal a-helix.
The C-terminal a-helices of the SaPI1-encoded CpmB protein dimers (orange) compete with gp46 for the same binding site on gp47. CpmA (pink) may be required to
remove gp46 in order to provide access for CpmB.

projections absent from the helper phage procapsids (Fig. 5A). The role of CpmA in size determination is less clear. CpmA is
CpmB, which has a structure similar to that of the SP of bacterio- only present in procapsids in small amounts, suggesting that its
phage f29, acts as an internal scaffolding protein (Dearborn et al., action is transient in nature (Poliakov et al., 2008). Deletion of
2011, Morais et al., 2003). CpmB binds as a dimer to the inside of cpmB in SaPI1 led to the formation of a large number of non-
the SaPI1 shell (Fig. 5C) and most likely competes with the isometric ‘‘monsters’’ (Damle et al., 2012; Dearborn et al., 2011),
cognate 80aSP for the same binding site on the 80a CP (Fig. 6B). and while CpmB alone could promote small capsid formation in
G.E. Christie, T. Dokland / Virology 434 (2012) 210–221 217

the absence of SP, CpmA had an inhibitory effect on capsid (Catalano, 2005; Feiss and Rao, 2012; Roy et al., 2012; Teschke,
assembly (Spilman et al., in press). Small procapsids lack the 2012). P4 and SaPIs have evolved different strategies to exploit the
internal scaffolding core that can be seen in reconstructions of DNA packaging machinery of their helper phages. P4 has simply co-
large procapsids (Spilman et al., 2011). The role of CpmA may be opted the P2 packaging machinery by incorporating the same
to reorganize the scaffolding core that would otherwise prevent packaging signals into its own genome. P2 and P4 contain identical
small capsid formation, or to bind SP to allow access to binding 55 bp cos site sequences that include the 19 bp cohesive ends found
sites on CP by CpmB (Fig. 6B). in virion DNA (Ziermann and Calendar, 1990). DNA packaging and
Size redirection depends on compatibility between CpmA/ cos site-specific cleavage requires the small (gpM) and large (gpP)
CpmB and the helper capsid proteins. SaPI2, for example, forms terminase subunits as well as procapsids (Pruss et al., 1975; Bowden
small capsids when mobilized by phage 80a, but not by phage 80, and Modrich, 1985). For both genomes, covalently closed circular
presumably due to incompatibility with the phage 80 CP, which DNA molecules are the preferred packaging substrate, unlike the
shares only 16% sequence identity with that of 80a (Christie et al., linear concatemers preferred by most bacteriophages (Black, 1989;
2010). Size determination of SaPIbov1 by 80a also appears to be Bowden and Modrich, 1985; Fujisawa and Morita, 1997; Pruss and
less efficient than for SaPI1 even though the CpmA and CpmB Calendar, 1978).
proteins are almost identical (Dearborn and Dokland, 2012). SaPIs, in contrast, redirect the specificity of the DNA packaging
Other factors, including relative protein expression levels, may machinery of their helpers (Fig. 7). Like their helper phages, SaPIs
also play a role in this process. replicate as linear concatemers, and are packaged as headfuls,
It should be pointed out that capsid size redirection is not resulting in virion DNA that is terminally redundant and partially
absolutely essential in either of these systems. P4 sid mutants are circularly permuted (Ruzin et al., 2001). The phage TerS protein
viable, although reduced in burst size (Diana et al., 1978; Shore recognizes a pac site on the phage genome that lies within the terS
et al., 1978). SaPI1 cpmAB mutants are also viable, and appear to be coding sequence (KD Lane, EK Read, GEC; unpublished), as is the case
transduced at normal frequency (Damle et al., 2012; Ram et al., for the pac site of several other phages that use headful packaging,
2012). Furthermore, in some phage/SaPI systems, size redirection including P22 (Wu et al., 2002) and PY100 (Schwudke et al., 2008). An
does not occur. For example, SaPIbov2 (27 kb) and SaPIbov5 do initial cut is then followed by several rounds of headful packaging.
not contain cpmAB homologs, and do not produce small capsids In the presence of the SaPI, a SaPI-encoded TerS subunit
(Novick et al., 2010; Ram et al., 2012). However, the fact that the together with the phage-encoded TerL directs the specific clea-
cpmAB genes are highly conserved when present and always come vage and packaging of SaPI DNA by binding to a SaPI-specific
together (Novick et al., 2010) suggest that they do confer an pac sequence that lies in an intergenic region upstream of
evolutionary advantage—presumably by interfering with helper the operon that encodes SaPI terS (JC Bento, KD Lane, EK Read,
phage growth. Both sid and cpmAB mutants have lost the ability to GEC; unpublished). The SaPI-encoded TerS is required for high
interfere with their helper phages (Damle et al., 2012; Diana et al., frequency transduction for both SaPI1 and SaPIbov1, while the
1978; Ram et al., 2012), and SaPIs that lack size redirection have phage-encoded TerS is required for packaging of helper phage
other interference mechanisms, as discussed below. DNA (Ubeda et al., 2009).
The compatibility of the SaPI-encoded TerS with the helper
phage TerL likely accounts for some of the observed SaPI-helper
DNA packaging specificity. For example, phage f13, a cos site phage, can induce
SaPI1 excision and replication but fails to produce SaPI1 transdu-
In the Caudovirales, DNA is packaged into the procapsids through cing particles (Ruzin et al., 2001). This is presumably due to an
a ring-shaped portal at one fivefold vertex in an ATP-dependent inability to form a functional hybrid between the cos-site based
process that requires a terminase complex, which consists of small DNA packaging machinery of the phage and the pac site-based
(TerS) and large (TerL) subunits (Black, 1989; Feiss and Rao, 2012; TerS subunit of SaPI1.
Fujisawa and Morita, 1997). The large terminase subunit is respon- Some SaPIs also influence DNA packaging at another level,
sible for prohead binding, DNA translocation and DNA cleavage, by interfering directly with the packaging of helper phage DNA.
while the small subunit is involved in DNA recognition and binding This novel mechanism requires the SaPI ppi (phage packaging

Fig. 7. Model for SaPI packaging redirection. Specific pac sites on the concatemeric phage DNA (top) are recognized by the phage-encoded small terminase (TerSphage; pink)
and packaged into procapsids through the action of the phage-encoded large terminase subunit (TerL). The DNA is cleaved when the capsid is full and the DNA is ready for
another round of packaging. Phage DNA can also be packaged into small capsids, but since the DNA will be only a fragment of the genome, the resulting virions are not
viable. The pac sites on SaPI DNA (bottom) are recognized specifically by the SaPI-encoded TerSSaPI subunit (blue), which also interacts with the phage-encoded TerL for
DNA packaging. While for most SaPIs the majority of capsids formed will be small, any SaPI DNA packaged into large capsids will be multimeric and able to be transduced.
Some SaPIs also encode an interference factor, Ppi, which specifically prevents packaging of phage DNA. Coloring of capsid proteins is as in Fig. 4B.
218 G.E. Christie, T. Dokland / Virology 434 (2012) 210–221

interference) gene (originally called pif; (Tormo-Mas et al., 2010)), of bacterial chromosomes, and can interrupt genes or bring in
which encodes a protein that binds directly to the phage TerS new phage-encoded functions via lysogenic conversion. The pirate
protein and blocks packaging of phage DNA (Ram et al., 2012). The elements we have described add a new dimension to phage-
known Ppi proteins fall into two conserved subsets, each mediated HGT. They differ from other phage-like elements in that
of which appears to target a different phage small terminase they do not encode their own capsids. They differ from other types
superfamily (Ram et al., 2012). of mobile DNA in that they have found a way to directly
manipulate bacteriophages, through changes in gene expression
Interference and morphogenesis, as vehicles for their own specific high fre-
quency transduction. P4 and the SaPIs both exhibit specialized
Both P4 and SaPIs interfere with the multiplication of their adaptations to the lifestyles of their helper phages that allow them
helper phages. In the case of P4, capsid size determination to exploit these phages to their advantage.
appears to be the primary interference mechanism. Interference How did these elements arise? One possibility is that the
with P2 by P4 can range from 5- to 10-fold in a simultaneous co- pirates evolved from temperate phages, retaining just those
infection to greater than 500-fold if P4 is given a ten minute phage-like functions required for integration/excision, replication
head start or is present as a multicopy plasmid (Diana et al., and helper exploitation. Alternatively, they may have been
1978; Deho and Ghisotti, 2005). Although there is some evidence independent extrachromosomal replicons that have acquired
that a still unidentified P4 function may augment P4 Sid for full genes conferring the ability to manipulate phage gene expression
interference with P2 growth, the degree of interference seen and utilize phage proteins for their own purpose. The answer may
when both phages are growing lytically correlates with the depend on the specific element, since the lifestyle of P4 differs
percentage of small capsids formed (Nilssen et al., 1996). greatly from the SaPIs. P4 can exist and replicate as a plasmid
Furthermore, P2 sir mutants, do not form small capsids, are also independently of P2, and it has been proposed that P4 evolved
resistant to interference and exhibit normal phage growth (Six from an ancestral plasmid replicon by acquisition of independent
et al., 1991). modules for site-specific integration and for helper exploitation
In the case of SaPIs, the situation is considerably more (Deho and Ghisotti, 2005). The complex web of mutual interac-
complex. There are at least three strategies for interference, not tions between P2 and P4 suggests that this is a finally tuned
all of which are used in the interactions between a particular SaPI and highly evolved relationship. The SaPI lifestyle more closely
and a specific helper phage. While small capsid formation resembles that of a prophage; it has a phage-like repressor and
certainly prevents packaging of a complete helper phage genome integration functions and it does not exist as an independent
and thereby interferes with phage growth, the loss of the ability extrachromosomal replicon. Accordingly, it has been suggested
to form small capsids by mutation of either SaPI1 cpmA or cpmB that SaPIs may have evolved from prophages (Novick et al., 2010).
alone does not relieve SaPI1 interference with 80a (Damle et al., However, the absence of genes encoding any virion structural
2012). The interference retained by cpmA or cpmB mutants does proteins and the acquisition of functions allowing exploitation of
not appear to depend on any SaPI1 functions other than cpmA or helper phages indicates that SaPIs are not merely some kind of
cpmB, suggesting a second direct role for these gene products in defective prophage, but like P4 have co-evolved with their helpers
helper interference. The effect of the size determination genes in a highly specific manner.
also differs for different helper phages. Despite differences in lifestyle and regulatory circuitry, P4 and
A second level at which interference has been documented is the SaPIs share certain common features (Table 1). Both encode
the inhibition of phage DNA packaging by the SaPI-encoded ppi integration/excision and replication functions and do not depend
genes (Ram et al., 2012). These genes fall into two different but on helper functions for these processes. Both have the ability to
related families, each of which appears to target different helper sense lytic multiplication of their respective helper phages
phages depending which family the phage small terminase and respond by excising and escaping from the bacterial host.
subunit belongs to. For example, the SaPI1 ppi gene does not This provides a clear evolutionary advantage, since lytic infection
interfere with the growth of 80a, but does block f12, while by a phage would mean the death of the host cell and the loss of
the SaPIbov2 ppi gene strongly interferes with 80a growth the pirate element. Remodeling of the helper phage capsid is
(Ram et al., 2012). Different allelic variants of both cpmAB and another conserved feature, and it is striking that these two pirates
ppi confer differing levels of interference, which in some cases have evolved quite different mechanisms to accomplish this
are additive and in others redundant. An additional SaPI gene outcome. While not obligatory for transduction of the pirate
involved in interference has also recently been identified. genome, capsid size redirection leads to the packaging of sub-
This gene, ORF17 in SaPI2, blocks growth of phage 80 (which is genomic fragments of the helper phage DNA and thereby inter-
not affected by the SaPI2 ppi or cpmAB genes) but not 80a, feres with phage propagation. This is likely of evolutionary benefit
and has homologs in other SaPIs as well (Ram et al., 2012). to the host, since fewer cells in the surrounding population would
The mechanism for this third interference function remains to be be lysed—and would also benefit the pirate, since it would
elucidated. increase the likelihood that bacteria infected by the transducing
particles carrying the pirate element would not also be infected
by a phage. The importance of interference in the pirate-helper
Conclusion and perspectives relationship is underscored by the fact that the SaPIs have evolved
at least three independent mechanisms for helper phage inter-
Horizontal gene transfer (HGT) is now commonly accepted ference. One remaining unresolved question is what the helper
to play a major role in prokaryotic evolution (Koonin and Wolf, phages get out of the three way relationship between the
2008; Toussaint and Chandler, 2012). The vehicles that drive bacterial host, the helper phage, and the pirate. Why have the
this ongoing exchange of genetic material, the so-called mobilome, helper phages not evolved resistance to this interference by losing
includes viruses, plasmids, transposons, and a variety of other the functions required to derepress the pirates or altering the
selfish elements. Bacteriophages play multiple roles as agents of genes targeted by the interference functions? The finely tuned
HGT. They mediate the exchange of fragments of chromosomal relationship between these pirates and their helpers suggests
DNA via generalized and specialized transduction. Temperate that these elements are highly co-evolved in a way that must be
phage integration and excision contributes to the remodeling of mutual benefit.
G.E. Christie, T. Dokland / Virology 434 (2012) 210–221 219

Molecular piracy, once thought to be a curiosity seen only in Christie, G.E., Matthews, A.M., King, D.G., Lane, K.D., Olivarez, N.P., Tallent, S.M.,
the P2/P4 system, has turned out to be considerably more wide- Gill, S.R., Novick, R.P., 2010. The complete genomes of Staphylococcus aureus
bacteriophages 80 and 80alpha—implications for the specificity of SaPI
spread. Many S. aureus genomes contain one or more SaPIs, and mobilization. Virology 407, 381–390.
the presence of similar elements in other Gram positive genera Conway, J.F., Duda, R.L., Cheng, N., Hendrix, R.W., Steven, A.C., 1995. Proteolytic
has led to their general designation as ‘‘phage-related chromoso- and conformational control of virus capsid maturation: the bacteriophage
HK97 system. J. Mol. Biol. 253, 86–99.
mal islands’’ (Novick et al., 2010). A BLAST search reveals P4-like Dale, E.C., Christie, G.E., Calendar, R., 1986. Organization and expression of the
elements in the genomes of a number of enterobacteria, including satellite bacteriophage P4 late gene cluster. J. Mol. Biol. 192, 793–803.
members of the genera Escherichia, Shigella and Salmonella. Damle, P.K., Wall, E.A., Spilman, M.S., Dearborn, A.D., Ram, G., Novick, R.P.,
Dokland, T., Christie, G.E., 2012. The roles of SaPI1 proteins gp7 (CpmA) and
Identification of the helper phages for these related elements,
gp6 (CpmB) in capsid size determination and helper phage interference.
and further study of the interactions between them, is likely to Virology 432, 277–282.
reveal additional mechanisms by which these pirates can exploit Dearborn, A.D., Dokland, T., 2012. Mobilization of pathogenicity islands by
their helpers. With the explosion of available genome sequences, Staphylococcus aureus strain Newman bacteriophages. Bacteriophage 2, 70–78.
Dearborn, A.D., Laurinmaki, P., Chandramouli, P., Rodenburg, C.M., Wang, S.,
we anticipate the discovery of similar elements in other systems Butcher, S.J., Dokland, T., 2012. Structure and size determination of bacter-
which will provide fertile ground for further study. iophage P2 and P4 procapsids: function of size responsiveness mutations.
J. Struct. Biol. 178, 215–224.
Dearborn, A.D., Spilman, M.S., Damle, P.K., Chang, J.R., Monroe, E.B., Saad, J.S.,
Christie, G.E., Dokland, T., 2011. The Staphylococcus aureus pathogenicity island
1 protein gp6 functions as an internal scaffold during capsid size determina-
Acknowledgments
tion. J. Mol. Biol. 412, 710–722.
Deho, G., Ghisotti, D., 2005. The Satellite Phage P4. In: Calendar, R.L. (Ed.), The
This work was supported by the National Institutes of Health Bacteriophages. Oxford University Press, New York, pp. 391–408.
Deho, G., Zangrossi, S., Ghisotti, D., Sironi, G., 1988. Alternative promoters in the
grants R21 AI067654 and R56 AI081837 to G.E.C.; R21 AI071982 development of bacteriophage plasmid P4. J. Virol. 62, 1697–1704.
and R01 AI083255 to T.D. Deho, G., Zangrossi, S., Sabbattini, P., Sironi, G., Ghisotti, D., 1992. Bacteriophage P4
immunity controlled by small RNAs via transcription termination. Mol.
Microbiol. 6, 3415–3425.
Diana, C., Deho, G., Geisselsoder, J., Tinelli, L., Goldstein, R., 1978. Viral interference
References at the level of capsid size determination by satellite phage P4. J. Mol. Biol. 126,
433–445.
Agarwal, M., Arthur, M., Arbeit, R.D., Goldstein, R., 1990. Regulation of icosahedral Dokland, T., 1999. Scaffolding proteins and their role in viral assembly. Cell Mol.
virion capsid size by the in vivo activity of a cloned gene product. Proc. Natl. Life Sci 56, 580–603.
Acad. Sci. U.S.A 87, 2428–2432. Dokland, T., 2012. gpO peptidase (Enterobacteria phage P2). In: Rawlings, N.D. and
Ahuja, S.K., Murphy, P.M., 1993. Molecular piracy of mammalian interleukin-8 Salvesen, G. (Eds.) Handbook of proteolytic enzymes, 3rd ed. Elsevier, Oxford,
receptor type B by herpesvirus saimiri. J. Biol. Chem. 268, 20691–20694. U.K., ISBN: 9780123822192.
Arnold, H.P., She, Q., Phan, H., Stedman, K., Prangishvili, D., Holz, I., Kristjansson, Dokland, T., Isaksen, M.L., Fuller, S.D., Lindqvist, B.H., 1993. Capsid localization of
J.K., Garrett, R., Zillig, W., 1999. The genetic element pSSVx of the extremely the bacteriophage P4 Psu protein. Virology 194, 682–687.
thermophilic crenarchaeon Sulfolobus is a hybrid between a plasmid and a Dokland, T., Lindqvist, B.H., Fuller, S.D., 1992. Image reconstruction from cryo-
virus. Mol. Microbiol. 34, 217–226. electron micrographs reveals the morphopoietic mechanism in the P2–P4
Ayers, D.J., Sunshine, M.G., Six, E.W., Christie, G.E., 1994. Mutations affecting two bacteriophage system. EMBO J 11, 839–846.
adjacent amino acid residues in the alpha subunit of RNA polymerase block Dokland, T., Wang, S., Lindqvist, B.H., 2002. The structure of P4 procapsids
transcriptional activation by the bacteriophage P2 Ogr protein. J. Bacteriol 176, produced by coexpression of capsid and external scaffolding proteins. Virology
7430–7438. 298, 224–231.
Barrett, K.J., Marsh, M.L., Calendar, R., 1976. Interactions between a satellite Duda, R.L., Martincic, K., Hendrix, R.W., 1995. Genetic basis of bacteriophage HK97
bacteriophage and its helper. J. Mol. Biol. 106, 683–707. prohead assembly. J. Mol. Biol. 247, 636–647.
Bertani, G., 1951. Studies on lysogenesis. I. The mode of phage liberation by Fane, B.A., Prevelige Jr., P.E., 2003. Mechanism of scaffolding-assisted viral
lysogenic Escherichia coli. J. Bacteriol 62, 293–300. assembly. Adv. Protein Chem. 64, 259–299.
Bertani, L.E., Six, E.W., 1988. The P2-like phages and their parasite, P4. In: Feiss, M., Rao, V.B., 2012. The bacteriophage DNA packaging machine. Adv. Exp.
Calendar, R. (Ed.), The Bacteriophages. Plenum Press, New York, pp. 73–143. Med. Biol. 726, 489–509.
Black, L.W., 1989. DNA packaging in dsDNA bacteriophages. Annu. Rev. Microbiol. Ferrer, M.D., Quiles-Puchalt, N., Harwich, M.D., Tormo-Mas, M.A., Campoy, S.,
43, 267–292. Barbe, J., Lasa, I., Novick, R.P., Christie, G.E., Penades, J.R., 2011. RinA controls
Bowden, D.W., Modrich, P., 1985. In vitro maturation of circular bacteriophage P2 phage-mediated packaging and transfer of virulence genes in Gram-positive
DNA. Purification of ter components and characterization of the reaction. bacteria. Nucleic Acids Res. 39, 5866–5878.
J. Biol. Chem 260, 6999–7007. Fitzgerald, J.R., Monday, S.R., Foster, T.J., Bohach, G.A., Hartigan, P.J., Meaney, W.J.,
Breitbart, M., Salamon, P., Andresen, B., Mahaffy, J.M., Segall, A.M., Mead, D., Azam, Smyth, C.J., 2001. Characterization of a putative pathogenicity island from
F., Rohwer, F., 2002. Genomic analysis of uncultured marine viral commu- bovine Staphylococcus aureus encoding multiple superantigens. J. Bacteriol.
nities. Proc. Natl. Acad. Sci. U.S.A 99, 14250–14255. 183, 63–70.
Briani, F., Deho, G., Forti, F., Ghisotti, D., 2001. The plasmid status of satellite Flaitz, C.M., Hicks, M.J., 1998. Molecular piracy: the viral link to carcinogenesis.
bacteriophage P4. Plasmid 45, 1–17. Oral Oncol. 34, 448–453.
Briani, F., Ghisotti, D., Deho, G., 2000. Antisense RNA-dependent transcription Forti, F., Dragoni, I., Briani, F., Deho, G., Ghisotti, D., 2002. Characterization of the
termination sites that modulate lysogenic development of satellite phage P4. small antisense CI RNA that regulates bacteriophage P4 immunity. J. Mol. Biol.
Mol. Microbiol. 36, 1124–1134. 315, 541–549.
Cali, S., Spoldi, E., Piazzolla, D., Dodd, I.B., Forti, F., Deho, G., Ghisotti, D., 2004. Forti, F., Polo, S., Lane, K.B., Six, E.W., Sironi, G., Deho, G., Ghisotti, D., 1999.
Bacteriophage P4 Vis protein is needed for prophage excision. Virology 322, Translation of two nested genes in bacteriophage P4 controls immunity-
82–92. specific transcription termination. J. Bacteriol. 181, 5225–5233.
Catalano, C.E., 2005. Viral Genome Packaging Machines: Genetics, Structure, and Fujimuro, M., Hayward, S.D., Yokosawa, H., 2007. Molecular piracy: manipulation
Mechanism. Landes Bioscience/Eurekah.com and Kluwer Academic/Plenum of the ubiquitin system by Kaposi’s sarcoma-associated herpesvirus. Rev. Med.
Publishers, Georgetown, TX and New York. Virol. 17, 405–422.
Chang, J.R., Poliakov, A., Prevelige, P.E., Mobley, J.A., Dokland, T., 2008. Incorpora- Fujisawa, H., Morita, M., 1997. Phage DNA packaging. Genes Cells. 2, 537–545.
tion of scaffolding protein gpO in bacteriophages P2 and P4. Virology 370, Garcia, E., Chain, P., Elliott, J.M., Bobrov, A.G., Motin, V.L., Kirillina, O., Lao, V.,
352–361. Calendar, R., Filippov, A.A., 2008. Molecular characterization of L-413C, a
Chang, J.R., Spilman, M.S., Rodenburg, C.M., Dokland, T., 2009. Functional domains P2-related plague diagnostic bacteriophage. Virology 372, 85–96.
of the bacteriophage P2 scaffolding protein: identification of residues involved Geisselsoder, J., Youdarian, P., Deho, G., Chidambaram, M., Goldstein, R., Ljung-
in assembly and protease activity. Virology 384, 144–150. quist, E., 1981. Mutants of satellite virus P4 that cannot derepress their
Choi, J., Means, R.E., Damania, B., Jung, J.U., 2001. Molecular piracy of Kaposi’s bacteriophage P2 helper. J. Mol. Biol. 148, 1–19.
sarcoma associated herpesvirus. Cytokine Growth Factor Rev. 12, 245–257. Halling, C., Calendar, R., 1990. Bacteriophage P2 ogr and P4 delta genes act
Christie, G.E., Anders, D.L., McAlister, V., Goodwin, T.S., Julien, B., Calendar, R., independently and are essential for P4 multiplication. J. Bacteriol. 172,
2003. Identification of upstream sequences essential for activation of a 3549–3558.
bacteriophage P2 late promoter. J. Bacteriol. 185, 4609–4614. Harwich, M.D., 2009. Transcriptional Profiling of Staphylococcal Bacteriophage
Christie, G.E., Calendar, R., 1990. Interactions between satellite bacteriophage P4 80a and Regulatory Interactions with Pathogenicity Island SaPI1. Ph.D.
and its helpers. Annu. Rev. Genet. 24 (465-90), 465–490. Dissertation, Virginia Commonwealth University, Richmond, VA.
Christie, G.E., Calendar, R., 1985. Bacteriophage P2 late promoters. II. Comparison Highlander, S.K., Hulten, K.G., Qin, X., Jiang, H., Yerrapragada, S., Mason . Jr, E.O.,
of the four late promoter sequences. J. Mol. Biol. 181, 373–382. Shang, Y., Williams, T.M., Fortunov, R.M., Liu, Y., Igboeli, O., Petrosino, J.,
220 G.E. Christie, T. Dokland / Virology 434 (2012) 210–221

Tirumalai, Y., Uzman, A., Fox, G.E., Cardenas, A.M., Muzny, D.M., Hemphill, L., Pettersen, E.F., Goddard, T.D., Huang, C.C., Couch, G.S., Greenblatt, D.M., Meng, E.C.,
Ding, Y., Dugan, S., Blyth, P.R., Buhay, C.J., Dinh, H.H., Hawes, A.C., Holder, M., Ferrin, T.E., 2004. UCSF Chimera—a visualization system for exploratory
Kovar, C.L., Lee, S.L., Liu, W., Nazareth, L.V., Wang, Q., Zhou, J., Kaplan, S.L., research and analysis. J. Comput. Chem. 25, 1605–1612.
Weinstock, G.M., 2007. Subtle genetic changes enhance virulence of methi- Poliakov, A., Chang, J.R., Spilman, M.S., Damle, P.K., Christie, G.E., Mobley, J.,
cillin resistant and sensitive Staphylococcus aureus. BMC Microbiol 7, 99. Dokland, T., 2008. Capsid size determination by Staphylococcus aureus patho-
Hu, C.C., Hsu, Y.H., Lin, N.S., 2009. Satellite RNAs and Satellite Viruses of Plants. genicity island SaPI1 involves specific incorporation of SaPI1 proteins into
Viruses 1, 1325–1350. procapsids. J. Mol. Biol. 380, 465–475.
Inouye, S., Sunshine, M.G., Six, E.W., Inouye, M., 1991. Retronphage phi R73: an Pountney, D.L., Tiwari, R.P., Egan, J.B., 1997. Metal- and DNA-binding properties
E. coli phage that contains a retroelement and integrates into a tRNA gene. and mutational analysis of the transcription activating factor, B, of coliphage
Science 252, 969–971. 186: a prokaryotic C4 zinc-finger protein. Protein Sci. 6, 892–902.
Isaksen, M.L., Dokland, T., Lindqvist, B.H., 1993. Characterization of the capsid Pruss, G.J., Calendar, R., 1978. Maturation of bacteriophage P2 DNA. Virology 86,
associating activity of bacteriophage P4’s Psu protein. Virology 194, 674–681. 454–467.
Johnson, J.E., 2010. Virus particle maturation: insights into elegantly programmed Pruss, G.J., Wang, J.C., Calendar, R., 1975. In vitro packaging of covalently closed
nanomachines. Curr. Opin. Struct. Biol. 20, 210–216. circular monomers of bacteriophage DNA. J. Mol. Biol. 98, 465–478.
Johnson, J.E., Chiu, W., 2007. DNA packaging and delivery machines in tailed Ram, G., Chen, J., Kumar, K., Ross, H.F., Ubeda, C., Damle, P.K., Lane, K.D., Penades,
bacteriophages. Curr. Opin. Struct. Biol. 17, 237–243. J.R., Christie, G.E., Novick, R.P., 2012. SaPI interference with helper phage
Julien, B., Calendar, R., 1996. Bacteriophage PSP3 and phiR73 activator proteins: reproduction is a paradigm of molecular parasitism. Proc. Natl. Acad. Sci. U.S.A
analysis of promoter specificities. J. Bacteriol. 178, 5668–5675. 109, 16300–16305.
Julien, B., Pountney, D., Christie, G.E., Calendar, R., 1998. Mutational analysis of a Rosenberg, D., 2009. The political economy of piracy in the South China Sea. Naval
satellite phage activator. Gene 223, 129–134. War Coll. Rev. 62, 43–58.
Kalionis, B., Dodd, I.B., Egan, J.B., 1986. Control of gene expression in the P2-related Roy, A., Bhardwaj, A., Datta, P., Lander, G.C., Cingolani, G., 2012. Small terminase
template coliphages. III. DNA sequence of the major control region of phage couples viral DNA binding to genome-packaging ATPase activity. Structure 20,
186. J. Mol. Biol. 191, 199–209. 1403–1413.
Kim, K.J., Sunshine, M.G., Lindqvist, B.H., Six, E.W., 2001. Capsid size determination Ruzin, A., Lindsay, J., Novick, R.P., 2001. Molecular genetics of SaPI1—a mobile
in the P2-P4 bacteriophage system: suppression of sir mutations in P2’s capsid pathogenicity island in Staphylococcus aureus. Mol. Microbiol. 41, 365–377.
gene N by supersid mutations in P4’s external scaffold gene sid. Virology 283, Sabbattini, P., Forti, F., Ghisotti, D., Deho, G., 1995. Control of transcription
49–58. termination by an RNA factor in bacteriophage P4 immunity: identification
Koonin, E., Wolf, Y., 2008. Genomics of bacteria and archaea: the emerging of the target sites. J. Bacteriol. 177, 1425–1434.
dynamic view of the prokaryotic world. Nucleic Acids Res. 36, 6688–6719. Saha, S., Haggard-Ljungquist, E., Nordstrom, K., 1989. Activation of prophage P4 by
Kuroda, M., Yamashita, A., Hirakawa, H., Kumano, M., Morikawa, K., Higashide, M., the P2 Cox protein and the sites of action of the Cox protein on the two phage
Maruyama, A., Inose, Y., Matoba, K., Toh, H., Kuhara, S., Hattori, M., Ohta, T., genomes. Proc. Natl. Acad. Sci. U.S.A 86, 3973–3977.
2005. Whole genome sequence of Staphylococcus saprophyticus reveals the Saha, S., Haggard-Ljungquist, E., Nordstrom, K., 1987. The cox protein of bacter-
pathogenesis of uncomplicated urinary tract infection. Proc. Natl. Acad. Sci. iophage P2 inhibits the formation of the repressor protein and autoregulates
U.S.A 102, 13272–13277. the early operon. EMBO J. 6, 3191–3199.
Kwan, T., Liu, J., DuBow, M., Gros, P., Pelletier, J., 2005. The complete genomes and Sauer, B., Calendar, R., Ljungquist, E., Six, E., Sunshine, M.G., 1982. Interaction of
proteomes of 27 Staphylococcus aureus bacteriophages. Proc. Natl. Acad. Sci. satellite phage P4 with phage 186 helper. Virology 116, 523–534.
U.S.A 102, 5174–5179. Sauer, B., Ow, D., Ling, L., Calendar, R., 1981. Mutants of satellite bacteriophage P4
that are defective in the suppression of transcriptional polarity. J. Mol. Biol.
La Scola, B., Desnues, C., Pagnier, I., Robert, C., Barrassi, L., Fournous, G., Merchat,
145, 29–46.
M., Suzan-Monti, M., Forterre, P., Koonin, E., Raoult, D., 2008. The virophage as
Schwudke, D., Ergin, A., Michael, K., Volkmar, S., Appel, B., Knabner, D., Konietzny, A.,
a unique parasite of the giant mimivirus. Nature 455, 100–104.
Strauch, E., 2008. Broad-host-range Yersinia phage PY100: genome sequence,
Lin, C.S., 1984. Nucleotide sequence of the essential region of bacteriophage P4.
proteome analysis of virions, and DNA packaging strategy. J. Bacteriol. 190,
Nucleic Acids Res. 12, 8667–8684.
332–342.
Lindqvist, B.H., Deho, G., Calendar, R., 1993. Mechanisms of genome propagation
Scott, J., Nguyen, S.V., King, C.J., Hendrickson, C., McShan, W.M., 2012. Phage-like
and helper exploitation by satellite phage P4. Microbiol. Rev. 57, 683–702.
streptococcus pyogenes chromosomal islands (SpyCI) and mutator phenotypes:
Lindsay, J.A., Ruzin, A., Ross, H.F., Kurepina, N., Novick, R.P., 1998. The gene for
control by growth state and rescue by a SpyCI-encoded promoter. Front.
toxic shock toxin is carried by a family of mobile pathogenicity islands in
Microbiol 3, 317.
Staphylococcus aureus. Mol. Microbiol. 29, 527–543.
Shore, D., Deho, G., Tsipis, J., Goldstein, R., 1978. Determination of capsid size by
Liu, J., Dehbi, M., Moeck, G., Arhin, F., Bauda, P., Bergeron, D., Callejo, M., Ferretti, V.,
satellite bacteriophage P4. Proc. Natl. Acad. Sci. U.S.A 75, 400–404.
Ha, N., Kwan, T., McCarty, J., Srikumar, R., Williams, D., Wu, J.J., Gros, P.,
Sinkovics, J., Horvath, J., Horak, A., 1998. The origin and evolution of viruses
Pelletier, J., DuBow, M., 2004. Antimicrobial drug discovery through bacter-
(a review). Acta Microbiol. Immunol. Hung. 45, 349–390.
iophage genomics. Nat. Biotechnol. 22, 185–191.
Six, E.W., 1975. The helper dependence of satellite bacteriophage P4: which gene
Liu, T., Renberg, S.K., Haggard-Ljungquist, E., 1998. The E protein of satellite phage functions of bacteriophage P2 are needed by P4? Virology 67, 249–263.
P4 acts as an anti-repressor by binding to the C protein of helper phage P2.
Six, E.W., 1963. A defective phage depending on phage P2. Bacteriol. Proc. 1963,
Mol. Microbiol. 30, 1041–1050.
138.
Liu, T., Renberg, S.K., Haggard-Ljungquist, E., 1997. Derepression of prophage P2 by Six, E.W., Klug, C.A., 1973. Bacteriophage P4: a satellite virus depending on a
satellite phage P4: cloning of the P4 epsilon gene and identification of its helper such as prophage P2. Virology 51, 327–344.
product. J.Virol 71, 4502–4508. Six, E.W., Lindqvist, B.H., 1978. Mutual derepression in the P2–P4 bacteriophage
Lynch, K.H., Stothard, P., Dennis, J.J., 2010. Genomic analysis and relatedness of system. Virology 87, 217–230.
P2-like phages of the Burkholderia cepacia complex. BMC Genomics 11, 599. Six, E.W., Sunshine, M.G., Williams, J., Haggard-Ljungquist, E., Lindqvist, B.H., 1991.
Marvik, O.J., Dokland, T., Nokling, R.H., Jacobsen, E., Larsen, T., Lindqvist, B.H., 1995. Morphopoietic switch mutations of bacteriophage P2. Virology 182, 34–46.
The capsid size-determining protein Sid forms an external scaffold on phage Spilman, M.S., Damle, P.K., Dearborn, A.D., Rodenburg, C.M., Chang, J.R., Wall, E.A.,
P4 procapsids. J. Mol. Biol. 251, 59–75. Christie, G.E., Dokland, T. Assembly of bacteriophage 80alpha capsids in a
Maxwell, K.L., Frappier, L., 2007. Viral proteomics. Microbiol. Mol. Biol. Rev. 71, Staphylococcus aureus expression system. Virology, http://dx.doi.org/10.1016/j.
398–411. virol.2012.08.031, in press.
McAlister, V., Zou, C., Winslow, R.H., Christie, G.E., 2003. Purification and in vitro Spilman, M.S., Dearborn, A.D., Chang, J.R., Damle, P.K., Christie, G.E., Dokland, T.,
characterization of the Serratia marcescens NucC protein, a zinc-binding 2011. A conformational switch involved in maturation of Staphylococcus
transcription factor homologous to P2 Ogr. J. Bacteriol. 185, 1808–1816. aureus bacteriophage 80alpha capsids. J. Mol. Biol. 405, 863–876.
Morais, M.C., Kanamaru, S., Badasso, M.O., Koti, J.S., Owen, B.A., McMurray, C.T., Sunshine, M.G., Sauer, B., 1975. A bacterial mutation blocking P2 phage late gene
Anderson, D.L., Rossmann, M.G., 2003. Bacteriophage phi29 scaffolding protein expression. Proc. Natl. Acad. Sci. U.S.A 72, 2770–2774.
gp7 before and after prohead assembly. Nat. Struct. Biol. 10, 572–576. Sureau, C., 2006. The role of the HBV envelope proteins in the HDV replication
Nilssen, O., Fossdal, C.G., Johansen, B.V., Lindqvist, B.H., 1996. Bacteriophage P4 cycle. Curr. Top. Microbiol. Immunol. 307, 113–131.
capsid-size determination and its relationship to P2 helper interference. Takeuchi, F., Watanabe, S., Baba, T., Yuzawa, H., Ito, T., Morimoto, Y., Kuroda, M.,
Virology 219, 443–452. Cui, L., Takahashi, M., Ankai, A., Baba, S., Fukui, S., Lee, J.C., Hiramatsu, K., 2005.
Nilsson, A.S., Haggard Ljungquist, E., 2005. The P2-Like Bacteriophages. In: Whole-genome sequencing of Staphylococcus haemolyticus uncovers the
Calendar, R.L. (Ed.), The Bacteriophages. Oxford University Press, New York, extreme plasticity of its genome and the evolution of human-colonizing
pp. 365–390. staphylococcal species. J. Bacteriol. 187, 7292–7308.
Nilsson, A.S., Karlsson, J.L., Haggard-Ljungquist, E., 2004. Site-specific recombina- Tallent, S.M., Langston, T.B., Moran, R.G., Christie, G.E., 2007. Transducing particles
tion links the evolution of P2-like coliphages and pathogenic enterobacteria. of Staphylococcus aureus pathogenicity island SaPI1 are comprised of helper
Mol. Biol. Evol. 21, 1–13. phage-encoded proteins. J. Bacteriol. 189, 7520–7524.
Novick, R.P., Christie, G.E., Penades, J.R., 2010. The phage-related chromosomal Teschke, C.M., 2012. Themes and variations of viral small terminase proteins.
islands of Gram-positive bacteria. Nat. Rev. Microbiol. 8, 541–551. Structure 20, 1291–1292.
Ott, D.E., 2008. Cellular proteins detected in HIV-1. Rev. Med. Virol. 18, 159–175. Tormo, M.A., Ferrer, M.D., Maiques, E., Ubeda, C., Selva, L., Lasa, I., Calvete, J.J.,
Pani, B., Ranjan, A., Sen, R., 2009. Interaction surface of bacteriophage P4 protein Novick, R.P., Penades, J.R., 2008. Staphylococcus aureus pathogenicity island
Psu required for complex formation with the transcription terminator Rho. DNA is packaged in particles composed of phage proteins. J. Bacteriol. 190,
J. Mol. Biol. 389, 647–660. 2434–2440.
G.E. Christie, T. Dokland / Virology 434 (2012) 210–221 221

Tormo-Mas, M.A., Mir, I., Shrestha, A., Tallent, S.M., Campoy, S., Lasa, I., Barbe, J., of integration into the host genome and spreading in the presence of a
Novick, R.P., Christie, G.E., Penades, J.R., 2010. Moonlighting bacteriophage fusellovirus. Virology 363, 124–133.
proteins derepress staphylococcal pathogenicity islands. Nature 465, 779–782. Wikoff, W.R., Liljas, L., Duda, R.L., Tsuruta, H., Hendrix, R.W., Johnson, J.E., 2000.
Toussaint, A, Chandler, M., 2012. Prokaryote genome fluidity: toward a system Topologically linked protein rings in the bacteriophage HK97 capsid. Science
approach of the mobilome. Methods Mol. Biol. 804, 57–80. 289, 2129–2133.
Ubeda, C., Maiques, E., Barry, P., Matthews, A., Tormo, M.A., Lasa, I., Novick, R.P., Wood, L.F., Tszine, N.Y., Christie, G.E., 1997. Activation of P2 late transcription by
Penades, J.R., 2008. SaPI mutations affecting replication and transfer and P2 Ogr protein requires a discrete contact site on the C terminus of the alpha
enabling autonomous replication in the absence of helper phage. Mol. subunit of Escherichia coli RNA polymerase. J. Mol. Biol. 274, 1–7.
Microbiol. 67, 493–503.
Wu, H., Sampson, L., Parr, R., Casjens, S., 2002. The DNA site utilized by
Ubeda, C., Maiques, E., Tormo, M., Campoy, S., Lasa, I., Barbe, J., Novick, R.P.,
bacteriophage P22 for initiation of DNA packaging. Mol. Microbiol. 45,
Penades, J.R., 2007. SaPI operon I is required for SaPI packaging and is
1631–1646.
controlled by LexA. Mol. Microbiol. 65, 41–50.
Ubeda, C., Olivarez, N.P., Barry, P., Wang, H., Kong, X., Matthews, A., Tallent, S.M., Yan, X., Sinkovits, R., Baker, T., 2007. AUTO3DEM—an automated and high through-
Christie, G.E., Novick, R.P., 2009. Specificity of staphylococcal phage and SaPI put program for image reconstruction of icosahedral particles. J. Struct. Biol. 157,
DNA packaging as revealed by integrase and terminase mutations. Mol. 73–82.
Microbiol. 72, 98–108. Yu, A., Haggard-Ljungquist, E., 1993. The Cox protein is a modulator of direction-
Wang, S., Chang, J.R., Dokland, T., 2006. Assembly of bacteriophage P2 and P4 ality in bacteriophage P2 site-specific recombination. J. Bacteriol. 175,
procapsids with internal scaffolding protein. Virology 348, 133–140. 7848–7855.
Wang, Y., Duan, Z., Zhu, H., Guo, X., Wang, Z., Zhou, J., She, Q., Huang, L., 2007. A Ziermann, R., Calendar, R., 1990. Characterization of the cos sites of bacteriophages
novel Sulfolobus non-conjugative extrachromosomal genetic element capable P2 and P4. Gene 96, 9–15.
CHAPTER TEN

THE FORCES BEHIND THE DISSEMINATION


OF BACTERIAL VIRULENCE
AND ANTIBIOTIC RESISTANCE

GABRIEL TRUEBA

It is intriguing that a large number of genes coding for virulence and antibiotic resistance are associated with
genes involved in intra-genomic mobility and horizontal gene transfer. In other cases, similar gene
associations have been observed to carry genes coding for functions such as photosynthesis, plant symbiosis,
degradation different substrates including pollutants, etc. A great variety of these structures, called mobile
genetic elements (MGEs), are widely disseminated in nature, and I will present the possible scenarios and
events in which natural selection may favour the genesis and propagation of these genetic structures.

Introduction
The horizontal transference of Mobile Genetic Elements (MGEs) is a major factor contributing to the
emergence of bacterial pathogens and antibiotic resistant bacteria (Ho Sui et al, 2009; Baquero, 2008).
Examples include enterohemorrhagic E. coli O104:H4 (Rasko et al, 2011), Cronobacter sakasaki (Kucerova,
2010) and highly virulent-methicillin resistant strains of Staphylococcus aureus (Diep, 2006). In addition to
clinically relevant examples, MGEs are also implicated in other important functions such as photosynthetic
carbon fixation in the oceans (Rohwer and Thurber, 2009), bacteria-plant symbiosis (Dobrindt, 2004), and
degradation of pollutants (Top, 2003).

MGEs are genetic structures that may contain both genes conferring selective advantages to the bacterial cells
and genes involved in horizontal gene transfer (hereinafter referred to as inter-genomic mobility) (Dobrindt,
2004; Jackson 2011), and translocation or transposition (hereinafter referred to as intra-genomic mobility).
Horizontal transference of MGEs is experimentally observable, in other cases it is assumed because of
molecular discrepancies (GC content, molecular phylogeny and codon usage) found when MGE and
housekeeping genes are compared. Additional evidences are: the existence of ancestral versions of MGEs in
distantly related bacteria (Gillings, 2008; Rowe-Magnus, 2001); the presence of remnants of mobility genes in
some MGEs, and irregular distribution of MGEs in members of the same bacterial species (Diep, 2006).

MGEs contain core and cargo regions: MGE core regions have genes coding for intra-genomic gene mobility
(integration, transposition) and intercellular transfer (conjugation and viral packaging); the cargo or moron
region contains genes coding diverse bacterial adaptive functions (Seth-Smith, 2009). MGEs could be
grouped according to the nature of the core and cargo regions. Core regions containing integrases and
relaxosomes (conjugation machinery) genes are named integrative conjugal elements (ICEs), those that have a
relaxosome and an origin of replication are known as plasmids, the ones containing phage-like integrases and
integration sites are known as integrons, and finally those containing many phage (viral) genes are
converting-phages (Rankin, 2011). Similarly, some MGEs are classified according to the cargo region in:
pathogenicity islands, metabolic islands, symbiotic islands, antibiotic resistance islands, etc.

The evolution of MGEs is complex and it involves the association of genes from different origins. The core
genes likely derive from selfish genetic elements (such as plasmids, phages and transposons) whose success
often depends on their aggressive replication without necessarily benefiting the bacterial host (Rankin, 2011;
Eberhard, 1990; Werren, 2011; Doolittle, 1980; Dionisio, 2005). Integrases in MGEs are genetically related to
phage tyrosine integrases; however, some MGE integrases form a separate phylogenetic cluster, which may
indicate that they emerged from an ancient phage-like structure (Napolitano, 2011). Relaxosome genes in
plasmid and ICEs are genetically related, however it is not possible to establish which structure is more
ancestral (Guglielmini, 2011). The origins of some virulence and antibiotic resistance genes in MGEs have
been linked to environmental bacteria and are reviewed extensively in other manuscripts (Martinez, 2012;
Wright, 2012).

Selfish genetic elements may prosper at the expense of the bacterial fitness; in addition, transposons and
phages may impose a cost on their host by disrupting the coding sequences of essential genes (Doolittle and
Sapienza 1980). Conversely, cargo genes alone could provide competitive advantage (in specific niches) to
the bacterial cell (Eberhard, 1990; Werren, 2011) while the combination of both (cargo and core genes) may
result in successful alliances which disseminate easily in nature (Werren, 2011).

The structural characteristics of MGEs (abundance of ISs and other recombinases) may provide evidence of
their possible origins, including: 1) gene capture; 2) integration into genetic platforms capable of moving
from cell to cell (plasmids or phages); 3) MGE gene (moron) exchange; and 4) integration into bacterial
chromosome and loss of unnecessary genes (Fig. 1). The present review summarizes current views and
presents the similarities in the origin of diverse MGEs.

Gene capture
Gene capture involves the joining of a DNA fragment coding for a meaningful function to a recombinase
gene, which could be transposase, tyrosine recombinase, or serine recombinase. Transposase is the most
abundant gene in nature (Aziz et al, 2010) and insertion sequences (ISs) may be the most common
transposable element in bacteria. Composite transposons arise when two ISs insert on the flanks of a gene,
leading to a region containing the two ISs elements with an intervening DNA fragment (Baquero, 2008;
Jackson et al, 2011; Iida, et al; Mahillon, 1998); in this way a gene could gain the ability to move from one
place to another within the genome (Figure 1). Similar events can take place when a gene becomes associated
with phage recombinases during specialized transduction (Jackson et al 2011; Gillings et al 2008; Wagner and
Waldor, 2002) or other illegitimate recombination events involving viral or plasmid integrases. If the
composite transposon or integrative gene inserts into a higher copy-number replicon (such as a plasmids) it
could increase its burden on bacterial metabolism. However if the captured gene codes for an adaptive feature
this event could improve the odds for the bacterial success (due to higher expression of the gene) and could
increase the abundance of this gene association in nature. Adaptive genes are very common in environments
wherever there is a selective pressure over a bacterial population (such as the presence of antibiotics).
Selection results in large numbers of bacteria carrying these genes which increases the chances for the
transposon gene capture.

Gene integration into platforms capable


of cell to cell transference
Insertion of the captured gene (composite transposon or integrative gene) into a conjugal plasmid or into a
viral genome would create a MGE able to mobilize horizontally between bacterial cells; those bacteria
benefiting from cargo genes will prosper, and so will these MGEs (Eberhard 1990; Dionisio et al, 2005).
There are many examples of such combinations that allow a pathogen to emerge and occupy a unique
ecological niche such as Vibrio cholerae, Bacillus anthracis, Yersinia pestis, etc (Hacker and Kaper, 2000). A
gene coding for a meaningless function, or a non-coding DNA, can be also associated with recombinases and
integrated into mobile platforms; however, in the absence of adaptive characteristics, these associations are
probably destined for extinction (Rowe-Magnus, 2001). This phenomenon may explain the abundance of
MGEs carrying genes that code for significant functions. Horizontal transference of MGEs into different
bacterial backgrounds (genomes) may result in gene combinations that offer compatible characteristics, for
instance MGEs coding for a virulence features may prosper in pathogenic bacteria, but not in non-pathogenic
bacteria.
MGE gene (moron) exchange
When more than one selective pressure has been maintained for long time over a bacterial population, there
may be a large number of bacterial cells carrying different MGEs. The existence of two or more MGEs inside
bacterial cells sets the stage for the next event, which is the exchange of adaptive genes (modules) between
MGEs (Baquero, 2008) and the formation of more complex MGEs such as integrons (Baquero, 2008; Rowe-
Magnus et al, 2002 ).

MGE Integration into bacterial chromosomes


and gene loss
Inactivating mutations in core genes (integrases, relaxosome genes, transposases, replicons, viral structural
genes, genes involved in bacterial lysis, etc.) could affect MGE fitness by reducing its ability to multiply or
move (Hacker and Kaper, 2000). Similarly, mutations in beneficial cargo genes could push the bacterial hosts
to extinction.

In some cases, the cargo genes provide such valuable benefits to the host that the loss of mobility genes does
not seem to diminish the success in chromosomally anchored MGEs (Eberhard, 1990; Wang et al, 2010;
Osborn and Böltner, 2002). Some anchored MGEs still contain mobility-associated genes and exhibit lateral
transference when the cell is infected with phages or plasmids capable of complementing this function
(Jackson et al, 2011; Napolitano Napolitano et al, 2011; Guglielmini et al, 2011; Wang et al, 2010). In other
cases there seems to be a pressure in MGEs to maintain the core genes: for instance, photosynthetic genes are
very common in phages infecting most abundant photosynthetic bacteria in oceans (Rohwer and Thurber,
2009), however these genes didn’t seem to be associated with anchored MGEs (Coleman et al, 2006). In this
case, viral genes may allow MGEs to disseminate in environments with low bacterial concentration such as
oceans (Whitman et al, 1998). Unlike conjugation, viral dissemination does not require direct cell-to-cell
contact; in this environment, phages could move genes long distances searching for new bacterial hosts.

Conclusions
The synergy of the cargo-core gene association could explain the evolutionary abundance of MGEs
(Guglielmini et al, 2011; Aziz et al, 2010); and the more successful, the more abundant. But it is important to
understand the environmental context that allows the survival and dissemination of MGEs. Those carrying
metabolic, virulence or antibiotic resistance genes, for example, could disseminate very fast among different
bacterial species in the presence of selective pressure (Peters et al, 1997). Hence, a cargo gene that is
advantageous under specific environmental conditions may be detrimental if the bacterial population expands
beyond this niche (Eberhard, 1990)

From the infectious disease point of view it is important to understand the environmental factors involved in
the persistence and dissemination of MGEs carrying virulence or antibiotic resistance genes. Environmental
stressors, such as antibiotic exposure, not only select for bacteria carrying antibiotic resistance genes, but
could also stimulate intra-genomic and inter-genomic mobility of MGEs carrying resistance genes
(McGannon et al, 2010; Guerin et al, 2009; Prudhomme et al, 2006; Beaber et al, 2004). A recent report
shows that intestines of antibiotic treated mice have far more MGEs carrying antibiotic resistance genes than
non-treated counterparts (Modi et al, 2012).

The understanding of the interactions between antibiotics and antibiotic resistance (and virulence) genes
should inform professionals about the risks of using sub-therapeutic doses of antibiotics as growth promoters
in food animals (Silbergeld et al, 2008). Finally this knowledge may lead to the development of new
approaches (such as eco-evo drugs) to control antibiotic resistance and the emergence of new bacterial
pathogens (Baquero et al, 2011).

Acknowledgments
The author thanks Paul Keim and Ana Trueba for their valuable suggestions.
References
Aziz, Ramy, Mya Breitbart, and Robert Edwards. “Transposases are the most abundant, most ubiquitous
genes in nature.” Nucleic Acids Research 38 (2010): 4207-4217.
Baquero, Fernado, Teresa Coque, and Fernando-de-la-Cruz F. “Ecology and evolution as targets: the need for
novel eco-evo drugs and strategies to fight antibiotic resistance.” Antimicrobial Agents Chemotherapy 55
(2011): 3649-3660.
Baquero, Fernando. “Modularization and Evolvability in Antibiotic Resistance.” In Evolutionary biology of
bacterial and fungal pathogens, 233-247. Washington: ASM Press, 2008.
Beaber, John, Blanca Hochhut, and Mathew Waldor. “SOS Response Promotes Horizontal Dissemination of
Antibiotic Resistance Genes.” Nature 427 (2004): 72-74.
Coleman, Maureen, Matthew Sullivan, Adam Martiny, Steglich Claudia, Kerrie Barry, Edward Delong, and
Sallie Chisholm. “Genomic Islands and the Ecology and Evolution of Prochlorococcus.” Science 311
(2006): 1768-1770.
Diep, Binh, Heather Carleton, Richard Chang, George Sensabaugh, and Françoise Perdreau-Remington.
“Roles of 34 Virulence Genes in the Evolution of Hospital- and Community-Associated Strains of
Methicillin-Resistant Staphylococcus aureus” The Journal of Infectious Diseases 193 (2006): 1495-1503.
Dionisio, F., I Conceição, A Marques, L Fernandes, and I Gordo, “The Evolution of a Conjugative Plasmid
and its Ability to Increase Bacterial Fitness.” Biology Letters 1 (2005): 250-252.
Dobrindt, Ulrich, Bianca Hochhut, Ute Hentschel, Jörg Hacker. “Genomic Islands in Pathogenic and
Environmental Microorganisms.” Nature Reviews Microbiology 2 (2004): 414-424.
Doolittle, W. Ford, and Carme Sapienza. “Selfish Genes, the Phenotype Paradigm and Genome Evolution.”
Nature 284 (1980): 601-603.
Eberhard, William. “Evolution in Bacterial Plasmids and Levels of Selection.” The Quarterly Review of
Biology 65 (1990): 3-22.
Gillings, Michael, Yan Boucher, Maurizio Labbate, Andrew Holmes, Samyuktha Krishnan, Marita Holley,
and H.W. Stokes. “The Evolution of Class 1 Integrons and the Rise of Antibiotic Resistance.” Journal of
Bacteriology 190 (2008): 5095-5100.
Guerin, Emilie, Guillaume Cambray, Neus Sanchez-Alberola, Susana Campoy, Ivan Erill, Sandra Da-Re,
Bruno Gonzalez-Zorn, Jordi Barbé, Marie-Cécile Ploy, and Didier Mazel. “The SOS Response Controls
Integron Recombination.” Science 324 (2009): 1034.
Guglielmini, Julien, Leonor Quintais, Maria-Pilar Garcillán-Barcia, Fernando de-la-Cruz, and Eduardo
Rocha. “The Repertoire of ICE in Prokaryotes Underscores the Unity, Diversity, and Ubiquity of
Conjugation” PLoS Genetics 7 (2011): e1002222.
Hacker, Jörg, and James Kaper, “Pathogenicity Islands and the Evolution of Microbes.” Annual Reviews of
Microbiology 54 (2000): 641-679.
Ho Sui, Shannan, Amber Fedynak, William Hsiao, Morgan Langille, and Fiona Brinkman. “The Association
of Virulence Factors with Genomic Islands.” PLoS One 4 (2009): e8094.
Iida, S, J. Meyer, and W. Arber. “Genesis and Natural History of IS-Mediated Transposons.” Cold Spring
Harbor Symposia of Quantitative Biology 45 (1980): 27-37.
Jackson, Robert, Boris Vinatzer, Dawn Arnold, Steve Dorus, and Jesús Murillo. “The Influence of the
Accessory Genome on Bacterial Pathogen Evolution.” Mobile Genetic Elements 1 (2011): 55-65.
Kucerova, Eva, Sandra Clifton, Xiao-Qin Xia, Fred Long, Steffen Porwollik, Lucinda Fulton, Catrina
Fronick, Patrick Minx, Kim Kyung, Wesley Warren, Robert Fulton, Dongyan Feng, Aye Wollam, Neha
Shah, Veena Bhonagiri, William Nash, Kymberlie Hallsworth-Pepin, Richard Wilson, Michael
McClelland, and Stephen Forsythe. “Genome Sequence of Cronobacter sakazakii BAA-894 and
Comparative Genomic Hybridization Analysis with other Cronobacter species.” PLoS One 5 (2010):
e9556.
Mahillon, J. “Transposons as Gene Haulers.” Acta Pathologica, Microbiologica et Immunologica
Scandinavica Suppl 84 (1998): 29-36.
Martínez, José. “Bacterial Pathogens: From Natural Ecosystems to Human Hosts.” Environmental
Microbiology 15 (2012): 325–333.
McGannon, Colleen, Cynthia Fuller, and Alison Weiss. “Different Classes Of Antibiotics Differentially
Influence Shiga Toxin Production.” Antimicrobrial Agents and Chemotherapy 54 (2010): 3790-3798.
Modi, Sheetal, Henry Lee, Catherine Spina, and James Collins. “Antibiotic Treatment Expands The
Resistance Reservoir And Ecological Network Of The Phage Metagenome.” Nature 499 (2013): 219-222.
Napolitano, Michael, Salvador Almagro-Moreno, Fidelma Boyd. “Dichotomy In The Evolution Of
Pathogenicity Island And Bacteriophage Encoded Integrases From Pathogenic Escherichia Coli Strains.”
Infection, Genetics and Evolution 11 (2011): 423-436.
Osborn, Mark, and Dietmar Böltner. “When Phage, Plasmids, And Transposons Collide: Genomic Islands,
And Conjugative- And Mobilizable-Transposons As A Mosaic Continuum.” Plasmid 48 (2002): 202-212.
Peters, M, E. Heinaru, E. Talpsep, H. Wand, U. Stottmeister, A. Heinaru, and A. Nurk. “Acquisition Of A
Deliberately Introduced Phenol Degradation Operon, Pheba, By Different Indigenous Pseudomonas
Species.” Applied and Environmental Microbiology 63 (1997): 4899-4906.
Prudhomme, Marc, Laetitia Attaiech, Guillaume Sanchez, Bernard Martin, and Jean-Pierre Claverys.
“Antibiotic Stress Induces Genetic Transformability In The Human Pathogen Streptococcus Pneumoniae.”
Science 313 (2006): 89-92.
Rankin, D., E. Rocha, and S. Brown, “What traits are carried on mobile genetic elements, and why?”
Heredity106 (2011): 1-10.
Rasko, David, Dale Webster, Jason Sahl, Ali Bashir, Nadia Boisen, Flemming Scheutz, Ellen Paxinos, Robert
Sebra, Chen-Shan Chin, Dimitris Iliopoulos, Aaron Klammer, Paul Peluso, Lawrence Lee, Andrey
Kislyuk, James Bullard, Andrew Kasarskis, Susanna Wang, John Eid, David Rank, Julia Redman, Susan
Steyert, Jakob Frimodt-Møller, Carsten Struve, Andreas Petersen, Karen Krogfelt, James Nataro, Eric
Schadt, and Matthew Waldor. “Origins Of The E. Coli Strain Causing An Outbreak Of Hemolytic-Uremic
Syndrome In Germany.” New England Journal of Medicine 365 (2011): 709-717.
Rohwer, Forest, and Rebbeca Vega Thurber, “Viruses manipulate the marine environment.” Nature 459
(2009): 207-212.
Rowe-Magnus, Dean, Anne-Marie Guerout, and Didier Mazel. “Bacterial Resistance Evolution By
Recruitment Of Super-Integron Gene Cassettes.” Molecular Microbiology 43 (2002): 1657-1669.
Rowe-Magnus, Dean, Anne-Marie Guerout, Pascaline Ploncard, broderick Dychinco, Julian Davies, and
Didier Mazel. “The evolutionary history of chromosomal super-integrons provides an ancestry for
multiresistant integrons.” Proceedings of the National Academy of Sciences of the United States of
America 98 (2001): 652-657.
Seth-Smith, Helena, and Nicholas Crouche. “Genome watch: breaking the ICE.” Nature Reviews
Microbiology 7 (2009): 328-329.
Silbergeld, Ellen, Jay Graham, and Lance Price. “Industrial food animal production, antimicrobial resistance,
and human health.” Annual Review of Public Health 29 (2008):151-69.
Top, Eva, and Dirk Springael. “The role of mobile genetic elements in bacterial adaptation to xenobiotic
organic compounds” Current Opinion in Biotechnolology 14 (2003): 262-269.
Wagner, Patrick, and Matthew Waldor. “Bacteriophage control of bacterial virulence.” Infection and
Immunity 70 (2002): 3985-3993.
Wang, Xiaoxue, Younghoon Kim, Qun Ma, Seok Hong, Karina Pokusaeva, Joseph Sturino, and Thomas
Wood. “Cryptic Prophages Help Bacteria Cope With Adverse Environments.” Nature Communications 1
(2010): 147.
Werren, JH. “Selfish Genetic Elements, Genetic Conflict, And Evolutionary Innovation.” Proceedings of the
National Academy of Sciences of the United States of America 108 Suppl 2 (2011): 10863-10870.
Whitman, William, David Coleman, and William Wiebe. “Prokaryotes: The Unseen Majority.” Proceedings
of the National Academy of Sciences of the United States of America 95 (1998): 6578-6583.
Wright, Gerard. “The origins of antibiotic resistance.” Handbook of Experimental Pharmacology 211 (2012):
13-30.
Figure 1. Steps in the evolution of mobile genetic elements (MGEs). Two ISs insert and capture a chromosomal gene, 1;
Composite transposons integrate into genetic platforms capable of moving from cell to cell (plasmids or phages), 2;
Successful MGEs exchange genes coding for adaptive functions, 3; MGEs with multiple morons are formed 4; Mobile
MGEs integrate into bacterial chromosome and lose unnecessary genes (chromosomally
Plasmid 48 (2002) 202–212
www.academicpress.com

When phage, plasmids, and transposons


collide: genomic islands, and conjugative- and
mobilizable-transposons as a mosaic continuum
A. Mark Osborn* and Dietmar Bo
€ltner
Department of Biological Sciences, University of Essex, Colchester, CO4 3SQ, UK
Received 29 August 2002

Abstract

Plasmids and bacteriophage represent the classical vectors for gene transfer within the horizontal gene
pool. However, the more recent discovery of an increasing array of other mobile genetic elements (MGE)
including genomic islands (GIs), conjugative transposons (CTns), and mobilizable transposons (MTns)
which each integrate within the chromosome, offer an increasingly diverse assemblage contributing to
bacterial adaptation and evolution. Molecular characterisation of these elements has revealed that they are
comprised of functional modules derived from phage, plasmids, and transposons, and further that these
modules are combined to generate a continuum of mosaic MGE. In particular, they are comprised of any
one of three distinct types of recombinase, together with plasmid-derived transfer and mobilisation gene
functions. This review highlights both the similarities and distinctions between these integrating trans-
ferable elements resulting from combination of the MGE toolbox.
Ó 2002 Elsevier Science (USA). All rights reserved.

Keywords: Gene transfer; Genomic islands; Conjugative transposons; Conjugation; Tyrosine recombinase; Resolvase;
Transposase

1. Mobile genetic elements and the horizontal gene Salmonella bacteriophage (Zinder and Lederberg,
pool 1952) and the introduction of the term plasmid
(Lederberg, 1952), following the earlier identifi-
The prokaryotic horizontal gene pool (HGP) cation of bacterial conjugation, research has re-
represents a rich tapestry of adaptive phenotypes vealed numerous combinations of genetic modules
conveyed within and between bacterial (and ar- derived from phage, plasmid (and transposons) to
chaeal) cells, by an increasingly diverse assem- generate an array of MGE that include conjuga-
blage of mobile genetic elements (MGE). Fifty tive and mobilizable transposons, genomic is-
years on from the discovery of transduction by lands, and integrons (Toussaint and Merlin,
2002).
Whilst plasmids and bacteriophage have well-
*
Corresponding author. Fax: +44-1206-872592. established genetic and phenotypic identities, the
E-mail address: osborn@essex.ac.uk (A. Mark Os- emerging families of mosaic MGE pose a con-
born). siderable challenge for the systematicist as to how

0147-619X/02/$ - see front matter Ó 2002 Elsevier Science (USA). All rights reserved.
PII: S 0 1 4 7 - 6 1 9 X ( 0 2 ) 0 0 1 1 7 - 8
A. Mark Osborn, D. B€oltner / Plasmid 48 (2002) 202–212 203

Fig. 1. The mosaic continuum of mobilizable and conjugative genetic elements. MGE (plasmids, phage, and transpo-
sons) that contribute key functional components are circled (dotted lines). MGE functional modules are as given in the
key. Black triangles inserted within the functional modules of the Anchored Genomic Island indicate that molecular
evidence exists for remnants or mutant forms of such modules (see text for further details).

best to classify such elements. Such complications recombinase family, and second, the occurrence of
arise not least from the utilisation of three distinct plasmid-related transfer genes on CTns, and
enzyme families catalysing the vital process of which are being discovered increasingly on GIs.
recombination between DNA molecules (Tous- However, whilst comparative analysis of these
saint and Merlin, 2002). These families are named elements reveals fascinating evolutionary insights,
after key conserved residues in their active site: (i) it is apparent on further inspection that they
tyrosine recombinases (reviewed in Esposito and represent just part of a continuum of mosaic
Scocca, 1997; Nunes-D€ uby et al., 1998), (ii) serine MGE (Fig. 1) that are either self-transmissible or
recombinases (resolvase/invertase) (reviewed in mobilizable between bacterial cells. Thus this re-
Smith and Thorpe, 2002), and (iii) DD-E recom- view will focus also on the emerging groups of
binases (transposases), named following the con- mobilizable transposons comprised of different
served aspartate and glutamate residues (Polard combinations of recombinases and plasmid-re-
and Chandler, 1995). Moreover, the situation is lated mobilisation functions.
complicated further by the additional inclusion on
MGE of genes encoding plasmid-related mobili-
sation and transfer functions. 2. Genomic islands
In this review we focus in particular on two
groups of MGE; the genomic islands (GIs) and Genomic islands, which are found in some
conjugative transposons (CTns), which from their bacterial strains but are absent from otherwise
names at least suggest vastly differing entities. Yet very closely related strains, are now recognised as
molecular comparison of their backbone modules important contributors to bacterial adaptation
identifies key similarities, namely the presence of and evolution. GIs, which vary in size between 10
phage-related integrases belonging to the tyrosine and 500 kb, were first identified as chromosomally
204 A. Mark Osborn, D. B€oltner / Plasmid 48 (2002) 202–212

located virulence genes in uropathogenic Escheri- receiving such islands is often altered dramatically
chia coli, differing in G + C content and codon (e.g., from non-pathogenic to pathogenic, or from
usage from the surrounding DNA (Hacker et al., non-symbiotic to symbiotic), this phenomenon
1983), and subsequently termed pathogenicity is- has been described as Ôevolution in quantum leapsÕ
lands (PAIs) by Hacker et al. (1990). More re- (Groisman and Ochman, 1996).
cently PAIs have been identified in a diverse range A number of the ecological islands have been
of animal pathogens from both Gram-negative demonstrated to be conjugative. For example the
(Yersinia, Salmonella, Vibrio, Helicobacter, and clc element (Ravatn et al., 1998) was initially
Neisseria) and Gram-positive genera (Staphylo- identified following conjugative transfer from
coccus, Listeria, and Clostridium) (Hacker and Pseudomonas strain B13 to P. putida Fl, and
Kaper, 2000). subsequently found to integrate using a tyrosine
In addition, PAIs have also been identified in a recombinase into either of two tRNA gene at-
number of plant pathogens including Erwinia, tachment sites. Similarly, the bph-sal element,
Xanthomonas (No€el et al., 2002), and Pseudomo- encoding biphenyl and salicylate metabolism
nas syringae (Jackson et al., 1999). PAIs carry functions, from Pseudomonas putida KF715 also
genes encoding a variety of phenotypes including transfers by conjugation and is believed to inte-
adhesins, secretion systems and iron uptake sys- grate within a specific insertion hot-spot (Nishi
tems critical to pathogenicity (reviewed in Hacker et al., 2000), whilst a third conjugative element
and Kaper, 2000). Typically PAIs are integrated from Pseudomonas aeruginosa JB2 encoding deg-
into, or near to, tRNA genes and are flanked by radation of hydroxy- and halo-aromatic com-
short direct repeats resembling phage attachment pounds is also believed to integrate within the
sites (Hacker and Kaper, 2000). In addition to the chromosome (Hickey et al., 2001). Conjugative
virulence genes they typically carry an integrase transfer has also been demonstrated for the 500 kb
gene related to that of phage lambda and be- Sym island from Mesorhizobium loti (Sullivan and
longing to the larger family of tyrosine recom- Ronson, 1998), and subsequently in the related
binases. Recent comparison of tRNA attachment islands from M. loti R7A and MAFF303099
sites for the broader group of elements utilising (Sullivan et al., 2002). Other likely members of
tyrosine recombinases, has revealed three sub-lo- this group of Ôconjugative genomic islandsÕ include
cations for integration within these genes. Signif- the related conjugative integrating elements R391
icantly, a phylogeny of integrase sequences has and SXT that carry metal and/or antibiotic resis-
also identified three groups that are consistent tance genes (see below).
with the tRNA sub-location classification (Wil- In Gram-positive bacteria an increasing num-
liams, 2002). ber of potentially transmissible GIs are identified.
In some GIs the integrase gene may be deleted In Streptococcus thermophilus, partial sequence
or non-functional, resulting in permanently an- analysis of the integrative element ICEStl (Burrus
chored islands (see below). Other integrases, et al., 2000) has shown this to be a mosaic con-
however, have been demonstrated to be func- taining a tyrosine recombinase related to that of
tional, and thus, potentially capable of horizontal the CTns Tn5276 and Tn5252, together with
gene transfer, especially if the island also carries plasmid pSK41- and Tn916-related transfer genes.
conjugal transfer genes, which is increasingly ob- Integration of this element has been shown to be
served (see below). Phenotypes carried by genomic site-specific within its host, whilst at present it is
islands are not limited to those encoding virulence not known whether this element is capable of in-
but also include antibiotic resistance e.g., the SRL dependent transfer. More recently, a 153 kb
PAI from Shigella flexneri (Turner et al., 2001), pathogenicity island has been identified in En-
degradation of xenobiotic compounds e.g., the clc terococcus faecalis MMH594 PAI (Shankar et al.,
element encoding chlorocatechol degradation 2002), that includes an integrase most closely re-
(Ravatn et al., 1998) and symbiosis (Sym islands) lated to that from Mesorhizobium loti GIs, and
(Sullivan and Ronson, 1998). Acquisition of such also carries transfer genes related to the Entero-
traits as functional units allows bacteria to re- coccus plasmids pAD1 and pAM373. Although
spond rapidly to environmental challenges and mobilisation of the MMH594 PAI has been
explore new ecological niches. As a consequence demonstrated, it is not currently known whether
such islands have been termed ÔecologicalÕ or Ôfit- the element is also self-transmissible.
nessÕ islands (Hacker and Carniel, 2001). Ac- Whilst the presence of plasmid-related transfer
cordingly, given that the phenotype of bacteria genes on GIs offers conjugation as a dissemination
A. Mark Osborn, D. B€oltner / Plasmid 48 (2002) 202–212 205

mechanism, GIs can also utilise other MGE and capable of excision and subsequent conjugative
mechanisms to facilitate their dispersion. The transfer to recipient cells.
15.2 kb PAI SaPI1 and the related SaPI2 from Prior to the determination of the sequences of
Staphylococcus aureus which both carry the tst these elements and the earlier demonstration of
gene for toxic shock syndrome toxin-1 (Lindsay the site-specific integration of SXT and R391 into
et al., 1998) both contain a putative integrase the prfC gene of E. coli (Hochhut and Waldor,
(tyrosine recombinase) and are flanked by direct 1999), the nature of R391, had represented
repeats. Whilst these may facilitate integration something of an enigma, being variously de-
into the chromosome, UV-inducible excision has scribed as a transmissible resistance factor, an
not been demonstrated, suggesting an absence of integrating plasmid, and subsequently as a con-
an excisionase on these elements. However, the jugative transposon (Murphy and Pembroke,
PAIs can be excised and circularised by certain 1995). The absence of any identifiable plasmid
Staphylococcal phage (80a and /13) to form cir- replicon (B€oltner et al., 2002) is consistent with
cular forms of the PAIs that can then re-integrate repeated failures to isolate ccc DNA and now
in a site-specific manner. Alternatively, replication confirms that these elements are not plasmids.
of the circular form can lead to transduction of Thus, this proposed classification of R391 and
the PAIs at high frequencies, and in the case of SXT as conjugative transposons might at first
SaPI1 this is a direct consequence of SaPI1 repli- seem appropriate, at least at the level of pheno-
cation interfering with that of the phage, in a type, i.e., recA-independent chromosomal inte-
semi-parasitic manner. Following encapsulation gration combined with conjugative transfer.
of SaPI1 and the subsequent transfer to another However, where these elements differ from ar-
cell, SaPI1 uses its own integrase to integrate into chetypal conjugative transposons, such as Tn916,
the recipient hostÕs chromosome (Ruzin et al., is in the absence of random chromosomal inte-
2001). Thus these elements, whilst lacking their gration (see below), with R391 and SXT only in-
own transfer system, are readily transferred intact tegrating into a single site on the E. coli
via phage transduction, offering another example chromosome (see Fig. 1). In this respect, they
of the numerous mechanistic variations found in share greater similarities with the GIs that simi-
the HGP. larly integrate into just one or two sites (typically
tRNA genes) on the chromosome. In retrospect,
on the basis of sequence data and phenotypic and
3. R391, SXT, and the IncJ elements molecular characterisation, R391, isolated 30
years ago, may in fact have been the first genomic
Two notable recent examples of the combina- island to be isolated, albeit a self-transmissable
tion of conjugative plasmid transfer genes with one.
phage-related integration systems are offered by
two related elements; R391 from a South African
isolate of Providencia rettgeri (Coetzee et al., 4. Anchored genomic islands and integrative plas-
1972) and the SXT element from Vibrio cholerae, mids
isolated in India (Waldor et al., 1996). They form
part of a larger series of elements, including R997 Analysis of the 43 kb genomic island (SGI1)
(Matthew et al., 1979) and pMERPH (Peters et al., carrying multi-drug resistances, in Salmonella
1991), classically, though now inappropriately, enterica serovar Typhimurium DT104 (Boyd et al.,
referred to as IncJ elements (following plasmid 2001) indicates a complex evolutionary history.
incompatibility nomenclature). The recent deter- With a failure to detect excision of the SGI1 island
mination of the DNA sequences of R391 (B€ oltner (Boyd et al., 2000) and the inability to demon-
et al., 2002) and SXT (Beaber et al., 2002) shows strate transfer of the multi-drug resistances
them to share conserved backbone regions, with (Threlfall et al., 1994), this island would now ap-
at least 95% identify to each other over 65 kb. In pear to be permanently anchored within the
particular, these backbone regions include a chromosome (Fig. 1). However, the DNA se-
phage-related integrase and associated regulatory quence of SGI1 suggests a possible previous ex-
genes, together with a conjugative transfer system istence as an integrative conjugative plasmid, as
related to that from the plasmid R27 (seen also in indicated by both the presence of a number of
SGI1 from S. enterica DT104—see below). How- ORFs related to genes from the IncHI1 plasmid
ever, in contrast to SGI1, both R391 and SXT are R27 encoding mating pair stabilisation and pilus
206 A. Mark Osborn, D. B€oltner / Plasmid 48 (2002) 202–212

assembly proteins; together with an ORF related also been reported, with reversible integration of
to the replication gene (repA) from the Rhodo- the plasmid pKLK106 into two lysine tRNA
pseudomonas plasmid pMG101. genes in Pseudomonas aeruginosa (Kiewitz et al.,
Past traces of a conjugative lifestyle may also 2000). We have also identified ORFs encoding
be indicated in the Neisseria gonorrhoeae Gono- potential tyrosine recombinases using the PFAM
coccal Genetic Island (GGI) by the presence of database within archaeal plasmids (Osborn and
traG and traH homologues (Dillard and Seifert, B€oltner, unpublished data) namely, ORF439 in
2001), though subsequent insertion mutagenesis the 41.2 kb conjugative plasmid pNOB8 from
suggests the fascinating possibility that these Sulfolobus sp. NOB8H2 (She et al., 1998), and the
genes function instead as a Type IV secretion related ORF457 in the pING plasmid from Sulf-
system acting as a potential DNA donation sys- olobus islandicus (Stedman et al., 2000). Whilst the
tem to facilitate DNA transformation, without function of their gene products is unknown, we
requiring autolysis (Hamilton et al., 2001). In- speculate that these plasmids may represent the
deed, the similarities between plasmid conjugative first examples of integrative plasmids in the ar-
transfer systems and the type IV protein secretion chaea.
systems including those carried by the cag PAI of
Helicobacter pylori or, responsible for extracellu-
lar transport of the pertussis toxin in Bordetella 5. Plasmid-located genomic islands
pertussis, suggest common evolutionary origins
for these systems (Christie, 2001). Given that the Localisation of GIs is not limited to the chro-
incorporation of plasmid-derived conjugative mosome, with a number of elements found lo-
transfer genes into the chromosome is readily cated on plasmids, in particular the PAI on the
demonstrated, it is tempting to speculate on the Shigella flexneri virulence plasmid pWR201
possibility for divergence and subsequent evolu- (Venkatesan et al., 2001) but also notably in the
tion over time of these genes to fulfil new func- discovery of a PAI carrying the three toxin genes
tional roles. Alternatively, such similarities on the plasmid pXO1 from Bacillus anthracis
between DNA and protein transporters may rep- (Okinaka et al., 1999). Whereas this PAI includes
resent divergent evolution of ancestral macro- an integrase gene, it is likely that its insertion into
molecule secretion systems. pXO1 is mediated by insertion sequences, as op-
Chromosomal integration of plasmids has long posed to a tyrosine recombinase (Fig. 1), as evi-
been recognised, most notably of the F plasmid in denced by the presence of copies of IS1627, which
the formation of E. coli Hfr strains. Integration of encode a DD-E type recombinase (transposase) at
F typically occurs by homologous recombination the flanking ends of the 44.3 kb element. Similarly,
between IS elements (IS2 or IS3a and 3b) present sequence analysis of a 27.5 kb PAI found on the
on the F plasmid and chromosome. Whilst inte- 80 kb plasmid p33071 in Rhodococcus equi again
gration by homologous recombination enables suggest involvement of transposons in PAI inser-
plasmids to form stable cointegrates, an increas- tion, as evidenced by the presence of two trans-
ing number of plasmids are reported to carry a poson-related resolvases at either end of the PAI
lambda-related integrase that facilitates recA-in- (Takai et al., 2000) These examples of PAIs that
dependent chromosomal integration and excision. utilise transposons for insertion represent yet an-
For example, the Streptomyces ambofaciens other intriguing illustration of the versatility of
11.2 kb conjugative circular plasmid pSAM2 mosaic MGE.
(Boccard et al., 1989) carries a tyrosine recom-
binase in addition to a rolling circle type replicon
and conjugative transfer genes (Fig. 1). Similarly, 6. Conjugative transposons
a lambda-like integrase mediates integration of
the 11.3 kb plasmid pSE101 into a threonine Conjugative transposons were first identified as
tRNA gene in its host Saccharoployspora eryth- chromosomally-borne transposons that were ca-
raea. In contrast, when pSE101 is, introduced into pable of conjugative transfer. Two such elements
Streptomyces lividans the element demonstrates were originally identified: Tn916 from Enterococ-
integration into multiple sites (Brown et al., 1994) cus faecalis (Franke and Clewell, 1981), which is
more akin to the relatively random integration of now the accepted archetype of the CTns, and
CTns. In Gram-negative bacteria tyrosine re- Tn5253 from Streptococcus pneumoniae (Shoe-
combinase-mediated integration of plasmids has maker et al., 1980). Whilst numerous CTns have
A. Mark Osborn, D. B€oltner / Plasmid 48 (2002) 202–212 207

subsequently been identified, albeit predominantly the TraG protein is homologous to the BctA
from Gram-positive bacteria, much of our mo- protein involved in conjugative transfer of the
lecular understanding of CTns stems from analy- Bacteroides pBF4 plasmid (Morgan and Macrina,
ses of Tn916 and the Bacteroides element 1997), and also to a number of proteins related to
CTnDOT. A full review of the CTns is beyond the the TraC protein from the F plasmid, which is
scope of this current paper, however, we refer involved in pilus assembly. Similarly the TraJ
readers to the excellent reviews by Salyers et al. protein from CTnDOT is related to the TrwI
(1995) and Scott and Churchward (1995). protein from the transfer operon of R388. The
Integration of the majority of CTns utilises a CTnDOT TraA protein is related to a number of
tyrosine recombinase, as is similarly the case for ParA proteins involved in plasmid partitioning.
most GIs. Classical CTns such as Tn916 demon- Of two additional ORFS upstream of the putative
strate semi-random integration in a manner more tra genes on the CTnDOT sequence (accession
redolent of classical transposons. However, Tn916 number: AF289050), the putative protein encoded
does demonstrate distinct preferences for AT-rich by ORF1 is homologous to relaxase proteins from
regions with a consensus target of 50 -TT/ both conjugative plasmids and mobilizable trans-
ATTTT(N6 )AAAAAA/TA-30 (Lu and Church- posons. Thus it is probable that the CTnDOT
ward, 1995), although no individual base within transfer operon represents a distant relative of
this generic sequence is conserved amongst the Gram-negative conjugative plasmid transfer sys-
numerous integration sites identified. In contrast, tems.
CTnDOT shows greater specificity and is found to For both Tn916 and CTnDOT, it is possible
integrate into only about seven sites in the Bac- that the difficulties in identifying relationships to
teroides chromosome (Bedzyk et al., 1992). well-known plasmid conjugation systems may be
Less is known about the transfer system of a consequence that these CTns have been isolated
Tn916, although FASTA analysis (Osborn and from organisms for which our knowledge of
B€oltner, unpublished data) of ORF21 of Tn916 plasmid biology is relatively sparse, notwith-
shows it to be a member of the FtsK/SpoIIIE standing the more detailed characterisation of the
family. SpoIIIE genes are believed to drive DNA Enterococcus pheromone-sensing plasmids pAD1
transport from the mother cell into the prespore (Francia et al., 2001) and pAM373 (De Boever et
during sporulation in Bacillus (Bath et al., 2000) al., 2000). Thus, as more plasmids are sequenced
and the increasing number of SpoIIIE homo- from these organisms, and in particular from
logues suggest that this may be a widespread Bacteroides, this may identify a possible closer
mechanism for DNA transport. Moreover, Spo- linkage between the transfer systems of classical
IIIE homologues are also responsible for conju- CTns and those from conjugative plasmids.
gative transfer of the Streptomyces plasmids Over recent years a number of elements iso-
pIJ101 (Pettis and Cohen, 2001) and pSAM2 lated from members of the proteobacteriaceae
(Hagege et al., 1993). FASTA analysis of ORF20 have been proposed as CTns, including R391, and
(Osborn and B€ oltner, unpublished data) identifies SXT (see above), CTnscr94 from Salmonella
relationships to certain Vibrio phage replication senftenberg (Hochhut et al., 1997) and the M. loti
proteins (Nasu et al., 2000) and the rep gene from strain R7A symbiosis island (Sullivan et al., 2002),
the Bacillus plasmid pGI3 (Hoflack et al., 1997), yet many of these elements would appear to have
suggesting a possible role for this ORF for DNA more in common with genomic islands, albeit
nicking prior to transfer. A functional origin of conjugative ones, on the basis of their preference
transfer has been located between ORFs 20 and for specific integration sites, in particular tRNA
21, and includes sequences related to both IncP genes, as opposed to the more random integration
and F plasmid nic sites (Jaworski and Clewell, exhibited by classical CTns. The 55 kb catabolic
1995). biphenyl degradation transposon Tn4371 from
The transfer system of CTnDOT is unrelated Ralstonia eutropha CH34 does, however, offer a
to that of Tn916-like elements, and is thought to better candidate for a conjugative transposon
represent a new class of conjugal transfer system within the proteobacteriaceae. This element that
(Bonheyo et al., 2001). Following our own FAS- utilises a tyrosine recombinase demonstrates semi-
TA analysis, we again found that many of the random integration into a number of sites present
putative ORFs described show no relationship to on both the CH34 chromosome and the co-resi-
known transfer genes. However, the putative TraP dent pMOL plasmid. However, whilst Tn4371
protein shows homology to DNA primases, whilst carries RP4/Ti-related transfer genes, conjugative
208 A. Mark Osborn, D. B€oltner / Plasmid 48 (2002) 202–212

transfer of the element has not been demon- own mob system by a helper plasmid, such as RP4
strated, although the transfer genes have been (Crellin and Rood, 1992).
demonstrated to be functional via mobilisation of A third type of mobilizable element, Tn5398
a second transposable element Tn-bph (Merlin from C. difficile 630 represents yet another fasci-
et al., 1999). nating variation on this theme, being mobilizable
and capable of semi-random integration upon
transfer to Bacillus subtilis, yet following analysis
7. Mobilizable transposons of the DNA sequence of this element, Tn5398 was
found to lack both a gene encoding any recogni-
Classical transposons utilise one of two types sable recombinase, required for integration, as
of recombinase. Firstly transposons such as Tn3 well as potential mobilisation genes. It does,
and cd utilise a serine recombinase (resolvase/in- however, carry a putative oriT site related to that
vertase). Alternatively, a second group including of the conjugative transposon Tn916 (Fig. 1). It is
Tn7, Tn10, and the IS3 family utilise the so-called postulated that Tn5398 utilises both resolvases
DD-E recombinase (transposase). Upon compar- and mob gene functions carried on the conjugative
ison of such classical transposons with their transposon Tn5397 which is co-resident in the
namesakes, the conjugative transposons (that same host (Farrow et al., 2001).
utilise a tyrosine recombinase), it is attractive to
conjecture upon the existence of truly randomly
integrating, conjugative MGE, perhaps combin- 8. Conjugative transposons or conjugative genomic
ing a DD-E or serine recombinase and a complete islands—that is the question
plasmid-related conjugative transfer system. At
present such systems await discovery, yet an in- The state of MGE nomenclature is a direct
creasing number of mobilizable transposons consequence of their highly mosaic composition,
(MTns), consisting of a transposon that includes with indistinct boundaries between a number of
an oriT site, enabling mobilisation by a co-resi- groups of elements. Obviously, historical prece-
dent conjugative plasmid, have been reported. dence in the naming of individual elements will
The majority of these have been identified in lead to future researchers trying to classify newly
Bacteroides isolates, with the Non-replicating discovered elements as part of existing groupings.
Bacteroides Units (NBUs), NBU1 and NBU2, However, as new DNA sequences are generated
and the mobilizable transposon Tn4555 all iden- and molecular mechanisms unravelled, it is clear
tified as utilising a tyrosine recombinase-based that some earlier classifications are inappropriate,
integration system (Fig. 1) to integrate into either whilst others have led to highly similar MGE be-
tRNA genes (NBU1 and NBU2) (Shoemaker ing classified with very different names. Arguably,
et al., 1996a; Wang et al., 2000), or semi-randomly this is most apparent with respect to the conju-
(Tn4555) [as is the case for Tn916, (Tribble et al., gative transposons (e.g., Tn916 and CTn-DOT)
1997)]. Intriguingly, NBU1, as described above and the increasing number of GIs that encode
for pSE 101, when transferred into E. coli, dem- their own conjugation systems (e.g., the clc ele-
onstrates semi-random integration (Shoemaker ment, the M. loti R7A symbiosis island, and
et al., 1996b), suggesting that site-specificity R391). On molecular comparison of these ele-
of tyrosine recombinase-based systems is a ments, they clearly share a tyrosine recombina-
host-specific phenomenon. Mobilisation of these tion-type integration mechanism, and both utilise
elements utilises oriT sequences and mob genes conjugative transfer systems that are related, al-
resembling those from Gram-positive plasmids beit to differing degrees, to those from conjugative
(Shoemaker et al., 2000; Smith and Parker, 1998; plasmids. Arguably the clearest distinction be-
Wang et al., 2000). tween CTns and GIs is with respect to integration
In contrast, Tn4451 and Tn4453 from Clos- site preference, namely, whether the element in-
tridium spp. represent a second type of MTn that tegrates into multiple sites (as is the case for
instead utilise a serine recombinase (the TnpX Tn916), or into one, or occasionally two, sites, as
resolvase) (Lyras and Rood, 2000) to mediate is typical for the GIs. Perhaps on this basis alone
excision and insertion, and additionally carry a CTns and a second group, for which we propose
plasmid-related mob gene (Fig. 1). These trans- the name Conjugative Genomic Islands, can be
poson-encoded mob determinants permit mobili- delineated. We would suggest that CTn is retained
sation of Tn4551—carrying plasmids lacking their for those classical CTns exhibiting random or
A. Mark Osborn, D. B€oltner / Plasmid 48 (2002) 202–212 209

semi-random integration, whilst those conjugative Beaber, J.W., Hochhut, B., Waldor, M.K., 2002. Geno-
integrating elements which have distinct site- mic and functional analyses of SXT, an integrating
preference, which in many cases will be for a antibiotic resistance gene transfer element derived
tRNA gene, or in the case of R391 and SXT for from Vibrio cholerae. J. Bacteriol. 184, 4259–4269.
Bedzyk, L.A., Shoemaker, N.B., Young, K.E., Salyers,
prfC, are termed conjugative genomic islands (a
A.A., 1992. Insertion and excision of Bacteroides
sub-family of the GIs). However, even here the
conjugative chromosomal elements. J. Bacteriol. 174,
waters are muddied by elements such as the mo- 166–172.
bilizable transposons NBU1 and NBU2, or the Boccard, F., Smokvina, T., Pernodet, J.L., Friedmann,
integrative plasmid pSE101 (see above) that show A., Guerineau, M., 1989. The integrated conjugative
single-site specificity in one host, but integration plasmid pSAM2 of Streptomyces ambofaciens is
into multiple sites in other recipients. As a third related to temperate bacteriophages. EMBO J. 8,
group we propose the adoption of the generic 973–980.
term mobilizable transposon, to cover MGE that B€
oltner, D., MacMahon, C., Pembroke, J.T., Strike, P.,
integrate (irrespective of their type of recombin- Osborn, A.M., 2002. R391: a conjugative integrating
mosaic comprised of phage, plasmid and transposon
ase) into the chromosome, and can be mobilised
elements. J. Bacteriol. 184, 5158–5169.
via conjugation to other cells, but lack a conju-
Bonheyo, G., Graham, D., Shoemaker, N.B., Salyers,
gation system of their own. This term has been A.A., 2001. Transfer region of a Bacteroides conju-
adopted already for a number of MGE (see gative transposon, CTnDOT. Plasmid 45, 41–51.
above), but a number of additional elements, Boyd, D., Peters, G.A., Cloeckaert, A., Boumedine,
would clearly fall within this group. K.S., Chaslus-Dancla, E., Imberechts, H., Mulvey,
Whilst some may ask ÔWhat’s in a name? That M.R., 2001. Complete nucleotide sequence of a 43-
which we call a rose by any other name would smell kilobase genomic island associated with the multi-
as sweet,Õ classification remains an important dis- drug resistance region of Salmonella enterica serovar
cipline within biology. For MGE, one approach is Typhimurium DT104 and its identification in phage
type DT120 and serovar Agona. J. Bacteriol. 183,
to accept their mosaic nature, and classify all
5725–5732.
MGE on the presence of key functional modules.
Boyd, D.A., Peters, G.A., Ng, L.-K., Mulvey, M.R.,
Whilst such an approach is extremely valuable 2000. Partial characterization of a genomic island
(Toussaint and Merlin, 2002), and will increas- associated with the multidrug resistance region of
ingly further benefit from ongoing developments Salmonella enterica Typhimurium DT104. FEMS
of a number of MGE-module specific websites, Microbiol. Lett. 189, 285–291.
the desire to also assign generic names to new and Brown, D.P., Idler, K.B., Backer, D.M., Donadio, S.,
existing elements remains compelling. We hope Katz, L., 1994. Characterization of the genes and
that the MGE research community will face these attachment sites for site-specific integration of plas-
challenges and be willing to embrace appropriate mid pSE101 in Saccharopolyspora erythraea and
Streptomyces lividans. Mol. Gen. Genet. 242, 185–
changes to nomenclature, as needed. As a lead, we
193.
point towards the numerous examples of reclas-
Burrus, V., Roussel, Y., Decaris, B., Guedon, G., 2000.
sification of bacterial genera and species that have Characterization of a novel integrative element,
resulted from ribosomal RNA-based phylogenetic ICEStl, in the lactic acid bacterium Streptococcus
studies over the last 15 years. thermophilus. Appl. Environ. Microbiol. 66, 1749–
1753.
Christie, P.J., 2001. Type IV secretion: intracellular
Acknowledgments transfer of macromolecules by systems ancestrally
related to conjugation machines. Mol. Micro. 40,
294–305.
The authors wish to thank Professor Julian
Coetzee, J.N., Datta, N., Hedges, R.W., 1972. R factors
Rood for an introduction to the serine-recom- from Proteus rettgeri. J. Gen. Microbiol. 72, 543–552.
binase-based mobilizable transposons. Crellin, P.K., Rood, J.I., 1992. Tn4451 from Clostridium
perfringens is a mobilizable transposon that encodes
the functional Mob protein, TnpZ. Mol. Microbiol.
References 28, 631–642.
De Boever, E.H., Clewell, D.B., Fraser, C.M., 2000.
Bath, J., Wu, L.J., Errington, J., Wang, J.C., 2000. Role Enterococcus faecalis conjugative plasmid pAM373:
of Bacillus subtilis SpoIIIE in DNA transport across complete nucleotide sequence and genetic analyses of
the mother cell-prespore division septum. Science sex pheromone response. Mol. Microbiol. 37, 1327–
290, 995–997. 1341.
210 A. Mark Osborn, D. B€oltner / Plasmid 48 (2002) 202–212

Dillard, J.P., Seifert, H.S., 2001. A variable genetic osa strain JB2. Appl. Environ. Microbiol. 67, 4603–
island specific for Neisseria gonorrhoeas is involved 4609.
in providing DNA for natural transformation and is Hochhut, B., Jahreis, K., Lengeler, J.W., Schmid, K.,
found more often in disseminated infection isolates. 1997. CTnscr94, a conjugative transposon found in
Mol. Microbiol. 41, 263–277. enterobacteria. J. Bacteriol. 179, 2097–2102.
Esposito, D., Scocca, J.J., 1997. The integrase family of Hochhut, B., Waldor, M.K., 1999. Site-specific integra-
tyrosine recombinases: evolution of a conserved tion of the conjugal Vibrio cholerae SXT element into
active site domain. Nucl. Acids Res. 25, 3605–3614. prfC. Mol. Micro. 32, 99–110.
Farrow, K.A., Lyras, D., Rood, J.I., 2001. Genomic Hoflack, L., Seurinck, J., Mahillon, J., 1997. Nucleotide
analysis of the erythromycin resistance element sequence and characterization of the cryptic Bacillus
Tn5398 from Clostridium difficile. Microbiology thuringiensis plasmid pG13 reveals a new type of
147, 2717–2728. rolling circle replicon. J. Bacteriol. 179, 5000–5008.
Francia, M.V., Haas, W., Wirth, R., Samberger, E., Jackson, R.W., Athanassopoulos, E., Tsiamis, G.,
Muscholl-Silberhorn, A., Gilmore, M.S., Ike, Y., Mansfield, J.W., Sesma, A., Arnold, D.L., Gibbon,
Weaver, K.E., An, F.Y., Clewell, D.B., 2001. Com- M.J., Murillo, J., Taylor, J.D., Vivian, A., 1999.
pletion of the nucleotide sequence of the Enterococ- Identification of a pathogenicity island, which con-
cus faecalis conjugative virulence plasmid pAD1 and tains genes for virulence and avirulence, on a large
identification of a second transfer origin. Plasmid 46, native plasmid in the bean pathogen Pseudomonas
111–127. syringae pathovar phaseolicola. Proc. Natl. Acad.
Franke, A.E., Clewell, D.B., 1981. Evidence for a Sci. (USA) 96, 10875–10880.
chromosome-borne resistance transposon (Tn916) Jaworski, D.D., Clewell, D.B., 1995. A functional origin
in Streptococcus faecalis that is capable of ‘‘conju- of transfer (oriT) on the conjugative transposon
gal’’ transfer in the absence of a conjugative plasmid. Tn916. J. Bacteriol. 177, 6644–6651.
J. Bacteriol. 145, 494–502. Kiewitz, C., Larbig, K., Klockgether, J., Weinel, C.,
Groisman, E.A., Ochman, H., 1996. Pathogenicity T€ummler, B., 2000. Monitoring genome evolution ex
islands: bacterial evolution in quantum leaps. Cell vivo: reversible chromosomal integration of a 106 kb
87, 791–794. plasmid at two tRNALys gene loci in sequential
Hacker, J., Bender, L., Ott, M., Wingender, J., Lund, B., Pseudomonas aeruginosa airway isolates. Microbiol-
Marre, R., Goebel, W., 1990. Deletions of chromo- ogy 146, 2365–2373.
somal regions coding for fimbriae and hemolysins Lederberg, J., 1952. Cell genetics and hereditary symbi-
occur in vivo and in vitro in various extra-intestinal oses. Physiol. Rev. 32, 403–430.
Escherichia coli isolates. Microb. Pathog. 8, 213– Lindsay, J.A., Ruzin, A., Ross, H.F., Kurepina, N.,
225. Novick, R.P., 1998. The gene for toxic shock toxin is
Hacker, J., Carniel, E., 2001. Ecological fitness, genomic carried by a family of mobile pathogenicity islands in
islands and bacterial pathogenicity. A Darwinian Staphylococcus aureus. Mol. Microbiol. 29, 527–543.
view of the evolution of microbes. EMBO Reports 2, Lu, F., Churchward, G., 1995. Tn916 target DNA
376–381. sequences bind the C-terminal domain of integrase
Hacker, J., Kaper, J.B., 2000. Pathogenicity islands and protein with different affinities that correlate with
the evolution of microbes. Ann. Rev. Microbiol. 54, transposon insertion frequency. J. Bacteriol. 177,
641–679. 1938–1946.
Hacker, J., Knapp, S., Goebel, W., 1983. Spontaneous Lyras, D., Rood, J.I., 2000. Transposition of Tn4451
deletions and flanking regions of the chromosomal and Tn4453 involves a circular intermediate that
inherited hemolysin determinant of an Escherichia forms a promoter for the large resolvase, TnpX.
coli O6 strain. J. Bacteriol. 154, 1145–1152. Mol. Micro. 38, 588–601.
Hagege, J., Pernodet, J.L., Sezonov, G., Gerbaud, C., Matthew, M., Hedges, R.W., Smith, J.T., 1979. Types of
Friedmann, A., Guerineau, M., 1993. Transfer beta-lactamase determined by plasmids in Gram-
functions of the conjugative integrating element negative bacteria. J. Bacteriol. 138, 657–662.
pSAM2 from Streptomyces ambofaciens: character- Merlin, C., Springael, D., Toussaint, A., 1999. Tn4371: a
ization of a kil-kor system associated with transfer. J. modular structure encoding a phage-like integrase, a
Bacteriol. 175, 5529–5538. Pseudomonas-like catabolic pathway, and RP4/Ti-
Hamilton, H.L., Schwartz, K.J., Dillard, J.P., 2001. like transfer functions. Plasmid 41, 40–54.
Insertion-duplication mutagenesis of Neisseria: use Morgan, R.M., Macrina, F.L., 1997. bctA: A novel
in characterization of DNA transfer genes in the pBF4 gene necessary for conjugal transfer in Bacte-
Gonococcal Genetic Island. J. Bacteriol. 183, 4718– roides spp. Microbiology 143, 2155–2165.
4726. Murphy, D.B., Pembroke, J.T., 1995. Transfer of the
Hickey, W.J., Sabat, G., Yuroff, A.S., Arment, A.R., IncJ plasmid R391 to recombination deficient Esc-
Perez-Lesher, J., 2001. Cloning, nucleotide sequenc- herichia coli K12: evidence that R391 behaves as a
ing, and functional analysis of a novel, mobile cluster conjugal transposon. FEMS Microbiol. Lett. 134,
of biodegradation genes from Pseudomonas aerugin- 153–158.
A. Mark Osborn, D. B€oltner / Plasmid 48 (2002) 202–212 211

Nasu, H., Iida, T., Sugahara, T., Yamaichi, Y., Park, Shoemaker, N.B., Smith, M.D., Guild, W.R., 1980.
K.S., Yokoyama, K., Makino, K., Shinagawa, H., DNAse-resistant transfer of chromosomal cat and let
Honda, T., 2000. A filamentous phage associated insertions by filter mating in pneumococcus. Plasmid
with recent pandemic Vibrio parahaemolyticus O3:K6 3, 80–87.
strains. J. Clin. Microbiol. 38, 2156–2161. Shoemaker, N.B., Wang, G.-R., Salyers, A.A., 1996a.
Nishi, A., Tominaga, K., Furukawa, K., 2000. A 90- The Bacteroides mobilizable insertion element,
kilobase conjugative chromosomal element coding NBU1, integrates into the 30 end of a Leu-tRNA
for biphenyl and salicylate catabolism in Pseudomo- gene and has an integrase that is a member of the
nas putida KF715. J. Bacteriol. 182, 1949–1955. lambda integrase family. J. Bacteriol. 178, 3594–
No€el, L., Thieme, F., Nennstiel, D., Bonas, U., 2002. 3600.
Two novel type III-secreted proteins of Xanthomonas Shoemaker, N., Wang, G., Salyers, A.A., 1996b. NBU1,
campestris pv. vesicatoria are encoded within the hrp a mobilizable site-specific integrated element from
pathogenicity island. J. Bacteriol. 184, 1340–1348. Bacteroides spp., can integrate nonspecifically in
Nunes-D€ uby, S.E., Kwon, H.J., Tirumalai, R.S., Ellen- Escherichia coli. J. Bacteriol. 178, 3601–3607.
berger, T., Landy, A., 1998. Similarities and differ- Shoemaker, N.B., Wang, G.-R., Salyers, A.A., 2000.
ences among 105 members of the Int family of Multiple gene products and sequences required
site-specific recombinases. Nucl. Acids Res. 26, 391– for the excision of the mobilizable integrated Bacte-
406. roides element NBU1. J. Bacteriol. 182, 928–
Okinaka, R.T., Cloud, K., Hampton, O., Hoffmaster, 936.
A.R., Hill, K.K., Keim, P., Koehler, T.M., Lamke, Smith, C.J., Parker, A.C., 1998. The transfer origin for
G., Kumano, S., Mahillon, J., Manter, D., Martinez, Bacteroides mobilizable transposon Tn4555 is related
Y., Ricke, D., Svensson, R., Jackson, P.J., 1999. to a plasmid family from Gram-positive bacteria. J.
Sequence and organization of pX01, the large Bacteriol. 180, 435–439.
Bacillus anthracis plasmid harboring the anthrax Smith, C.M., Thorpe, H.M., 2002. Diversity in the serine
toxin genes. J. Bacteriol. 181, 6509–6515. recombinases. Mol. Microbiol. 44, 299–307.
Peters, S.E., Hobman, J.L., Strike, P., Ritchie, D.A., Stedman, K.M., She, Q., Phan, H., Holz, I., Singh, H.,
1991. Novel mercury resistance determinants carried Prangishvili, D., Garrett, R., Zillig, W., 2000. pING
by IncJ plasmids pMERPH and R391. Mol. Gen. family of conjugative plasmids from the extremely
Genet. 228, 294–299. thermophilic archaeon Sulfolobus islandicus: insights
Pettis, G.S., Cohen, S.N., 2001. Unravelling the essential into recombination and conjugation in Cre-
role in conjugation of the Tra protein of Streptomy- narchaeota. J. Bacteriol. 182, 7014–7020.
ces lividans plasmid pIJ101 Anton. V. Leeuwen. Int. J. Sullivan, J.T., Ronson, C.W., 1998. Evolution of rhizo-
Gen. Mol. Microbiol. 79, 247–250. bia by acquisition of a 500 kb symbiosis island that
Polard, P., Chandler, M., 1995. Bacterial transposases integrates into a phe-tRNA gene. Proc. Natl. Acad.
and retroviral integrases. Mol. Microbiol. 15, 13–23. Sci. (USA) 95, 5145–5149.
Ravatn, R., Studer, S., Springael, D., Zehnder, A.J.B., Sullivan, J.T., Trzebiatowski, J.R., Cruickshank, R.W.,
van der Meer, J.R., 1998. Chromosomal integration, Gouzy, J., Brown, S.D., Elliot, R.M., Fleetwood,
tandem amplification, and deamplification in Pseu- D.J., McCallum, N.G., Rossbach, U., Stuart, G.S.,
domonas putida F1 of a 105-kilobase genetic element Weaver, J.E., Webby, R.J., de Bruijn, F.J., Ronson,
containing the chlorocatechol degradative genes C.W., 2002. Comparative sequence analysis of the
from Pseudomonas sp. strain B13. J. Bacteriol. 180, symbiosis island of Mesorhizobium loti strain R7A. J.
4360–4369. Bacteriol. 184, 3086–3095.
Ruzin, A., Lindsay, J., Novick, R.P., 2001. Molecular Takai, S., Hines, S.A., Sekizaki, T., Nicholson, V.M.,
genetics of SaPI1—a mobile pathogenicity island in Alperin, D.A., Osaki, M., Takamatsu, D., Nakam-
Staphylococcus aureus. Mol. Microbiol. 41, 365–377. ura, M., Suzuki, K., Ogino, N., Kakuda, T., Dan,
Salyers, A.A., Shoemaker, N.B., Stevens, A.M., Li, L.- H., Prescott, J.F., 2000. DNA sequence and com-
Y., 1995. Conjugative transposons: an unusual and parison of virulence plasmids from Rhodococcus equi
diverse set of integrated gene transfer elements. ATCC 33701 and 103. Infect. Immun. 68, 6840–
Microbiol. Rev. 59, 579–590. 6847.
Scott, J.R., Churchward, G.G., 1995. Conjugative Threlfall, E.J., Frost, J.A., Ward, L.R., Rowe, B., 1994.
transposition. Ann. Rev. Microbiol. 49, 367–397. Epidemic in cattle and humans of Salmonella
Shankar, N., Baghdayan, A.S., Gilmore, M.S., 2002. typhimurium DT104 with chromosomally integrated
Modulation of virulence within a pathogenicity multiple drug resistance. Vet. Rec. 137, 577.
island in vancomycin-resistant Enterococcus faecalis. Toussaint, A., Merlin, C., 2002. Mobile elements as a
Nature 417, 746–750. combination of functional modules. Plasmid 47, 26–
She, Q., Phan, H., Garrett, R.A., Albers, S.V., Stedman, 35.
K.M., Zillig, W., 1998. Genetic profile of pNOB8 Tribble, G.D., Parker, A.C., Smith, C.J., 1997. The
from Sulfolobus: the first conjugative plasmid from Bacteroides mobilizable transposon Tn4555 inte-
an archaeon. Extremophiles 2, 417–425. grates by a site-specific recombination mechanism
212 A. Mark Osborn, D. B€oltner / Plasmid 48 (2002) 202–212

similar to that of the Gram-positive bacterial element tance to sulfamethoxazole, trimethoprim, and strep-
Tn916. J. Bacteriol. 179, 2731–2739. tomycin in Vibrio cholerae O139. J. Bacteriol. 178,
Turner, S.A., Luck, S.N., Sakellaris, H., Rajakumar, K., 4157–4165.
Adler, B., 2001. Nested deletions of the SRL Wang, J., Shoemaker, N., Wang, G.-R., Salyers, A.A.,
Pathogenicity Island of Shigella flexneri 2a. J. 2000. Characterization of a Bacteroides mobilizable
Bacteriol. 183, 5535–5543. transposon, NBU2, which carries a functional linco-
Venkatesan, M.M., Goldberg, M.B., Rose, D.J., Grot- mycin resistance gene. J. Bacteriol. 182, 3559–3571.
beck, E.J., Burland, V., Blattner, F.R., 2001. Com- Williams, K.P., 2002. Integration sites for genetic
plete DNA sequence and analysis of the large elements in prokaryotic tRNA and tmRNA genes:
virulence plasmid of Shigella flexneri. Infect. Immun. sublocation preference of integrase subfamilies.
69, 3271–3285. Nucl. Acids Res. 30, 866–875.
Waldor, M.K., Tschape, H., Mekalanos, J.J., 1996. A Zinder, N.D., Lederberg, J., 1952. Genetic exchange in
new type of conjugative transposon encodes resis- Salmonella. J. Bacteriol. 64, 679–699.
Integrons and the Origin of Antibiotic
Resistance Gene Cassettes
Super integrons with thousands of gene cassettes may have set the
stage for pathogens to develop antibiotic resistance very rapidly
Didier Mazel

ntegrons, which are natural genetic engi- the late 1930s, and in the late 1940s, mycobac-

I neering platforms, can incorporate open


reading frames and convert them to
functional genes by ensuring correct ex-
pression. Although such integrons play a
teria developed resistance to streptomycin.
These single-resistance phenotypes were not
entirely unforeseen. For example, laboratory
studies had demonstrated penicillin-resistant
major role in multidrug resistance phenomena point mutants. In contrast, investigators did not
among gram-negative species, similar platforms anticipate multidrug resistance because the co-
occur in numerous bacterial species, where they appearance of multiple mutations conferring
can play other roles. Meanwhile, we have evi- such phenotypes was considered to be beyond
dence suggesting that they are a major source of the potential of a given bacterial population.
multidrug resistance integrons and their resis- However, in 1956 in Japan, six years after
tance gene cassettes observed in clinical isolates. physicians began prescribing—and companies
Recruiting exogenous genes represents a began massively producing—the antibiotics
rapid adaptation against antimicrobial com- streptomycin (Sm), tetracycline (Tc), and chlor-
pounds, and the integron functional platform amphenicol (Cm), investigators were observing
seems perfectly suited for capturing those spe- isolates of Shigella dysenteriae resistant to up to
cific genes that enable bacterial pathogens to four antibiotics simultaneously (Tc, Cm, Sm,
face challenges posed by multiple antibiotic Su).
treatment regimes. With the discovery of the This emergence of multiple resistant strains
superintegron and the thousands of cassettes could not be attributed to mutation alone. It was
entrapped in integrons of environmental spe-
soon established that bacteria were acquiring
cies, we now have a better view of the immense
genes that confer resistance, relying on that
resources embedded within this system. How-
means to escape antimicrobial activity rather
ever, several questions of importance about this
than on mutations arising in resident genes. At
system are still without satisfactory answers,
that time, transposons and integrons began ap-
including insights about specific recombination
pearing among human gram-negative pathogens
processes, cassette genesis, and cassette ex-
as well as the commensal bacterial population,
change dynamics within complex bacterial pop-
playing a key role in disseminating resistance
ulations.
Didier Mazel is genes while hitchhiking on conjugative plasmids
head of the Unité and interspecies transfer.
Antibiotic Resistance-Encoding Integrons Nevertheless, integrons were not formally
“Plasticité du Gé-
nome Bactérien”- With few exceptions, antibiotic resistance in identified as agents of resistance gene recruit-
CNRS URA 2171, bacterial pathogens was identified soon after ment until the late 1980s, following the obser-
dept Structure et particular drugs were introduced into clinical vation that loci in transposons and resistance
Dynamique des Gé- practice, illustrating the genetic flexibility of (R)-plasmids expressing different antibiotic re-
nomes, Institut Pas- bacteria. For instance, pathogens developed re- sistance spectra share the same genetic back-
teur, Paris, France. sistance to sulfonamides (Su) and penicillin in bone and differ only in the resistance genes that

520 Y ASM News / Volume 70, Number 11, 2004


they harbor. Regardless of this lag in FIGURE 1
awareness, however, those integrons
were certainly part of the first multi-
drug resistance outbreaks in the 1950s.
One proof is that Tn21, an integron-
containing transposon, was involved in
the resistance phenotype propagated by
plasmid NR1 (R100) in the very first
events in Japan.
In light of the six-year time scale for
the emergence of multidrug-resistant
Shigella strains in Japan in 1956, it was
hardly disputable that bacteria were al-
ready equipped with appropriate ge-
netic tools for meeting the challenge of
multidrug assaults. Indeed, molecular
studies during the last 15 years show
how powerful the bacterial integron re-
combination machinery is, despite its
relative functional simplicity.
Thus, all known integrons are com-
posed of three essential elements for
procuring exogenous genes: (i) a gene
coding for an integrase (intI), (ii) a pri-
mary recombination site (attI), and (iii)
a strong promoter (Pc). Integron inte-
grases recombine, in a recA-indepen-
dent manner, discrete units of circular-
ized DNA known as gene cassettes
downstream of the resident Pc pro- Integron-mediated gene capture and the model for cassette exchange. Outline of the
moter at the proximal attI site, permit- process by which circular antibiotic resistance gene cassettes (antR) are repeatedly
ting expression of their encoded pro- inserted at the specific attI site in a class1 integron downstream of the strong promoter
PC. intI1, integrase encoding gene; Int, integrase IntI1.
teins (Fig. 1). Even considering only
those that differ in nucleotide sequence
by more than 5%, more than 70 differ- from 57 bp to 141 bp in length, and their nucle-
ent resistance cassettes have been described, and otide sequence similarities are primarily re-
they confer resistance to all beta-lactams, all stricted to the inverse core-site and the core-site
aminoglycosides, chloramphenicol, tri- (Fig. 2).
methoprim, streptothricin, rifampin, erythro- Three classes of integrons (RI) involved in
mycin, and antiseptics of the quaternary ammo- multidrug resistance expression were defined on
nium compound family. the basis of homology of their integrase genes.
Moreover, all these integron-inserted cas- Each class appears capable of sharing and ac-
settes share several specific structural character- quiring the same gene cassettes. Several cassettes
istics. For instance, the boundaries of each inte- appear to belong in two different classes of
grated cassette are defined by two GTTRRRY integrons, and the class 1 integrase can recom-
(core-site) sequences in the same orientation, bine several structurally diverse attC sites. The
which are the targets of the recombination pro- IntI integrases belong to the catalytic family of
cess. The integrated cassettes also generally in- the tyrosine (Y) recombinases that are involved
clude a single gene and an imperfect inverted in the movement of numerous phages through
repeat located at the 3⬘ end of the gene called an site-specific recombination (such as the lambda
attC site (or “59-base element”), a diverse family phage integrase, ␭Int) or in fundamental cellular
of sequences that function as recognition sites processes such as chromosome dimer resolution
for the site-specific integrase. The attC sites vary in cell division (XerC/D). In spite of these rela-

Volume 70, Number 11, 2004 / ASM News Y 521


FIGURE 2

Structural comparison of a “classical” multi-resistant integron and the V. cholerae N16961 super-integron. Top, schematic representation
of In40; the various resistance genes are associated with different attC sites (see text). Antibiotic resistance cassettes confer resistance
to the following compounds: aacA4, aminoglycosides; carb4, beta-lactams; cmlA2, chloramphenicol; qac, quarternary ammonium
compounds; oxa9, beta-lactams. The sul gene, which provides resistance to sulfonamides, is not a gene cassette. Bottom, the ORFs are
separated by highly homologous sequences, the VCRs.

tionships, the integron integrases have a unique repeats), led to discovery of another type of inte-
characteristic among the Y-recombinases, as they gron, a superintegron (SI) (Fig. 2). This distinct
recombine sequences that are only remotely re- type of integron is now known to be an integral
lated. component of many g-proteobacterial genomes.
The RI platforms are defective for self-trans- The integron discovered in chromosome 2 of
position, but this defect is often complemented V. cholerae possesses a specific integrase, IntIA,
through association with IS, transposons, that is related to the RI integrases and also is
and/or conjugative plasmids which can serve as responsible for inserting coding sequences
vehicles for the intra- and interspecies trans- (ORFs) into a unique chromosomal attI site.
mission of genetic material. With this system, However, this SI has two structural characteris-
bacteria are capable of stockpiling exogenous tics that distinguish it from other RIs—namely,
genetic loci to establish an appreciable anti- the large number of cassettes that it gathers and
microbial armamentarium. Some bacteria habor the high homology observed among the attC
up to eight different resistance cassettes in a
sites of these cassettes, which in the case of V.
single integron.
cholerae are known as VCRs.
These key features plus the sedentary nature
Another Type of Integron: the of the functional platform (IntIA ⫹ attI site)
Chromosomal Superintegrons define a superintegron. Such SI structures also
A pivotal question is, what are the origins of the are found among the Vibrionaceae and their
RI and their cassettes? The degree of homology close relatives, the xanthomonads, and a branch
between the three RI integrases (45–58%) sug- of the pseudomonads. They share the same gen-
gests that their evolutionary divergence extends eral characteristics, such as their large number
longer than the 50 years since antibiotics came of more than 20 cassettes and a high homology
into use. In the late 1990s, studies examining the between their endogenous cassette attC sites.
relationship between RI gene cassette arrays and More importantly, they predate the antibiotic
a cluster of repeated sequences identified in the era, and have been identified in isolates collected
Vibrio cholerae genome, called VCRs (V. cholerae from the 19th century.

522 Y ASM News / Volume 70, Number 11, 2004


The SI carried in three Vibrio species whose Y-recombinase family. Moreover, the integron
genomes are sequenced all show a large number platform appears to be ancient based on the
of cassettes, from 72 in V. parahaemolyticus to species-specific clustering of the respective SI
more than 200 in V. vulnificus. In the case of the integrase genes in a pattern that mostly adheres
V. cholerae strain El Tor N16961, the SI gathers to the line of descent among the bacterial species
at least 216 mostly unidentified genes in an in which they are found. Thus, the establishment
array of 179 cassettes that starts from the of SIs likely predates speciation within respec-
VchintIA gene and occupies about 3% of the tive genera, suggesting that integrons are ancient
genome. The high level of identity shared by the structures that steered the evolution of bacterial
attC sites carried by the majority of these cas- genomes for hundreds of millions of years.
settes suggests that they are assembled in the However, it is possible that transfer of either a
SI-carrying species through the physical associ- part or all of a SI occurred long ago, perhaps
ation of an attC site with an incoming DNA explaining some of the discrepancies observed
fragment. However, the mechanics of this pro- between the SI-integrase and 16S rRNA gene
cess are not known. trees, such as for V. fischeri.
Integrons that do not share all the character- Because the RIs and SIs have a common struc-
istics of the SI have been found in the genome of tural organization, the antiquity of SIs suggests
two Shewanella species. These integrons gather that they are the ancestors of RIs. Hence, we
only a handful of cassettes with structurally proposed that RIs evolved from SIs through
heterogeneous attC sites, a situation reminiscent entrapment of intI genes and their cognate attI
of the RI cassette arrays. Furthermore, integron sites by mobile structures, such as easily and
integrase-like genes have also been identified in randomly assembled compound transposons.
the genomes of other proteobacteria, including Furthermore, we noticed that 12 different resis-
Nitrosomonas europaea, Microbulbifer degrad- tance cassettes carry an attC site almost identical
ans, Geobacter sulfurreducens, and Treponema to those specifically found in and characteristic
denticola. Apart from the one carried in N. of Xanthomonas and Vibrio SIs (Fig. 3). Once
europea, they have not been characterized. mobile, one can imagine that the subsequent
Using PCR primers directed against con- harvesting of cassettes from various SI sources
served regions of the integron-integrase genes led to contemporary RIs, including the great
and attC sites, Harold Stokes of Macquarie Uni- diversity of attC sites associated with these gene
versity, Sydney, Australia, and Steven Schmidt cassettes.
of the University of Colorado, Boulder, and Two recent observations support this account
their respective collaborators amplified 19 new of how RIs could have arisen from SIs. The first
integron integrases from markedly different en- came from Henning Sørum of the Norwegian
vironmental DNA samples, including from na- School of Veterinary Science in Oslo and his
tional parks or metal-contaminated soils. Un- collaborators. While analyzing a plasmid asso-
fortunately, in most cases their protocol does ciated with trimethoprim resistance in V. salmo-
not permit determining the source of these inte- nicida, they found that resistance is due to a
grons, be it the endogenous SI of a soil bacte- dfrA1 cassette carried in an integron (Genbank
rium or an integron located on a mobile struc- #AJ277063) that contains a “classical” dfrA1
ture. However, their findings support the cassette in second position and is followed by six
hypothesis that integrons are widespread among cassettes. This new integron has two features
bacterial populations either as components of that corroborate our model: (i) its integrase orig-
mobile DNA elements or the chromosome, and inated from a Vibrio species, or one from a
that they are not confined to pathogenic or closely related genus, as attested to by its 74%
multidrug-resistant bacteria. similarity to the V. cholerae and V. mimicus
IntIA; and (ii) the attC sites of three of the seven
cassettes are structurally homogeneous, a char-
Phylogeny Indicates Integrons Are
acteristic until now only found in the SI cassette
Ancient, Super Integrons Possibly
arrays. Because this integron is surrounded by IS
Ancestral
sequences, we suspect that it corresponds to an
According to comparative analyses, the inte- intermediate of what could possibly became a
gron-integrases form a specific clade within the true RI by harvesting other resistance cassettes,

Volume 70, Number 11, 2004 / ASM News Y 523


FIGURE 3

A model for super-integrons as the source of the resistance gene cassettes of multi-resistant integrons. The pool of RI gene cassettes
is depicted as being derived from the reservoirs of SIs. The attC sites are represented as colored triangles. The structural relationships
between the attC sites of resistance cassettes found in RIs and those characteristic of identified, or unknown yet, SIs are indicated by
a larger triangle of the same color.

using the mobility of the assembled compound Meanwhile, another resistance cassette, cod-
transposon and multiple transfers. ing for a novel carbenicillinase, has been identi-
The second observation from our group in- fied in the SI of another V. cholerae isolate. This
volves SI cassettes that could be recruited by a cassette, like the catB9 cassette, is structurally
RI, and, moreover, confer a resistance pheno- identical to the other V. cholerae SI cassettes, as
type. The majority of the examined SI cassettes its attC site is a canonical VCR. Along with
appear unique to the host species, and a major- results demonstrating that SI cassettes are sub-
ity of their encoded genes have no counterparts strates for the integrase of class 1 integrons
in the database or are the sole homologs of when present on a high-copy-number plasmid,
unassigned ORFs. Nevertheless, the reservoir of these results suggest that environmental condi-
adaptive functions residing within SIs includes tions, such as the presence of antibiotics, dictate
cassettes with significant homology to known which of the randomly recruited cassettes are
antibiotic resistance genes. We have demon- retained within the RIs of clinical isolates.
strated that class 1 integrons can randomly re- It is likely that the assembly of complex RIs
cruit any gene cassettes harbored within a SI. having more than two resistance cassettes oc-
After we applied selective pressure for antibiotic curred through recombination between differ-
resistance, we discovered a chloramphenicol ent RI, rather than through successive direct
acetyltransferase gene cassette, catB9, in the V. recruitment from the SI cassette arrays of envi-
cholerae SI. ronmental species. Niches exist which could fa-

524 Y ASM News / Volume 70, Number 11, 2004


vor the exchange of cassettes between RI on a nomycetes that produce aminoglycosides, and
high scale. The recent characterization of a re- the APH encoded in transposons such as Tn5.
markable collection of cassettes, integrons, and The best example is certainly the strong similar-
plasmids circulating in a single wastewater ity in sequence and genetic organization ob-
treatment plant, as well as the unpredicted dem- served by Gerard Wright of McMaster Univer-
onstration that aerobic, gram-positive Coryne- sity, Hamilton, Ontario, Canada, between the
bacteria can be a reservoir of class 1 RI in vancomycin resistance gene cluster found in en-
poultry litter, indicates that environments other terococci and those of the glycopeptide-produc-
than obvious clinical settings exist in which cas- ing actinomycetes. While no other direct evi-
sette exchanges among RI may be common. dence is presently available, many antibiotic-
producing strains other than those used
industrially exist in nature and could be a source
Trying To Trace Sources of Resistance
of resistance genes.
Genes in Integron Cassettes
The recruitment of general housekeeping
As for antibiotic resistance genes that are not genes provides an alternate route by which re-
carried in cassettes, key questions about their sistance determinants might evolve. For in-
origins and how they disseminate remain unan- stance, several examples of chromosomally en-
swered. Nonetheless, evidence suggests that coded homologs whose functions are not related
horizontal gene transfer among prokaryotes is a to antibiotic resistance have been identified in
perpetual activity. So ancient is this process that the genomes of Serratia marcescens, Providen-
determining a definitive source for any particu- cia stuartii, Streptomyces spp., and Mycobacte-
lar resistance marker will surely prove diffi- rium spp.
cult—unless a recent event can be identified. The marked differences in codon usage
In 1973, Raoul Benveniste and Julian Davies among cassettes within the same integrons indi-
proposed that intrinsically resistant or antibiot- cate that these several antibiotic resistance de-
ic-producing organisms routinely supply resis- terminants are of diverse origins. However, evo-
tance genes to clinical isolates, and some recent lutionary rates and the passing of genes through
findings substantiate their hypothesis. Their intermediates undoubtedly created descendants
proposal was based in part on the structural that are appreciably divergent from the original
relationship between the aminoglycoside phos- loci, meaning the original source of a given
photransferase (APH), which protects the acti- resistance gene will remain elusive.

ACKNOWLEDGMENTS
I thank all the members, past and present, of my group. I especially thank D. Rowe-Magnus and M.-C. Ploy for their critical
reading of the manuscript. This work was supported by the Pasteur Institute and the CNRS.

SUGGESTED READING
Collis, C. M., G. D. Recchia, M. J. Kim, H. W. Stokes, and R. M. Hall. 2001. Efficiency of recombination reactions catalyzed
by class 1 integron integrase IntI1. J. Bacteriol. 183:2535–2542.
Hall, R. M., and C. M. Collis. 1998. Antibiotic resistance in gram-negative bacteria: the role of gene cassettes and integrons.
Drug Resist. Updates 1:109 –119.
Nandi, S., J. J. Maurer, C. Hofacre, and A. O. Summers. 2004. Gram-positive bacteria are a major reservoir of Class 1
antibiotic resistance integrons in poultry litter. Proc. Natl. Acad. Sci. USA 101:7118 –7122.
Nemergut, D. R., A. P. Martin, and S. K. Schmidt. 2004. Integron diversity in heavy-metal-contaminated mine tailings and
inferences about integron evolution. Appl. Environ. Microbiol. 70:1160 –1168.
Rowe-Magnus, D. A., A.-M. Guerout, P. Ploncard, B. Dychinco, J. Davies, and D. Mazel. 2001. The evolutionary history of
chromosomal super-integrons provides an ancestry for multi-resistant integrons. Proc. Natl. Acad. Sci. USA 98:652– 657.
Rowe-Magnus, D. A., A. M. Guerout, L. Biskri, P. Bouige, and D. Mazel. 2003. Comparative analysis of superintegrons:
engineering extensive genetic diversity in the Vibrionaceae. Genome Res 13:428 – 442.
Rowe-Magnus, D. A., A. M. Guerout, and D. Mazel. 2002. Bacterial resistance evolution by recruitment of super-integron
gene cassettes. Mol. Microbiol. 43:1657–1669.
Stokes, H. W., A. J. Holmes, B. S. Nield, M. P. Holley, K. M. Nevalainen, B. C. Mabbutt, and M. R. Gillings. 2001. Gene cassette
PCR: sequence-independent recovery of entire genes from environmental DNA. Appl. Environ. Microbiol. 67:5240 –5246.
Tennstedt, T., R. Szczepanowski, S. Braun, A. Puhler, and A. Schluter. 2003. Occurrence of integron-associated resistance
gene cassettes located on antibiotic resistance plasmids isolated from a wastewater treatment plant. FEMS Microbiol. Ecol.
45:239 –252.

Volume 70, Number 11, 2004 / ASM News Y 525


Molecular Microbiology (2002) 43(6), 1657–1669

Bacterial resistance evolution by recruitment of


super-integron gene cassettes
Dean A. Rowe-Magnus,† Anne-Marie Guerout and clinically relevant pathogens at high frequency. These
Didier Mazel* results demonstrate that otherwise phenotypically
Unité de Programmation Moléculaire et Toxicologie sensitive strains may still be a genetic source for
Génétique, CNRS URA 1444, Département des the evolution of resistance to clinically relevant
Biotechnologies, Institut Pasteur, 25 rue du Dr Roux, antibiotics through integron-mediated recombination
75724 Paris, France. events.

Summary Introduction
The capture and spread of antibiotic resistance deter- The horizontal transfer of genetic material within micro-
minants by integrons underlies the rapid evolution of bial communities has been instrumental in the emergence
multiple antibiotic resistance among diverse Gram- of novel functions and species (de la Cruz and Davies,
negative clinical isolates. The association of multiple 2000; Ochman et al., 2000). The ‘antibiotic resistance
resistance integrons (MRIs) with mobile DNA ele- phenomenon’ is perhaps the most striking recent example
ments facilitates their transit across phylogenetic of the impact of horizontal transfer on microbial adapta-
boundaries and augments the potential impact of tion. This phenomenon refers to the rapid and widespread
integrons on bacterial evolution. Recently, ancestral emergence of similar antibiotic resistance profiles among
chromosomal versions, the super-integrons (SIs), phylogenetically diverse Gram-negative clinical isolates
were found to be genuine components of the over the last half-century (Davies, 1994). The source of
genomes of diverse bacterial species. SIs possess these antibiotic resistance determinants is often unknown,
evolutionary characteristics and stockpiles of but many are thought to have evolved or, more signifi-
adaptive functions, including cassettes related to cantly, been acquired in response to aeons of natural
antibiotic resistance determinants previously charac- bacterial competition (Davies, 1994; 1997). At the root of
terized in clinical isolates, which suggest that MRIs antibiotic resistance gene acquisition is the unique
and their resistance genes were originally recruited activity of integrons. Integrons are natural genetic engi-
from SIs and their pool of amassed genes. However, neering systems that incorporate circularized open
the recombination activity of integrons has never reading frames, called gene cassettes, and convert them
been demonstrated in a bacterium other than to functional genes (Martinez and de la Cruz, 1990; Collis
Escherichia coli. We introduced a naturally occurring and Hall, 1992; Collis et al., 1993; Hall and Collis, 1995).
MRI (TpR, SulR) on a conjugative plasmid into Vibrio The integron platform consists of an integrase gene of the
cholerae, a species known to harbour a SI. We show tyrosine recombinase family (Nunes-Duby et al., 1998)
that MRIs can randomly recruit genes directly from and a primary recombination site, called the attI site,
the cache of SI cassettes. By applying a selective located proximal to the integrase gene. The integrase
constraint for the development of antibiotic resis- catalyses recombination between its cognate attI site
tance, we demonstrate bacterial resistance evolution and a secondary target called an attC site (or 59 base
through the recruitment a novel, but phenotypically element). The attC site is normally found associated
silent, chloramphenicol acetyltransferase gene from with a single open reading frame (ORF) that encodes an
the V. cholerae SI and its precise insertion into the adaptive function, and the ORF–attC structure is termed
MRI. The resulting resistance profile (CmR, TpR, SulR) a gene cassette (Hall and Stokes, 1993; Recchia and
could then be disseminated by conjugation to other Hall, 1995; Stokes et al., 1997; Sundstrom, 1998).
Recombination between the attI and attC sites leads to
insertion of the gene cassette downstream of a resident
Accepted 24 December, 2001. *For correspondence. E-mail
mazel@pasteur.fr; Tel. (+33) 1 40 61 32 84; Fax (+33) 1 45 68 88 34. promoter, Pc, within the integron (Levesque et al., 1994;

Present address: Department of Microbiology and Division of Clini- Collis and Hall, 1995) that drives expression of the
cal Integrative Biology, Sunnybrook and Women’s College Hospital encoded product. The strongest promoter variant of Pc
Health Sciences Centre Research Institute, 2075 Bayview Avenue,
Toronto, Canada M4N 3N5, and The Department of Laboratory (TTGACAN17TAAACT) is six times more efficient than
Medicine and Pathobiology, University of Toronto, Toronto, Canada. the derepressed PTac promoter (Levesque et al., 1994).
© 2002 Blackwell Science Ltd
1658 D. A. Rowe-Magnus et al.

The most notable gene cassettes identified within for antibiotic resistance genes whereas those of SIs are
integrons are those conferring resistance to antibiotics. of mainly unknown function. Third, MRIs are commonly
More than 70 different antibiotic resistance genes, cover- associated with mobile DNA elements. Conversely, the
ing most classes of antimicrobials presently in use, are coevolution of SI integrases with their host genome
structured as gene cassettes (Hall and Collis, 1998; (Rowe-Magnus et al., 2001) strongly suggests that SIs
Mazel and Davies, 1999). The stockpiling of these cas- are sedentary. Finally, whereas the attC sites of the gene
settes in integrons, to create multiresistance integrons cassettes of MRIs are highly variable in length and
(MRIs), has contributed substantially to the current sequence, the attC sites of SI gene cassettes are closely
dilemma in the treatment of infectious disease, as inte- related and species specific (Rowe-Magnus et al., 2001).
grons containing up to eight resistance cassettes have These relationships led to the proposal that MRIs evolved
been found in multiple-resistant clinical isolates (Naas from SIs through the entrapment of intI genes and their
et al., 2001). Furthermore, integron platforms are often cognate attI sites by mobile DNA elements (Rowe-
found embedded within mobile DNA elements such as Magnus et al., 2001). The subsequent harvesting of
transposons and/or conjugative plasmids that can serve cassettes from various SI sources then led to the estab-
as vehicles for the intra- and interspecies transmission of lishment of contemporary MRIs. However, the ability of
the resistance genes that have been amassed by inte- integrons to recruit gene cassettes has never been
grons. The proficiency of this partnership is confirmed by demonstrated to occur in a bacterium other than E. coli.
the marked differences in codon usage among cassettes Furthermore, the direct recruitment of cassettes from a SI
within the same MRI, indicating that the antibiotic resis- by a MRI has never been observed.
tance determinants are of diverse origin. Functional analysis of the SI cassettes of V. cholerae
Five classes of MRIs have been defined based on the strain N16961 unveiled a variety of adaptive functions,
homology of the integrase genes (Martinez and de la Cruz, including metabolic activities, virulence traits and poten-
1990; Sundstrom et al., 1991; Arakawa et al., 1995; tial antibiotic resistance determinants (Rowe-Magnus
Hochhut et al., 2001; Sorum et al., GenBank accession et al., 1999; Heidelberg et al., 2000). We sought to deter-
no. AJ277063). They share between 39% and 58% amino mine if any of these ORFs could confer resistance to
acid identity, which suggests that their evolutionary current antibiotics and become a source of the resistance
divergence has extended over a much longer period of genes found in MRIs. Here, we show that the gene cas-
time than the 50 years of the antibiotic era. The likely sette reservoirs of SIs are a source of the gene cassettes
ancestors of MRIs are the recently discovered chromo- identified within MRIs. We demonstrate that MRIs ran-
somal super-integrons (SIs) that have been identified in the domly recruit gene cassettes directly from SIs. By then
genomes of diverse Gram-negative proteobacteria (Mazel applying a selection for the development of antibiotic
et al., 1998; Clark et al., 2000; Rowe-Magnus et al., 2001; resistance, we followed the real-time evolution of bac-
Vaisvila et al., 2001). The first SI was discovered in the terial resistance through the recruitment by a MRI of a
V. cholerae genome (Mazel et al., 1998). Clustered in one novel, but phenotypically silent, chloramphenicol acetyl-
region of chromosome II, the V. cholerae SI spans transferase gene from the V. cholerae SI. The resulting
126 kb and harbours 214 open reading frames of mainly resistance profile (CmR, TpR, SulR) could then be dis-
unassigned function (Rowe-Magnus et al., 1999; seminated by conjugation to other clinically relevant
Heidelberg et al., 2000) in 179 cassettes. Its cassettes vary pathogens at high frequency.
extensively in base composition and codon usage, and
appear to be of bacterial, viral and eukaryotic origin. MRIs Results
and SIs share an identical structural organization, with an
The evolution of a MRI through the recruitment of a
intI gene divergently transcribed from an upstream cluster
novel chloramphenicol acetyltransferase gene cassette
of gene cassettes (Rowe-Magnus and Mazel, 1999).
from the V. cholerae SI by a class 1 integron
Furthermore, the integrases of both MRIs and SIs are
functional for cassette recombination (Rowe-Magnus The majority of integron gene cassettes are known to
et al., 2001), and their integrases form a group related to be promoterless (Hall, 1997) and are probably poorly
but distinct from other members of the l family of site- expressed as a result. Using plasmids containing cloned
specific tyrosine recombinases (Nunes-Duby et al., 1998). class 1 integron fragments that differed only in the order of
Despite these commonalities, four major differences their inserted resistance gene cassettes, Collis and Hall
exist between MRIs and SIs. First, MRIs typically contain (1995) have shown that cassettes closest to the promoter,
fewer than six cassettes, with the largest described to PC, are more highly expressed than distal cassettes. Thus,
date containing eight cassettes. As is the case in V. depending on the genetic context in which they are found,
cholerae, SIs may contain hundreds of cassettes. the potential antimicrobial activity of silent determinants
Second, the cassettes found within MRIs typically code encoded in SI gene cassettes may be overlooked. We

© 2002 Blackwell Science Ltd, Molecular Microbiology, 43, 1657–1669


Bacterial trafficking of integron gene cassettes 1659
Table 1. Potential antibiotic resistance cas-
ORF number on chromosome II b Proposed functionc E-valuec settes in the V. cholerae N16961 SIa.

VCA0300 (catB9) Chloramphenicol acetyltransferase 5 e–71


VCA0328, VCA0341, VCA0463d Fosfomycin glutathione-transferase 6 e–4
VCA0387 Phosphinothricin acetyltransferase 6 e–31
VCA0473 Streptogramin acetyltransferase 2 e–31

a. These four cassettes are also present in the 63.32 SI (see Experimental procedures); 10
other genes found in the N16961 SI cassettes (VCA0316, VCA0400, VCA0402, VCA0417,
VCA0436, VCA0442, VCA0474, VCA0479, VCA0496, VCA0505) code for potential acetyl-
transferases and a glutathione transferase.
b. The 214 ORFs in the 179 cassettes in the SI of V. cholerae strain N16961 are numbered
from VCA0292 to VCA0506 (Heidelberg et al., 2000).
c. According to BLAST analysis (http://www.ncbi.nlm.nih.gov/BLAST/).
d. Cassettes are identical in sequence.

determined the sensitivity profiles of V. cholerae isolates cassette homologues that we observed in the SI of V.
obtained since 1935. Our profiles indicated that early V. cholerae N16961 could confer resistance to their sug-
cholerae isolates, including strain N16961, were sensitive gested corresponding antibiotic (Table 1), we sought to
to the classes of antibiotics active against Gram-negative relocate SI gene cassettes to the insertion site of a class
bacteria. To determine if any of the resistance gene 1 integron (Fig. 1). This would place the SI cassettes

Fig. 1. The V. cholerae conjugative assay system. For clarity, only a portion of the V. cholerae SI on chromosome II is shown. The 214 ORFs
of the 179 SI gene cassettes are numbered from VCA0292 to VCA0506. Any of these (represented as VCA0xxx) may be recruited by In3.
The V. cholerae signature attC sites, the VCRs, are shown as black triangles; the unrelated attC sites typical of MRI cassettes are shown as
white and stripped triangles in In3. The dark wavy lines and black boxes represent chromosomal DNA and ORFs that are not part of the SI
structure. The class 1 integron–integrase protein, IntI1, is expressed from p112 (large white arrow) under the control of the IPTG-inducible trc
promoter. IntI1 catalyses site-specific recombination between the attC sites of gene cassettes, leading to random excision of the SI gene
cassettes (white and stripped boxes) in circular form. The subsequent integration of the free cassettes at the attI1 site (small white circle)
of In3, a naturally occurring class 1 integron on the conjugative plasmid R388, places the cassette downstream of an active promoter, Pc (bent
arrow), that drives expression of the cassette-encoded protein. R388 and its derivatives are then conjugated (thick black arrow) to a NalR
recipient and transconjugants are selected on the appropriate sets of antibiotics. VchintIA, the V. cholerae SI integrase gene; stripped box, a
potential antibiotic resistance gene cassette. The dfrB2 gene cassette in the In3 codes for trimethoprim resistance. The relative positions of
the attI1.1 and dfrfus primers are shown (small arrows).

© 2002 Blackwell Science Ltd, Molecular Microbiology, 43, 1657–1669


1660 D. A. Rowe-Magnus et al.

downstream of a fully active resident promoter to ensure


their expression and allow us to ascertain if resistance
could be attained by high-level expression of these antibi-
otic resistance gene cassette homologues.
In order to detect the trafficking of cassettes between
SIs and MRIs, we utilized a conjugation system involving
two plasmids. The first, p112, contains the intI1 gene,
which codes for the integrase of class 1 integrons under
the control of the inducible trc promoter. The second har-
bours a naturally occurring class 1 target integron, In3, on
the conjugative plasmid R388 (Avila and de la Cruz, 1988;
Martinez and de la Cruz, 1990). In3 contains the strong
promoter variant of PC, the dfrB2 gene cassette, which
confers resistance to trimethoprim, and an open reading
frame of unknown function, ORFA. We verified by poly-
merase chain reaction (PCR) that the resistance gene
cassette homologues shown in Table 1 were also present
in the SI of V. cholerae CIP 63.32 (see Experimental Pro-
cedures). Both plasmids were introduced into V. cholerae
strain CIP 63.32 to create strain 1275. As the complete
sequence of the SI of V. cholerae strain CIP 63.32 was
not known, we extended our selection to include not only Fig. 2. Thin-layer chromatography of the [14C]-Cm CAT assay
products. The CAT assay was performed as described in the
the antibiotics listed in Table 1 but also those for which
Experimental procedures. Lanes: 1, control strain UB5201 (R388);
resistance determinants structured as integron gene cas- 2, UB5201a (R388::catB9).
settes had already been described (Mazel and Davies,
1999). Bacterial resistance evolution was then assayed
by monitoring integrase-mediated integration of SI cas- of an arabinose-inducible promoter. Transformants were
settes into In3 (Fig. 1). found to be resistant to Cm in an induction-dependent
Following induction of intI1 gene expression, R388 was manner, whereas control strains containing vector without
transferred to E. coli and transconjugants were plated on insert remained susceptible regardless of the induction
a number of different antibiotics. Of the antibiotics tested, state (data not shown).
bacteria that were resistant to chloramphenicol (Cm)
were recovered (MIC of 128 mg ml–1). Sequence analysis
An integron-mediated switch of V. cholerae from a
of several transconjugants revealed that a SI cassette,
Cm-sensitive to a Cm-resistant state
corresponding to VCA0300 of V. cholerae strain N16961
(Table 1), had been precisely inserted into In3. The According to our antibiograms, the parent strain V.
VCA0300 cassette contained a single ORF of 630 cholerae 63.32 was sensitive to Cm (minimum inhibitory
nucleotides with significant homology to chloramphenicol concentration <1 mg ml–1). The fact that the catB9 gene
acetyltransferases (CATs) (64.9%, 65.1%% and 65.9% cassette did not contain a promoter potentially explained
amino acid identity to CATB6, CAT X82455 and CATB8 this sensitivity. In the CmR E. coli transconjugants, catB9
respectively; accession numbers AJ223604, X82455 and was precisely inserted into the first position of In3, which
AF227506; Fig. 2). Typical of integron gene cassettes, this placed the cassette downstream of the resident PC pro-
cassette did not contain a promoter element. Several moter. This suggested that in its native SI position the
E. coli transconjugants were selected and tested for the catB9 gene might not be sufficiently expressed to confer
presence of CAT activity. A standard CAT assay (Gorman resistance. To investigate this possibility, a 3.8 kb region
et al., 1982) verified 1-acetoxy- and 3-acetoxy-CAT activ- containing the attI4 site and the first seven cassettes up
ities in the transconjugants, whereas E. coli harbouring to the catB9 gene of the V. cholerae strain 63.32 SI was
native R388 showed no CAT activity (Fig. 3). This novel cloned downstream of the PLac promoter in the plasmid
CAT gene cassette was designated catB9. pSU38 (Fig. 4). In addition, sequential deletions of the cas-
To verify that the catB9 gene cassette alone was settes within the fragment were constructed in the same
responsible for the observed CmR phenotype and that no vector. Each construct was then tested for its ability to
other element carried by R388 participated in the CmR permit growth of E. coli transformants on media containing
phenotype, we amplified catB9 by PCR from V. cholerae 25 mg ml–1 Cm. Transformants with the catB9 gene in its
strain 63.32 and expressed it in E. coli under the control native seventh position were sensitive to Cm. Deletion
© 2002 Blackwell Science Ltd, Molecular Microbiology, 43, 1657–1669
Bacterial trafficking of integron gene cassettes 1661

Fig. 3. Alignment (A) and phylogenetic relationships (B) of the known chloramphenicol (CAT), streptogramin (SatA) and virginiamycin
acetyltransferase gene (VatB). Accession numbers follow the corresponding designations. Resistance determinants that are structured as gene
cassettes within integrons are boxed. The novel CATB9 enzyme is marked with a grey box.

© 2002 Blackwell Science Ltd, Molecular Microbiology, 43, 1657–1669


1662 D. A. Rowe-Magnus et al.

Fig. 4. Cloning of the sequential deletions of the cassettes upstream of the catB9 gene and the corresponding resistance phenotype
conferred by the various constructs. The integrase gene of the V. cholerae SI, VchintIA (thick grey arrow), and the 3.8 kb fragment spanning
the attI4 site and the first seven cassettes, including the catB9 (stripped box) gene, is shown. Sequential cassette deletions were generated
by PCR and all products were cloned into the multiple cloning site (MCS) of the plasmid pSU38. E. coli transformants were tested for their
ability to grow on media containg 25 mg ml–1 Cm. The relative hybridization positions of the primers used to create the various constructs (I4,
I4Cm1 to I4Cm6 and cat2) are marked as arrows. Symbols: Top: horizontal lines represent the PCR products obtained with the corresponding
primer pairs; –, no growth; +, growth. Bottom: black triangles, VCRs; open boxes, ORFs; the PLac promoter of pSU38 is indicated with a bent
arrow.

of the first two cassettes to create transformants that transconjugants containing the catB9 cassette in the first
harboured the catB9 gene in the sixth and fifth positions position of In3, suggested that the parent V. cholerae
relative to the PLac promoter remained sensitive to the strain 63.32 was sensitive to Cm because the catB9
antibiotic. Deletion of the third, fourth, fifth and sixth cas- gene was poorly expressed in its native seventh position
settes, which placed the catB9 gene in the fourth, third, of the SI.
second and first positions relative to the PLac promoter, To determine if V. cholerae strain 63.32 could make a
gave rise to CmR transformants. Furthermore, the growth direct switch from a Cm-sensitive to a Cm-resistant state
rate of the CmR transformants in the presence of Cm through integrase-mediated relocation of its SI gene cas-
conformed with the position of the catB9 cassette relative settes to a class 1 integron, V. cholerae strain 1275 was
to the PLac promoter, i.e. the closer the catB9 cassette submitted to intI1 induction and then plated directly on
was placed to the PLac promoter, the faster the growth rate media containing 25 mg ml–1 Cm. V. cholerae isolates that
of the transformants in the presence of Cm (data not were resistant to Cm were obtained. PCR and sequenc-
shown). These results, along with those of the E. coli ing verified that the catB9 gene had been relocated to the

© 2002 Blackwell Science Ltd, Molecular Microbiology, 43, 1657–1669


Bacterial trafficking of integron gene cassettes 1663

attI1 site of In3 in the resistant clones. These results cassettes by a MRI, including the catB9 gene cassette
confirmed that the catB9 gene cassette was phenotypi- and three other potential antibiotic resistance determi-
cally silent in its native position in the SI of V. cholerae nants, demonstrates that integrons randomly recruit cas-
strain CIP 63.32. The resulting MRI (CmR, TpR, SulR) sette-encoded functions that are later selected according
could then be readily conjugated, at high frequency to environmental conditions, as demonstrated by our dis-
(>10–2), among clinically relevant bacterial species covery of catB9 by selective constraint.
such as Salmonella typhimurium, Shigella sonnei,
Pseudomonas aeruginosa and V. cholerae strain 569B,
Clonal expansion of V. cholerae isolates harbouring the
which does not harbour the VCA0300 gene cassette
catB9 gene cassette
within its SI. The evolution of antibiotic resistance due to
the positional activation of a single traceable marker Thirty V. cholerae isolates, representing O:1, O:139 and-
allowed us to determine a frequency of recombination and non-O:1/non- O:139 serovars collected between 1937
conjugation (RCF) of 7 ¥ 10–7 for a single-copy SI cas- and 1993, were tested for the presence of the catB9 gene
sette by a MRI. These results clearly demonstrated that cassette by PCR (Table 3). In our sample pool, 22
cassette-encoded phenotypes that might be quiescent isolates were negative for the catB9 cassette, while eight
in certain genetic contexts can be presented through isolates tested positive for the catB9 cassette. Sixteen of
integrase-mediated relocation events. the 30 isolates examined were from Asia and the Indian
subcontinent, and subsequent analysis focused on this
subset as it was the most statistically complete. Interest-
Integrons randomly recruit SI cassettes
ingly, all the Asian isolates that tested negative for the
To detect any V. cholerae SI cassette relocation event catB9 gene were collected before 1961 and all of the pos-
to In3, we conducted allele-specific fusion primer PCR itive isolates were collected after 1961. Furthermore, the
directly on V. cholerae strain 1275 without further selec- data indicated that theses isolates could be divided into
tion. In this procedure, a unique fusion primer was clonal groups. For example, the O:1 isolates collected
designed to hybridize specifically to the VCR–dfrB2 cas- from this region before 1961 (569B, 62.14, 62.15, 62.16)
sette junction that would arise from an IntI1-mediated were of the same clonal origin (A.-M. Guerout, D. A.
recombination event of any V. cholerae SI cassette at Rowe-Magnus and D. Mazel, manuscript in preparation)
the attI1 site of plasmid R388. After cloning of the bulk and lacked the catB9 cassette. However, the catB9-
PCR product, we selected and sequenced the inserts of positive O:1 isolates collected from this region after 1961
10 random clones. We identified 10 different cassettes (N16961, 63.32, 68.10, 254) represented the expansion
(Table 2), nine of which had homologues in the SI of V. of a different O:1 clone that harboured the catB9 cassette.
cholerae N16961 (Heidelberg et al., 2000). In addition, the Other differences in SI order and content were consistent
bulk PCR product was subjected to nested PCR using with this observation. Two late O:139 isolates (104151,
primers specific for the gene cassettes listed in Table 1. 104152) of clonal origin also harboured the catB9 cas-
Sequencing of the amplified fragments confirmed the inte- sette. There was no apparent correlation between serovar
gration of the catB9, VCA0328, VCA0387 and VCA0473 type and the presence of the catB9 cassette, as it
gene cassettes at the attI1 site of In3. Thus, our detec- was found among O:1, O:139 and-non-O:1/non- O:139
tion of 14 independent recruitment events of SI gene serovar types.

Table 2. Cassettes recruited from the SI of V. cholerae strain 63.32 by the class I integron, In3, of R388.

V. cholerae N16961 homologue

V. cholerae CIP 63.32 cassette clone Accession no. E value Function


–77
1330 D82477 3e Chloramphenicol acetyltransferase
1335 F82451 7e–86 Probable acetyltrafsferase
1336 H82462 5e–33 Hypothetical protein
1337 H82465 5e–66 Hypothetical protein
1138 E82472 2e–89 Hypothetical protein
1339 G82462 2e–41 Hypothetical protein
1340 D82471 3e–67 Hypothetical protein
1341 AF462020 – Hypothetical protein
1342 D82474 6e–35 Hypothetical protein
1343 E82471 4e–18 Hypothetical protein
1344 H82471 4e–42 Hypothetical protein

© 2002 Blackwell Science Ltd, Molecular Microbiology, 43, 1657–1669


1664 D. A. Rowe-Magnus et al.
Table 3. V. cholerae strains examined for the catB9 gene cassette.

Strain name or CIP designation Date of isolation Place of isolation Serovar catB9 Reference

N16961 1975 Bangladesh O:1 + ATCC 39315


63.32 1961 Hong-Kong O:1 + CIP
104151 1992 Madras O:139 + CIP
104152 1993 Bangladesh O:139 + CIP
68.10 1968 Vietnam O:1 + CIP
1024 1961 Philippines ND + 930070
J. M. Fournier, IP
62.16 1959 Thailand O:1 – CIP
569B 1940 Indian O:1 – ATCC 25870
62.15 1959 Thailand O:1 – CIP
62.14 1959 Thailand O:1 – CIP
1144 1947 Egypt ND – 930051
J. M. Fournier, IP
1145 1953 India ND – 960640
J. M. Fournier, IP
1143 1959 India ND – 960652
J. M. Fournier, IP
1023 1937 Indonesia ND – 920189
J. M. Fournier, IP
723 1954 Thailand Non-O:1/O:139 – Vicente et al. (1997)
104153 1989 Bangladesh Non-O:1/O:139 – CIP
104154 1987 Bangladesh Non-O:1/O:139 + CIP

ND, not determined.

Discussion novel phenotypically silent cat gene cassette, catB9, in a


V. cholerae SI, clearly demonstrating that environmental
Bacteria have a remarkable genetic flexibility that almost conditions dictate which of the randomly recruited
immediately permits the adaptive execution of events cassettes are retained and observed within the MRIs of
resulting from alterations in their genome order and clinical isolates.
content. In this regard, the directed gene capture activity Each integron gene cassette is an individual mobile
of integrons has played a pivotal role in the evolution genetic element, and the discovery of the chromosomal
of antibiotic resistance among Gram-negative bacteria SIs shifts their present numbers from the tens to the
(Martin et al., 1990; Jones et al., 1997; Hall and Collis, thousands. To date, more than 20 SIs have been
1998; Mazel and Davies, 1999; Rowe-Magnus and Mazel, discovered among diverse proteobacterial genera, and
1999). However, recent findings significantly expand the just five Vibrio SIs contain a transferable genetic reservoir
impact of integrons on bacterial genome engineering. that is equivalent in size to a small genome (Rowe-
The paradigm for integron-mediated gene acquisition has Magnus et al., 1999; 2001; Vaisvila et al., 2001). Further-
been the class 1 integron. The common association of more, several novel integron-integrases and hundreds
class 1 integrons with mobile DNA elements such as the of unique gene cassettes have been readily amplified
Tn21 transposon family and conjugative plasmids has led from a variety of environmental samples (Nield et al.,
to the widespread occurrence of integrons among bacte- 2001; Stokes et al., 2001). By tracking the movement of
rial populations (Martinez and de la Cruz, 1988; Nesvera a single traceable marker, we were able to determine
et al., 1998; Ploy et al., 1998; Liebert et al., 1999; Naas a RCF of 7 ¥ 10–7 for a V. cholerae SI cassette. Although
et al., 1999; Poirel et al., 1999a,b; 2000; Dalsgaard et al., this rate may seem low, the potential number of inte-
2000a). The perpetual phenomenon of lateral gene trans- gron gene cassettes and the widespread presence of
fer (Jain et al., 1999) will inevitably lead to an encounter class 1 integrons in natural bacterial populations creates
between a class 1 integron and a bacterial species har- a scenario in which bacterial chromosomal super-
bouring a SI. This is supported by the recent finding of integrons represent caches of dispersable, unpredicted
class 1 integrons in many V. cholerae isolates (Dalsgaard adaptive functions. The existence of other mobile DNA
et al., 1999; 2000a,b; Falbo et al., 1999) and a new fourth elements such as transposons, conjugative plasmids,
class of resistance integron in the SXTET constin of V. phages and entire chromosomes, in Hfr strains for
cholerae (Hochhut et al., 2001). We have shown that example, along with the vast reservoirs of integron gene
class 1 integrons can randomly recruit the gene cassettes cassettes supports the concept of a single massive
harboured within SIs. By applying a selective pressure for genetic pool that is available to and shared among
the development of antibiotic resistance, we discovered a bacteria.
© 2002 Blackwell Science Ltd, Molecular Microbiology, 43, 1657–1669
Bacterial trafficking of integron gene cassettes 1665

Although the class 1 integron is the most ubiquitous identical in length and sequence to the signature attC
among multidrug-resistant bacterial populations, its activ- sites of various SIs (Rowe-Magnus et al., 2001). Nine of
ity has never been demonstrated in a bacterium other these are associated with the signature attC sites of a
than E. coli and the establishment of a functional in vitro Xanthomonas species SI (also known as XSRs for Xan-
integron system has proven difficult. This is the first thomonas species repeats) and two are associated with
demonstration that the class 1 integron is fully functional the signature attC sites of the V. cholerae SI (also known
in bacteria other than E. coli. Furthermore, the fact that as VCRs for V. cholerae repeats; Barker et al., 1994). We
the class 1 integron is active in V. cholerae indicates that found that a VCR abutted the catB9 gene. Interestingly,
integrase function is either not dependent on a co-factor the closest relatives to CATB9 (CATB6, CATB8 and
or, if a co-factor is required for integrase function, it is not CAT accession no. X82455) are also structured as inte-
specific to E. coli. Thus, MRIs have likely contributed sig- gron gene cassettes, but their associated attC sites
nificantly to the capture and dissemination of V. cholerae are strikingly different as they belong to the XSR or
SI gene cassettes, as well as those of other bacterial SIs, SPR (for Shewanella putefaciens repeats) types. Our
among bacterial populations. identification of a third VCR-associated antibiotic re-
Although most of their encoded genes have no homo- sistance gene cassette (CARB4 and dfrVI are also VCR-
logues in the database, at least some of the SI cassettes associated; Mazel et al., 1998) suggests that SIs are an
encode adaptive functions including pathogenicity and important source of the antibiotic resistance gene cas-
antibiotic resistance determinants. In V. cholerae, two settes found in MRIs.
pathogencity genes, the heat-stable toxin gene, sto The genetic organization of a MRI promotes coexpres-
(Ogawa and Takeda, 1993), and the mannose–fucose- sion of its gene cassettes from a single promoter, PC, and
resistant haemagglutinin gene, mrhA (van Dongen et al., selection for one resistance determinant often co-selects
1987), have been found to be cassette encoded. In this for the maintenance of the entire array. Studies are cur-
study we have identified the first functional antibiotic re- rently under way in our laboratory to determine if similar
sistance gene cassette, catB9, to be found in a SI. In addi- promoters reside within the attI regions of various SIs.
tion, several potential progenitor cassettes with significant Even if this is the case, it is inconceivable that a single
homology to aminoglycoside, phosphinotricin, fosfomycin promoter could drive expression of the potentially hun-
and streptothricin resistance genes are present as well. dreds of cassettes within a SI. As a case in point, the
A variety of metabolic functions whose activities are not catB9 gene cassette in the V. cholerae strain 63.32 SI was
related to antibiotic resistance or virulence have also been phenotypically silent when located in its native seventh
determined for the gene cassettes of SIs in a number of position. Sequential repositioning of the cassette closer to
bacterial species (Barker and Manning, 1997; Rowe- a strong promoter was paralleled by an increase in the
Magnus et al., 2001). As the majority of the cassettes growth rate of E. coli transformants in the presence of Cm,
examined thus far appear to be unique to the host species and integron-mediated repositioning of the catB9 cassette
(Rowe-Magnus et al., 1999; Stokes et al., 2001; Vaisvila into the first position of In3 produced CmR E. coli transcon-
et al., 2001), the reservoir of adaptive functions that can jugants and V. cholerae 1275 inductants. The identifica-
be propagated by MRIs is potentially immense. Our tion of cassettes that contain their own promoters coupled
results confirm that integrons operate as a potent general with transcriptional readthrough, the identification of cas-
gene capture system, and this activity may have a re- settes that do not code for a discernible ORF but contain
sounding impact on bacterial adaptation. identifiable –35 and –10 transcription initiation signals
A striking difference between MRIs and SIs is the nature and cassette repositioning are alternative ways in which
of the attC sites that are associated with their accumu- promoterless cassettes may be expressed.
lated gene cassettes. The attC sites associated with the Gene cassettes are probably exchanged in marine and
gene cassettes of SIs display a strikingly high degree of soil habitats and the forces governing their transfer are
species-specific sequence relatedness, whereas the attC probably highly variable. The antibiotic therapy regimes
sites of the cassettes found in MRIs share very little of clinical environments typically select for resistance to
sequence homology. Furthermore, nearly identical cas- the antibiotics being administered. We observed that the
settes can be found in the SIs of remote species but each catB9 gene cassette was present in the SIs of Asian iso-
is associated with the signature attC site of that species lates collected after 1961 but absent from isolates of the
(Rowe-Magnus et al., 1999). These observations led to same region collected prior to 1961. Several lines of evi-
the proposal that gene cassettes are assembled within dence, however, indicate that the intervention of a selec-
individual SI-harbouring bacteria and are then dissemi- tive pressure for resistance to Cm around this point in time
nated by MRIs (Rowe-Magnus et al., 2001). In support of to force V. cholerae to acquire the catB9 cassette or
this notion, to date 12 antibiotic resistance gene cassettes favouring the expansion of a clone already harbouring the
have been identified with attC sites that are virtually catB9 cassette is unlikely. First, all of the V. cholerae

© 2002 Blackwell Science Ltd, Molecular Microbiology, 43, 1657–1669


1666 D. A. Rowe-Magnus et al.

isolates that tested positive for the catB9 gene cassette to circumvent the necessary ascent and selection of com-
remained sensitive to Cm. Second, Cm has not been pensatory mutations and still allow resistance to persist in
widely used in the treatment of cholera (Kaper et al., the absence of selection. Thus, otherwise phenotypically
1995) and the presence of the catB9 gene could not be sensitive strains may still be a genetic source for the evo-
correlated with the specific use of Cm in the treatment of lution of resistance to clinically relevant antibiotics through
epidemic and pandemic cholera. Furthermore, the catB9 integrase-mediated recombination events.
cassette was variably present among the serovars of both
epidemic and non-epidemic strains of V. cholerae. Our
Experimental procedures
findings thus raise the intriguing question of why there is
a cat gene cassette in the V. cholerae SI. There is no Bacterial strains and media
reason to assume that the original function of the resist-
The following E. coli strains were used in this work:
ance gene homologues found in SIs was the defensive
DH5a (supE44DlacU169(F80lacZDM15) DArgFhsdR17recA1
inactivation of the antibiotics developed by human indus- endA1gyrA96thi-1relA1), b2150 (DdapA::(erm-pir) thrB1004,
try or medicine. Indeed, more than 10 000 different natural pro, thi, strA, hsdS, lacZ DM15 (F¢ lacZ DM15 lacIq, traD36,
antibiotics produced by ascomycetes alone have been proA +, proB +) ), TOP10 (F- mcrA D(mrr-hdsRMS-mcrBC)
catalogued (Berdy, 1995), any number of which may have F80 lacZDM15 recA1 deoR araD139 D(ara-leu) 7697 galU
been the original substrates of the SI gene cassettes. galK rpsL endA1 nupG) and UB5201 (Martinez and de la
These genes may also have had a function aside from Cruz, 1990). V. cholerae strains were provided by the Collec-
tion de l’Institut Pasteur (CIP). Antibiotics were used at the
conferring antibiotic resistance. In support of this notion,
following concentrations: nalidixic acid (Nal), 30 mg ml–1;
we have found resistance gene homologues in the SI cas- ampicillin (Ap), 100 mg ml–1; kanamycin (Kan), 25 mg ml–1; chlo-
settes of a V. metschnikovii strain isolated in 1888 (Rowe- ramphenicol (Cm), 25 mg ml–1; trimethoprim (Tp), 87 mg ml–1;
Magnus et al., 1999), 40 years before the discovery of fosfomycin (Fm), 250 mg ml–1; phosphinothricine (Pt), 50 mg
penicillin. Thus, the gene cassettes of SIs, be they silent ml–1 in Mueller–Hinton media. Susceptibility to virginiamycin
genes or pseudogenes, may provide a genetic basis for and pristinamycin was determined by a disc diffusion assay
the evolution and subsequent retention of novel antimi- with commercially available antibiotic discs (Sanofi Diagnos-
tics Pasteur, Marne-la-Coquette, France) containing 30 mg of
crobial, virulence or metabolic functions.
the antibiotic. Diaminopimelic acid (DAP) was supplemented
The integron system allows bacteria to scavenge when necessary to a final concentration of 0.3 mM. PCR-
foreign genes that may ultimately endow an adaptive amplified genes were cloned using the TA TOPO cloning kit
advantage. Likewise, as the cassettes are presumably (Invitrogen). Primers were obtained from Genset (France).
subject to episodic selection, unproductive gene cas- Sequencing was performed by MWG-Biotech AG (Germany)
settes may be eliminated through integrase-mediated or on a Pharmacia 4 ¥ 4 sequencing system.
deletion events (Collis and Hall, 1992) to reduce the
genetic burden. With respect to the evolution of antibiotic
Detection of resistance gene cassette homologues in
resistance, this may be particularly important. Several
V. cholerae strain CIP 63.32
studies have shown that, when acquiring antibiotic re-
sistance, a concomitant cost to fitness compared with To verify that the resistance gene cassette homologues iden-
susceptible bacteria may arise in resistant strains in the tified in V. cholerae strain N16961 were also present in the
absence of selective pressure (Schrag and Perrot, 1996; SI of V. cholerae CIP 63.32, specific primers VC473 (TGCTG
TTTTGATGGTTGCTATGAT), VC387 (AAGAGGACATTTTG
Levin et al., 1997; Schrag et al., 1997; Bjorkman et al.,
TGGAAATTG), VC328 (AATGGAGTTTCTGATGAAAATTA)
2000). The subsequent evolution of resistant bacteria and VC300 (TCCCTCTTGAGGCGTTTGTTATGT) were de-
in the absence of selection commonly resulted in the signed from the sequence of strain N16961 and used in a
accumulation of secondary mutations that compensated PCR reaction with VCR1 (GTCCCTCTTGAGGCGTTTGTTA)
for these costs, rather than reversion to a drug-sensitive and strain CIP 63.32 as matrix.
state. Alternatively, as we have shown for V. cholerae,
‘positional regulation’ probably governs the expression
Conjugation assays
of many cassette-encoded functions as the majority
are promoterless and their transcription will be directly Plasmid R388 (Martinez and de la Cruz, 1990) was intro-
dependent on their position relative to a sufficiently active duced into V. cholerae 63.32 by conjugation from E. coli strain
promoter (Levesque et al., 1994; Collis and Hall, 1995). b2150. The intI1 gene was amplified using the primers
IN1RBS (GAATTCGAGCTCTAACAAAGGAGCAAGCCATG
To this end, the evolution of promoterless antibiotic resis-
AAAACCGCCACTGCG) and IN1END (GGATCCATCAGG
tance gene cassettes within integron environments may
CAACGACGGGCTGCTG), digested with EcoRI and BamHI
permit bacteria to alternate between sensitive and resis- and cloned into pTRC99A (Pharmacia) digested with the
tant states to offset the fitness constraints of resistance same enzymes to create plasmid p112. p112, which contains
under non-selective conditions. This could allow bacteria the intI1 gene under the control of the isopropyl-beta-D-

© 2002 Blackwell Science Ltd, Molecular Microbiology, 43, 1657–1669


Bacterial trafficking of integron gene cassettes 1667
thiogalactopyranoside (IPTG)-inducible synthetic Ptrc pro- catB9 gene was amplified by PCR using Pfu polymerase
moter, was then introduced by electroporation (Chiang and (Promega) with the primer pair I4 (CGCGGATCCGCGAA
Mekalanos, 2000) into V. cholerae strain 63.32 (R388) to CACTTAACAAAAACTGGG) and cat2, and V. cholerae strain
create V. cholerae strain 1275. E. coli UB5201 served as the 63.32 genomic DNA as matrix. The product was digested with
recipient for conjugations with V. cholerae strain 1275. The BamHI and PstI and cloned into pSU38 digested with the
recombination and conjugation frequency (RCF) is the ratio same enzymes. Sequential deletions were cloned into the
of CmR to TpR transconjugants (average of three independent SmaI and PstI sites of pSU38 following digestion of the Pfu
trials). The precise location of the recombination event was PCR products obtained with 14Cm1 (TGTGGTCGGAGAA
established by PCR and sequencing of 10 random clones per AAATTATGA), I4Cm2 (GTTGCCAACGAGAAAAAAGTA),
test set. I4Cm3 (GTTTCTATCAAGAAAAATCATCAAA), I4Cm4 (GGT
GTTCATCAGCTATGTGAA), I4Cm5 (GAGCTTTATGGAGC
TAACTATG) or I4Cm6 (TGTGCTTTTAGGAAAATTTAAAAA
CAT cassette cloning and CAT assay
TG) and cat2 by PstI. Plasmids were transformed into E.
The CAT gene was amplified by PCR using the primers CAT1 coli TOP10 (Invitrogen) and selected on Kan plates. To
(GCGGAATTCACCATGAACTTCTTTACGTCTCCATTTTC) test the Cm phenotype, the transformants were both plated
and CAT2 (AAACTGCAGTTGTTATGTGCGTGCTTTTGA on 25 mg ml–1 Cm and inoculated into liquid culture con-
CT) from V. cholerae strain 63.32 genomic DNA. The product taining 25 mg ml–1 Cm. Growth following incubation at 37°C
was digested with EcoRI and PstI, cloned into pBAD24 was followed.
(Guzman et al., 1995) digested with the same enzymes (to
create pBADVchCAT) and transformed into DH5a. CmR was
induced by addition of arabinose to a final concentration of Nucleotide accession number
0.4%. Control strains were grown in 0.4% glucose to repress The catB9 and 1341 gene cassettes have been deposited in
expression of the CAT gene or contained pBAD24 without GenBank under the accession numbers AF462019 and
insert. The CAT activity assay was conducted as previously AF462020 respectively.
described (Gorman et al., 1982) on cell extracts from
the control E. coli strain UB5201 (R388) and from
UB5201a(R388::catB9) Acknowledgements

We dedicate this paper to the memory of Professor Maurice


Allele-specific fusion primer PCR and nested PCR Hofnung, former Chef de l’Unité PMTG, for his encourage-
Following intI1 induction, primers DFRFUS (CACAACGGCT ment, guidance and friendship. We also thank Professor
GCCTAACGCCC) and attI1.1 (GAATTCGGCTTGTTATGACT Julian Davies (The University of British Columbia) for his
GTTTTTTTGTAC) were used in a PCR reaction with V. support, Brigitte Bourachot for technical help, Jean-Michel
cholerae strain 1275 as matrix. DFRFUS is a unique fusion Foumier for V. cholerae strains, and Jean-Marc Ghigo and
primer designed to hybridize specifically to the VCR–dfrB2 Martina Ochs for critical reading of the manuscript. D.R.-M.
cassette junction that would arise from an IntI1-mediated is an EMBO and a Fondation de la Recherche Medicale
recombination event of any V. cholerae SI cassette at the attI1 (FRM) Post-doctoral fellow. This work was supported by the
site of plasmid R388. The PCR was conducted as follows: Institut Pasteur, the CNRS, the Programme de Recherche
94°C, 30 s; 60°C, 30 s; 72°C, 2 min, for 30 cycles with Pfu poly- Fondamentale en Microbiologie et Maladies Infectieuses et
merase. The PCR products were cloned using a TA cloning kit Parasitaires from the MENRT and the DGA (contrat no.
and transformed into TOP10 cells (invitrogen). Ten colonies 0134020).
were chosen at random for sequence analysis. Homologues
of the following V. cholerae strain N16961 SI cassettes
References
were identified: accession numbers D82477 (catB9), F82451,
H82462, H82465, E82472, G82462, D82471, D82474, Arakawa, Y., Murakami, M., Suzuki, K., Ito, H.,
E82471 and H82471. The primers VCR2 (Mazel et al., 1998) Wacharotayankun, R., Ohsuka, S., et al. (1995) A novel
and DFRFUS yielded no PCR product. None of the primers integron-like element carrying the metallo-beta-lactamase
alone produced a PCR product nor did either combination of gene blaIMP. Antimicrobial Agents Chemotherapy 39:
primers on uninduced cultures. 1612–1615.
Nested PCR was conducted using primers cat2, 328-1 Avila, P., and de la Cruz, F. (1988) Physical and genetic map
(ACAAATCTGCGCTCCCAACTC), 387-1 (GGGAACATGCA of the IncW plasmid R388. Plasmid 20: 155–157.
AAACCTTGCAAC) or 473-1 (CAATGCTATAAGGCGGCAC Barker, A., Clark, C.A., and Manning, P.A. (1994) Identifica-
ATC) with the primer attI1.1 and the above bulk PCR product tion of VCR, a repeated sequence associated with a
as matrix. The PCR was conducted as described above. The locus encoding a hemagglutinin in Vibrio cholerae O1.
amplified fragments were sequenced directly with the corre- J Bacteriol 176: 5450–5458.
sponding cassette-specific primers to confirm the attI site Barker, A., and Manning, P.A. (1997) VlpA of Vibrio
location of each cassette. cholerae O1: the first bacterial member of the alpha
2-microglobulin lipocalin superfamily. Microbiology 143:
Construction of sequential cassette deletion plasmids 1805–1813.
Berdy, J. (1995) Are actynomycetes exhausted as a source
The 3.8 kb region from the VchintIA gene to the end of the of secondary metabolites? In: Ninth Symposium on the

© 2002 Blackwell Science Ltd, Molecular Microbiology, 43, 1657–1669


1668 D. A. Rowe-Magnus et al.
Actynomycetes, Moscow, Russia, 10–15 July 1994. New vectors containing the arabinose PBAD promoter. J
York: Allerton Press, pp. 13–34. Bacteriol 177: 4121–4130.
Bjorkman, J., Nagaev, I., Berg, O.G., Hughes, D., and Hall, R.M. (1997) Mobile gene cassettes and integrons:
Andersson, D.I. (2000) Effects of environment on com- moving antibiotic resistance genes in gram-negative bac-
pensatory mutations to ameliorate costs of antibiotic resis- teria. Ciba Foundation Symp 207: 192–202; discussion
tance. Science 287: 1479–1482. 202–5.
Chiang, S.L., and Mekalanos, J.J. (2000) Construction of a Hall, R.M., and Collis, C.M. (1995) Mobile gene cassettes
Vibrio cholerae vaccine candidate using transposon and integrons: capture and spread of genes by site-
delivery and FLP recombinase-mediated excision. Infect specific recombination. Mol Microbiol 15: 593–600.
Immun 68: 6391–6397. Hall, R.M., and Collis, C.M. (1998) Antibiotic resistance in
Clark, C.A., Purins, L., Kaewrakon, P., Focareta, T., and gram-negative bacteria: the role of gene cassettes and
Manning, P.A. (2000) The Vibrio cholerae O1 chromoso- integrons. Drug Resistance Updates 1: 109–119.
mal integron. Microbiology 146: 2605–2612. Hall, R.M., and Stokes, H.W. (1993) Integrons: novel DNA
Collis, C.M., and Hall, R.M. (1992) Gene cassettes from the elements which capture genes by site-specific recombina-
insert region of integrons are excised as covalently closed tion. Genetica 90: 115–132.
circles. Mol Microbiol 6: 2875–2885. Heidelberg, J.F., Eisen, J.A., Nelson, W.C., Clayton, R.A.,
Collis, C.M., and Hall, R.M. (1995) Expression of antibiotic Gwinn, M.L., Dodson, R.J., et al. (2000) DNA sequence of
resistance genes in the integrated cassettes of integrons. both chromosomes of the cholera pathogen Vibrio
Antimicrob Agents Chemother 39: 155–162. cholerae. Nature 406: 477–483.
Collis, C.M., Grammaticopoulos, G., Briton, J., Stokes, H.W., Hochhut, B., Lotfi, Y., Mazel, D., Faruque, S.M., Woodgate,
and Hall, R.M. (1993) Site-specific insertion of gene cas- R., and Waldor, M.K. (2001) Molecular analysis of anti-
settes into integrons. Mol Microbiol 9: 41–52. biotic resistance gene clusters in Vibrio cholerae O139
Dalsgaard, A., Forslund, A., Petersen, A., Brown, D.J., Dias, and O1 SXT constins. Antimicrob Agents Chemother 45:
F., Monteiro, S., et al. (2000a) Class 1 integron-borne, 2991–3000.
multiple-antibiotic resistance encoded by a 150-kilobase Jain, R., Rivera, M.C., and Lake, J.A. (1999) Horizontal gene
conjugative plasmid in epidemic Vibrio cholerae O1 strains transfer among genomes: the complexity hypothesis. Proc
isolated in Guinea-Bissau. J Clin Microbiol 38: 3774–3779. Natl Acad Sci USA 96: 3801–3806.
Dalsgaard, A., Forslund, A., Tam, N.V., Vinh, D.X., and Cam, Jones, M.E., Peters, E., Weersink, A.M., Fluit, A., and
P.D. (1999) Cholera in Vietnam: changes in genotypes and Verhoef, J. (1997) Widespread occurrence of integrons
emergence of class I integrons containing aminoglycoside causing multiple antibiotic resistance in bacteria. Lancet
resistance gene cassettes in Vibrio cholerae O1 strains 349: 1742–1743.
isolated from 1979 to 1996. J Clin Microbiol 37: 734– Kaper, J.B., Morris, J.G. Jr and Levine, M.M. (1995) Cholera.
741. Clin Microbiol Rev 8: 48–86.
Dalsgaard, A., Forslund, A., Serichantalergs, O., and Levesque, C., Brassard, S., and Lapointe, J., and Roy, P.H.
Sandvang, D. (2000b) Distribution and content of class 1 (1994) Diversity and relative strength of tandem promoters
integrons in different Vibrio cholerae O-serotype strains for the antibiotic-resistance genes of several integrons.
isolated in Thailand. Antimicrob Agents Chemother 44: Gene 142: 49–54.
1315–1321. Levin, B.R., Lipsitch, M., Perrot, V., Schrag, S., Antia, R.,
Davies, J.E. (1994) Inactivation of antibiotics and the dis- Simonsen, L., Walker, N.M., and Stewart, F.M. (1997) The
semination of resistance genes. Science 264: 375–382. population genetics of antibiotic resistance. Clin Infect Dis
Davies, J.E. (1997) Origins, acquisition and dissemination of 24 (Suppl. 1): S9–S16.
antibiotic resistance determinants. Ciba Foundation Symp Liebert, C.A., Hall, R.M., and Summers, A.O. (1999) Trans-
207: 15–27. poson Tn21, flagship of the floating genome. Microbiol Mol
de la Cruz, F., and Davies, J. (2000). Horizontal gene trans- Biol Rev 63: 507–522.
fer and the origin of species: lessons from bacteria. Trends Martin, C., Timm, J., Rauzier, J., Gomez-Lus, R., Davies,
in Microbiol 8: 128–133. J., and Gicquel, B. (1990) Transposition of an antibiotic
van Dongen, W.M.A.M., van Vlerken, M.M.A., and De Graaf, resistance element in mycobacteria. Nature 345: 739–743.
F.K. (1987) Nucleotide sequence of a DNA fragment Martinez, E., and de la Cruz, F. (1988) Transposon Tn21
encoding a Vibrio cholerae haemagglutinin. Mol General encodes a RecA-independant site-specific integration
(Life Sci Adv) 6: 85–91. system. Mol Gen Genet 211: 320–325.
Falbo, V., Carattoli, A., Tosini, F., Pezzella, C., Dionisi, A.M., Martinez, E., and de la Cruz, F. (1990) Genetic elements
and Luzzi, I. (1999) Antibiotic resistance conferred by a involved in Tn21 site-specific integration, a novel mecha-
conjugative plasmid and a class I integron in Vibrio nism for the dissemination of antibiotic resistance genes.
cholerae O1 El Tor strains isolated in Albania and Italy. EMBO J 9: 1275–1281.
Antimicrob Agents Chemother 43: 693–696. Mazel, D., and Davies, J. (1999) Antibiotic resistance in
Gorman, C.M., Moffat, L.F., and Howard, B.H. (1982) Recom- microbes. Cell Mol Life Sci 56: 742–754.
binant genomes which express chloramphenicol acetyl- Mazel, D., Dychinco, B., Webb, V.A., and Davies, J. (1998)
transferase in mammalian cells. Mol Cell Biol 2: A distinctive class of integron in the Vibrio cholerae
1044–1051. genome. Science 280: 605–608.
L.M., Belin, D., Carson, M.J., and Beckwith, J. (1995) Tight Naas, T., Poirel, L., Karim, A., and Nordmann, P. (1999)
regulation, modulation, and high-level expression by Molecular characterization of In50, a class 1 integron

© 2002 Blackwell Science Ltd, Molecular Microbiology, 43, 1657–1669


Bacterial trafficking of integron gene cassettes 1669
encoding the gene for the extended-spectrum beta- Poirel, L., Le Thomas, I., Naas, T., Karim, A., and Nordmann,
lactamase VEB-1 in Pseudomonas aeruginosa. FEMS P. (2000) Biochemical sequence analyses of GES-1, a
Microbiol Lett 176: 411–419. novel class A extended-spectrum b-lactamase, and the
Naas, T., Mikami, Y., Imai, T., Poirel, L., and Nordmann, P. class 1 integron In52 from Klebsiella pneumoniae. Anti-
(2001) Characterization of In53, a class 1 plasmid- and microbial Agents Chemother 44: 622–632.
composite transposon-located integron of Escherichia coli Recchia, G.D., and Hall, R.M. (1995) Gene cassettes: a new
which carries an unusual array of gene cassettes. J class of mobile element. Microbiology 141: 3015–3027.
Bacteriol 183: 235–249. Rowe-Magnus, D.A., and Mazel, D. (1999) Resistance gene
Nesvera, J., and Hochmannova, J., and Patek, M. (1998) An capture. Curr Opin Microbiol 2: 483–488.
integron of class 1 is present on the plasmid pCG4 from Rowe-Magnus, D.A., Guerout, A.-M., and Mazel, D. (1999)
gram-positive bacterium Corynebacterium glutamicum. Super-integrons. Res Microbiol 150: 641–651.
FEMS Microbiol Lett 169: 391–395. Rowe-Magnus, D.A., Guerout, A.-M., Ploncard, P., Dychinco,
Nield, B.S., Holmes, A.J., Gillings, M.R., Recchia, G.D., B., Davies, J., and Mazel, D. (2001) The evolutionary
Mabbutt, B.C., Nevalainen, K.M., and Stokes, H.W. (2001) history of chromosomal super-integrons provides an
Recovery of new integron classes from environmental ancestry for multi-resitant integrons. Proc Natl Acad Sci
DNA. FEMS Microbiol Lett 195: 59–65. USA 98: 652–657.
Nunes-Duby, S.E., Kwon, H.J., Tirumalai, R.S., Ellenberger, Schrag, S.J., and Perrot, V. (1996) Reducing antibiotic
T., and Landy, A. (1998) Similarities and differences among resistance. Nature 381: 120–121.
105 members of the Int family of site-specific recombi- Schrag, S.J., Perrot, V., and Levin, B.R. (1997) Adaptation to
nases. Nucleic Acids Res 26: 391–406. the fitness costs of antibiotic resistance in Escherichia coli.
Ochman, H., Lawrence, J.G., and Groisman, E.A. (2000) Proc R Soc Lond B Biol Sci 264: 1287–1291.
Lateral gene transfer and the nature of bacterial evolution. Stokes, H.W., O’Gorman, D.B., Recchia, G.D., Parsekhian,
Nature 405: 299–304. M., and Hall, R.M. (1997) Structure and function of 59-base
Ogawa, A., and Takeda, T. (1993) The gene encoding the element recombination sites associated with mobile gene
heat-stable enterotoxin of Vibrio cholerae is flanked by cassettes. Mol Microbiol 26: 731–745.
123-base pair direct repeats. Microbiol Immunol 37: 607– Stokes, H.W., Holmes, A.J., Nield, B.S., Holley, M.P.,
616. Nevalainen, K.M., Mabbutt, B.C., and Gillings, M.R. (2001)
Ploy, M.C., Courvalin, P., and Lambert, T. (1998) Character- Gene cassette PCR: sequence-independent recovery of
ization of In40 of Enterobacter aerogenes BM2688, a class entire genes from environmental DNA. Appl Environ
1 integron with two new gene cassettes, cmlA2 and qacF. Microbiol 67: 5240–5246.
Antimicrobial Agents Chemother 42: 2557–2563. Sundstrom, L. (1998) The potential of integrons and
Poirel, L., Guibert, M., Bellais, S., Naas, T., and Nordmann, connected programmed rearrangements for mediating
P. (1999a) Integron- and carbenicillinase-mediated re- horizontal gene transfer. APMIS 84 (Suppl.): 37–42.
duced susceptibility to amoxicillin–clavulanic acid in iso- Sundstrom, L., Roy, P.H., and Skold, O. (1991) Site-specific
lates of multidrug-resistant Salmonella enterica serotype insertion of three structural gene cassettes in transposon
typhimurium DT104 from French patients. Antimicrob Tn7. J Bacteriol 173: 3025–3028.
Agents Chemother 43: 1098–1104. Vaisvila, R., Morgan, R.D., Posfai, J., and Raleigh, E.A.
Poirel, L., Naas, T., Guibert, M., Chaibi, E.B., Labia, R., and (2001) Discovery and distribution of super-integrons
Nordmann, P. (1999b) Molecular and biochemical charac- among pseudomonads. Mol Microbiol 42: 587–601.
terization of VEB-1, a novel class A extended-spectrum Vicente, A.C.P., Coehlo, A.M., and Salles, C.A. (1997) Detec-
beta-lactamase encoded by an Escherichia coli integron tion of Vibrio cholerae and V. mimicus heat-stable toxin
gene. Antimicrob Agents Chemother 43: 573–581. gene sequence by PCR. J Med Microbiol 46: 398–402.

© 2002 Blackwell Science Ltd, Molecular Microbiology, 43, 1657–1669


P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

Annu. Rev. Microbiol. 2000. 54:641–79


Copyright c 2000 by Annual Reviews. All rights

PATHOGENICITY ISLANDS AND THE EVOLUTION


OF MICROBES

Jörg Hacker
Institut für Molekulare Infektionsbiologie, Universität Würzburg, D-97070 Würzburg,
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

Germany; e-mail: j.hacker@mail.uni-wuerzburg.de

James B. Kaper
Center for Vaccine Development and Department of Microbiology & Immunology,
University of Maryland School of Medicine, Baltimore, Maryland 21201;
e-mail: jkaper@umaryland.edu

Key Words gene transfer, virulence, genome structure, plasmid, bacteriophage


■ Abstract Virulence factors of pathogenic bacteria (adhesins, toxins, invasins,
protein secretion systems, iron uptake systems, and others) may be encoded by partic-
ular regions of the prokaryotic genome termed pathogenicity islands. Pathogenicity
islands were first described in human pathogens of the species Escherichia coli, but
have recently been found in the genomes of various pathogens of humans, animals, and
plants. Pathogenicity islands comprise large genomic regions [10–200 kilobases (kb)
in size] that are present on the genomes of pathogenic strains but absent from the
genomes of nonpathogenic members of the same or related species. The finding that
the G+C content of pathogenicity islands often differs from that of the rest of the
genome, the presence of direct repeats at their ends, the association of pathogenicity
islands with transfer RNA genes, the presence of integrase determinants and other mo-
bility loci, and their genetic instability argue for the generation of pathogenicity islands
by horizontal gene transfer, a process that is well known to contribute to microbial evo-
lution. In this article we review these and other aspects of pathogenicity islands and
discuss the concept that they represent a subclass of genomic islands. Genomic islands
are present in the majority of genomes of pathogenic as well as nonpathogenic bac-
teria and may encode accessory functions which have been previously spread among
bacterial populations.

CONTENTS
INTRODUCTION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 642
COMMON FEATURES OF PATHOGENICITY ISLANDS . . . . . . . . . . . . . . . . . . 643
OCCURENCE OF PATHOGENICITY ISLANDS . . . . . . . . . . . . . . . . . . . . . . . . . 645
VIRULENCE-ASSOCIATED GENES ON PATHOGENICITY ISLANDS . . . . . . . . 649
Adhesins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 649
Secretion Systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 650

0066-4227/00/1001-0641$14.00 641
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

642 HACKER ¥ KAPER

Invasins, Modulins, and Effectors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 654


Toxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 656
Iron Uptake Systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 658
Other Virulence-Associated Genes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 659
REGULATION OF PATHOGENICITY ISLANDS . . . . . . . . . . . . . . . . . . . . . . . . . 661
JUNCTION SITES OF PATHOGENICITY ISLANDS . . . . . . . . . . . . . . . . . . . . . . 664
THE ROLE OF tRNA LOCI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 665
MOBILITY GENES ON PATHOGENICITY ISLANDS . . . . . . . . . . . . . . . . . . . . . 667
TRANSFER AND DELETION OF PATHOGENICITY ISLANDS . . . . . . . . . . . . . 668
CONCLUSION: Pathogenicity Islands, Genomic Islands,
and the Evolution of Microbes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 670
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

INTRODUCTION

Over the last few years, the genome sequences of >20 bacterial species have
been determined and analyzed. It became obvious that microbial genomes consist
primarily of core sequences, with a fairly homogeneous G+C content and codon
usage, which encode housekeeping functions and which carry gene clusters with
relatively low mutational capacity. These core sequences include ribosomal-RNA-
specific genes and genes that encode key metabolic proteins such as ATPases [for
a review see Hacker et al (37) and Morschhäuser et al (78)]. In addition to the
core genome, sequences were discovered that differ from the rest of the genome
in their G+C content and codon usage and that have been acquired via horizontal
gene transfer. For example, the overall amount of horizontally transferred DNA
in the Escherichia coli K-12 genome has been calculated to be ∼17% (63). These
regions, which have been designated genomic islands, encode accessory functions
such as additional metabolic activities, antibiotic resistance, or properties involved
in microbial fitness, symbiosis, or pathogenesis.
It has long been known that important pathogenicity factors can be encoded by
mobile genetic elements, which are capable of lateral gene transfer. The presence
of genes encoding diphtheria toxin of Corynebacterium diphtheriae on a lysogenic
bacteriophage was discovered nearly half a century ago, and in subsequent years
a variety of toxins have been shown to be encoded on bacteriophages, including
Shiga toxin of enterobacteria, cholera toxin of Vibrio cholerae, neurotoxins of
Clostridium botulinum, and the cytotoxin of Pseudomonas aeruginosa (11, 110).
Other examples of mobile virulence elements include the heat-stable enterotoxin
gene of E. coli, which is part of a transposon, and genes encoding a V. cholerae
hemagglutinin, which are located on an integron structure (68). In addition, impor-
tant virulence-associated genes of gram-negative pathogens (e.g. Shigella flexneri,
Salmonella enterica and Yersinia spp.) as well as of gram-positive pathogens
(e.g. Clostridium tetani and Enterococcus faecalis) are located on plasmids (33,
48, 82). Other virulence determinants are located on the chromosome, where
they are often associated in so-called virulence blocks or virulence cassettes. In
the early 1980s, it was discovered that the presence of chromosomally located
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 643

virulence-associated genes of uropathogenic E. coli (UPEC) differs among E. coli


strains (38, 66). This observation was also made for intestinal E. coli pathogens
and other pathogenic bacteria of different species and genera (69). In 1990, these
regions were termed pathogenicity islands (PAIs) (34). Although the presence of
PAIs was first described for human pathogens of the family Enterobacteriaceae, it
has become clear in recent years that animal and plant pathogens may also contain
PAIs. The majority of PAIs are located on the chromosome; however, they can
also be part of bacterial plasmids and phages (37).
This article describes the structures and functions of the various PAIs known to
date and discusses virulence functions encoded on PAIs and their impact on infec-
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

tious diseases. In addition, the mobility of PAI elements is summarized. Last, but
not least, we discuss PAIs as examples of genomic islands present in the majority of
bacterial genomes, where they act as landmarks for lateral gene transfer. The sig-
nificance of genomic islands, including PAIs, for microbial evolution is discussed.

COMMON FEATURES OF PATHOGENICITY ISLANDS

PAIs in pathogenic E. coli were the first described. It was soon discovered that
pathogenic bacteria of species other than E. coli, both gram positive and gram
negative, contain in their genomes DNA segments which share many of the below
mentioned features of PAIs. A simplified model of a bacterial PAI is shown in
Figure 1. PAIs possess most, if not all, of the characteristics listed below (36, 37).
PAIs carry genes encoding one or more virulence factors. They were first
described in human pathogens but are also present in plant pathogens such as
Pseudomonas syringae (50). One should keep in mind that in some pathogenic

Figure 1 Model of a bacterial pathogenicity island. The thin bold line represents regions of the
core genome; pathogenicity island-specific sequences are indicated. The box represent genes. The
arrows indicate the presence of direct repeats at the ends of the pathogenicity island. Abbreviations:
DR, direct repeats; int, integrase gene; vir, virulence-associated gene; mob, mobility gene; 1mob,
pseudo-mobility gene. mob genes encode integrases, transposases, or other proteins involved in
mobility of the prokaryotic genome.
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

644 HACKER ¥ KAPER

bacteria virulence factors contribute to pathogenic potential, whereas in other, non-


pathogenic bacteria the same factors may be important for survival and replication
in particular ecological niches, where they do not create any pathogenic potential
(e.g. the iron uptake system in pathogenic Yersinia species and nonpathogenic
E. coli) (88). In nonpathogenic hosts, these DNA segments may act as fitness is-
lands or ecological islands (see section on iron uptake systems below) rather than
as PAIs. A summary of important virulence features encoded by PAIs is given in
Table 1 and discussed below.
PAIs are present in the genomes of pathogenic organisms but absent from the
genomes of nonpathogenic organisms of the same or closely related species. PAIs
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

were first described as chromosomal DNA regions, but the increasing amount of
sequence data from extrachromosomal elements supports the view that PAIs may
be also part of plasmids or bacteriophage genomes.
PAIs occupy relatively large genomic regions. The majority of PAIs cover
DNA regions of ≥10–200 kb. Strains of various species, however, may also
carry insertions of small pieces of DNA which may encode virulence factors.
These DNA regions have been termed pathogenicity islets. From a heuristic point
of view, it is useful to distinguish between islands and islets to account for the

TABLE 1 Major virulence features encoded by pathogenicity islands


Virulence feature Examples

Adherence factors Diarrheagenic Escherichia coli


Uropathogenic Escherichia coli
Vibrio cholerae
Listeria spp.
Toxins Uropathogenic Escherichia coli
Staphylococcus aureus
Iron uptake systems Uropathogenic Escherichia coli
Shigella flexneri
Yersinia spp.
Invasions, modulins, effectors Diarrheagenic Escherichia coli
Salmonella spp.
Shigella spp.
Listeria spp.
Type III secretion systems Diarrheagenic Escherichia coli
Pseudomonas syringae
Erwinia spp.
Yersinia spp.
Salmonella spp.
Shigella spp.
Type IV secretion system Helicobacter pylori
Agrobacterium tumefaciens
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 645

apparent differences in size and structure. Some authors, however, prefer the term
PAI also for smaller DNA regions (28).
PAIs often consist of DNA regions that differ from the core genome in G+C
content and in different codon usage, which may reflect the generation of PAIs by
horizontal gene transfer. It should be mentioned here that differences in the G+C
contents of PAIs and the core genome will not be observed if the DNAs of the
donors and recipients have similar or identical G+C contents.
PAIs are often flanked by small directly repeated (DR) sequences. These se-
quences may be generated after integration of PAI-specific DNA regions into the
host genome via recombination.
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

PAIs are often associated with transfer RNA (tRNA) genes. tRNA loci often
act as integration sites for foreign DNA. The association of PAIs and tRNA loci
may therefore reflect the generation of PAIs by horizontal gene transfer.
PAIs often carry cryptic or functional genes encoding mobility factors such as
integrases, transposases, and insertion sequence (IS) elements or parts of these
elements. PAIs often do not represent homogeneous pieces of DNA but rather
are made up of mosaic-like structures which have been generated by a multistep
process.
PAIs often represent unstable DNA regions. Deletions of PAIs may occur via
the direct repeats (DRs) at their ends or via IS elements or other homologous
sequences located on PAIs. Additionally, a few PAIs [e.g. the high PAI (HPI)
of Yersinia pseudotuberculosis] have the capacity to move from one tRNA site to
another (7), and other PAIs may be mobilized and transmitted by bacteriophages
(e.g. those of Staphylococcus aureus and V. cholerae) (56, 64). Particular PAIs
represent integrated plasmids, conjugative transposons, or bacteriophages or parts
of such elements.

OCCURENCE OF PATHOGENICITY ISLANDS

PAIs occur in the genomes of various human, animal, and plant pathogens. A
list of PAIs described up to now (see Table 2) shows that many members of the
Enterobacteriaceae (e.g. E. coli, S. flexneri, S. enterica, and Yersinia spp.) cause
either intestinal or extraintestinal infections via virulence factors encoded on PAIs.
Enterobacteria, as well as V. cholerae, P. syringae, and others, show frequent gene
transfer via plasmids and bacteriophages. Such extrachromosomal elements in-
deed seem to represent one source of PAIs (37). Recently it was demonstrated
that the genome of the causative agent of Legionnaires’ disease, Legionella pneu-
mophila, also contains DNA segments which carry type IV secretion-specific genes
and which can be considered PAIs (96, 97).
In addition, bacteria that have the capacity to take up DNA from the environment
via natural transformation, such as Helicobacter pylori and Neisseria gonorrhoeae,
may carry PAIs (10; JP Dillard & HS Seifert, submitted for publication).
It seems, however, that naturally transformable organisms frequently introduce
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.
P1: FRD/FQU

646
August 11, 2000
P2: FQP

TABLE 2 Pathogenicity islands (PAIs) and PAI-associated genes of various pathogensa


11:13

HACKER

Associated
¥

Organism Description Functions Size (kb) Junction sequences

E. coli 536 (UPEC) PAI I536 Hemolysin 70 DR 16 bp selC


KAPER

E. coli 536 (UPEC) PAI II536 Hemolysin, P-fimbriae 190 DR 18 bp leuX


E. coli 536 (UPEC) PAI III536 S-Fimbriae 25 thrW
E. coli 536 (UPEC) PAI IV536 Yersiniabactin synthesis, uptake 45 asnT
Annual Reviews

E. coli J96 (UPEC) PAI IJ96 Hemolysin, P-fimbriae 170 pheV


E. coli J196 (UPEC) PAI IIJ96 Hemolysin, P-fimbriae, 110 DR 135 bp pheR
cytotoxic necrotizing factor 1 (CNF1)
E. coli CFT073 (UPEC) PAI ICFT073 Hemolysin, P-fimbriae 58 DR 9 bp metV
E. coli K1 kps PAI Capsule n.d. pheV
AR110-19

E. coli E2348/69 (EPEC) LEE Type III secretion, invasion 35 selC


E. coli O157:H7 (EHEC) LEE Type III secretion, invasion 43 selC
E. coli EPEC2 LEE Type III secretion, invasion 35 pheU
E. coli ETEC Tia-PAI Invasion 46 selC
Y. enterocolitica HPI Yersiniabactin synthesis, transport 43 DR 17 bp asnT
Y. pseudotuberculosis HPI Yersiniabactin synthesis, transport 36 DR 17 bp asnT, U, W
Y. pestis HPI ( pgm locus) Yersiniabactin synthesis, transport 102 IS100 asnT
Hemin uptake DR 17 bp
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.
P1: FRD/FQU
August 11, 2000

Yersinia spp. Yop virulon Type III secretion, effectors (YOPs) 47 Plasmid
Pathogenic E. coli HPI Yersiniabactin synthesis, transport 42 asnT
P2: FQP

S. flexneri SHI-1 Enterotoxin, protease 51 IS elements


11:13

S. flexneri SHI-2 Aerobactin synthesis, transport 23–30 selC


Shigella spp. Entry region Type III secretion, invasion 37 Plasmids
S. typhimurium SPI-1 Type III secretion, invasion into 40
epithelial cells, apoptosis
S. typhimurium SPI-2 Type III secretion, invasion into monocytes 40 valV
S. typhimurium SPI-3 Invasion, survival in monocytes 17 selC
S. typhimurium SPI-4 Invasion, survival in monocytes 25 Putative
Annual Reviews

tRNA gene
S. dublin SPI-5 Enteropathogenesis 7 serT
V. cholerae VPI TCP-adhesin, regulator 39.5 DR 30 bp ssrA
H. pylori cag Pai Type IV secretion, cag-antigen 40 DR 31 bp glr
AR110-19

D. nodosus vap region Vap-antigens 12 Dr 19 bp serV


D. nodosus vrl region Vrl-antigens 27
N. gonorrhoeae PAI Serum resistance, cytotoxin 60–70
L. pneumophila icm/dot region Type IV secretion ∼20
lvh region Type IV secretion 23
B. fragilis BfPAI Fragilysin, metalloprotease II 6
B. pertussis ptx-ptl locus Pertussis toxin tRNA (Asp)
P. syringae hrp cluster Type III secretion, effectors 35 Plasmid
PATHOGENICITY ISLANDS

X. campestis hrp cluster Type III secretion, effectors ∼20


(Continued )
647
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.
P1: FRD/FQU

648
August 11, 2000
P2: FQP
11:13

HACKER

TABLE 2 (Continued )
¥

Associated
Organism Description Functions Size (kb) Junction sequences
KAPER

Erwinia spp. hrp cluster Type III secretion, effectors


R. solanacearum hrp cluster Type III secretion, effectors
A. tumefaciens T-DNA Crown gall tumor induction 20 DR 25 bp Plasmid
Annual Reviews

Opine production
S. aureus SaPI1 Toxic shock syndrome toxin-1, 15.2 DR 17 bp
putative superantigen
L. monocytogenes prf vir gene cluster Phospholipases, listeriolysin 9
Metalloprotease, ActA protein regulator
AR110-19

L. ivanovii prf vir gene cluster Phospholipases, listeriolysin 9


(LIPI-1) Metalloprotease, ActA protein regulator
Internalins, Sphingomyelinase 18
LIPI-2 Internalin CD 4 tRNA Thr
inlCD-region
C. difficile Pathogenicity locus Tcd toxins 19
a
DR, direct repeat; T-DNA, transferred DNA; LEE, Locus of enterocyte effacement; Tia, toxigenic invasion locus A; HPI, high PAI; SHI, Shigella PAI; SPI, Salmonella PAI; VPI,
Vibrio PAI; BfPAI, Bacteriodes f ragilis PAI; SaPI, Staphylococcus aureus PAI; LIPI, Listeria ivanovii PAI; Tcd, Clostridium difficile toxin.
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 649

small pieces of DNA into their genomes and that the generation of PAIs repre-
sents an exceptional event. Gram-positive bacteria such as Listeria ivanovii and
S. aureus carry “classical” PAIs which exhibit the majority of the features dis-
cussed above. Other gram-positive pathogens, such as Listeria monocytogenes and
Clostridium difficile, carry on their genomes so-called virulence or pathogenicity
gene clusters, which exhibit a few features of PAIs but differ in many aspects
(23, 61, 64). It is obvious from Table 2 that the occurrence of PAIs is not restricted
to a particular group of pathogens; rather, PAIs are distributed among all groups
of pathogens. In the following sections, virulence factors encoded by PAI-specific
genes are described.
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

VIRULENCE-ASSOCIATED GENES
ON PATHOGENICITY ISLANDS
Virulence factors encoded on PAIs represent the entire spectrum of bacterial viru-
lence factors, from adhesins to toxins to host defense avoidance mechanisms. This
section briefly reviews the major classes of virulence factors found on PAIs but is
by no means an exhaustive summary.

Adhesins
PAIs located in the genomes of various species and pathotypes encode adhesins,
which mediate the capacity of microbes to attach to specific eukaryotic receptor
molecules. Thus, P fimbriae, which represent important adherence factors of
UPEC, are encoded by UPEC-specific PAIs (35). These attachment factors have
the capacity to bind to galactose–α1-4–galactose-specific receptor molecules on
uroepithelial cells. P-fimbrial genes ( pap or prs) are often linked to gene clusters
hly and cfn, respectively encoding the UPEC-specific toxins α-hemolysin and
cytotoxic necrotizing factor 1, a linkage that argues for a strong coevolution of
these factors (4). In addition, UPEC, as well as sepsis- or meningitis-causing
E. coli, have the capacity to produce S fimbriae, which bind to sialic acid-specific
receptors on uroepithelial cells and on brain cells (79). S-fimbria-specific genes
(sfa) are part of a PAI that also carries genes for the iron uptake system iro, which
was initially found in Salmonella spp. (U Dobrindt, G Gottschalk, G Blum-Oehler,
J Hacker, unpublished data). Sequence data from PAIs of different UPEC isolates
show the presence of additional genes with significant homologies to adhesin gene
clusters such as those encoding the Proteus mirabilis fimbriae (Pmf), the heat-
resistant hemagglutinin of enterotoxigenic E. coli, and a saliva-binding protein of
Streptococcus sanguis (35).
The locus of enterocyte effacement (LEE) PAI of enteropathogenic and en-
terohemorrhagic E. coli (EPEC and EHEC, respectively) encodes an important
intestinal adherence factor called intimin [reviewed elsewhere (51, 52, 81)]. In-
timin is a 94- to 97-kDa outer-membrane protein encoded by the eae gene, which
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

650 HACKER ¥ KAPER

shows similarity to the Yersinia invasin adhesin. The role of intimin in disease has
been shown in volunteer and animal studies using isogenic eae mutants. EPEC
and EHEC interact with intestinal epithelial cells in a characteristic pattern, called
attaching and effacing, in which the epithelial cell brush border is effaced and the
bacteria intimately adhere to the epithelial cell membrane. Marked cytoskeletal
changes, including accumulation of polymerized actin, are seen in the epithelial
cell directly beneath the adherent bacteria. Intimin mediates the intimate adher-
ence of the bacteria to the host cell, whereas other factors on the LEE PAI (see
below) signal cytoskeletal changes. Intimin binds to the translocated intimin re-
ceptor (Tir) protein, which is translocated from the bacterium to the host cell via a
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

type III secretion system encoded on the LEE PAI (58). Tir is also encoded on the
LEE PAI and has effector functions in addition to serving as a receptor for intimin
(see below).
The Vibrio PAI (VPI) of V. cholerae (54, 55) encodes the toxin-coregulated
pilus (TCP), a type 4 pilus that is an essential intestinal adherence factor for this
species. The importance of the TCP as an intestinal colonization factor has been
shown in volunteer and animal studies [reviewed by Kaper et al (53)], although
the TCP has never been shown to actually mediate adherence to epithelial cells.
The TCP definitely mediates interbacterial adherence, and its role in intestinal
colonization may be to increase the colonizing mass of bacteria while some other
factor directly binds the bacteria to epithelial cells. Downstream of the tcp gene
cluster on the VPI is the acf (accessory colonization factor) gene cluster, whose
products play a role in chemotaxis, thereby assisting in intestinal colonization.
Another type 4 pilus of V. cholerae is the mannose-sensitive hemagglutinin,
which is encoded by a cluster of 16 genes (msh) flanked by 7-base-pair (-bp) DR
sequences (67). The 16.7-kb msh region is inserted between the yhdA and mreB
genes, which are adjacent to each other on the E. coli chromosome. Volunteer
studies have shown that the mannose-sensitive hemagglutinin is not involved in
intestinal colonization or any other aspect of cholera (102), thus indicating that,
although this region has features of a PAI, it is not involved in pathogenicity. How-
ever, finding that the mannose-sensitive hemagglutinin is essential for formation of
biofilms on abiotic surfaces (111) led Marsh & Taylor (67) to call this an environ-
mental persistence island. Another fitness or persistence island, in V. cholerae El
Tor, encodes an exopolysaccharide (EPSETr ) that is also involved in biofilm forma-
tion (117). EPSETr also mediates the rugose colony type and chlorine resistance.
Thus, the fitness/persistence islands encoding the mannose-sensitive hemagglu-
tinin and exopolysaccharide may be essential for maintenence of V. cholerae in an
environmental reservoir between epidemics.

Secretion Systems
Five distinct mechanisms for extracellular secretion of proteins, known as types I
through V, have been described in gram-negative bacteria. Such secretion mecha-
nisms are essential for the extracellular secretion of virulence factors to the surface
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 651

of the host cell or their direct translocation into the host cell. Extracellular protein
secretion via type II, IV, and V mechanisms requires the sec general secretion
pathway, whereas proteins secreted via type I and III mechanisms do not require
the sec system. Type III and type IV systems are the mechanisms that are most
closely associated with PAIs, although types I, II, and V can also be found on PAIs.

Type III Secretion Systems


Type III secretion systems have been found on PAIs in Salmonella, Yersinia,
Shigella, EPEC, and EHEC as well as in plant pathogens such as P. syringae,
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

Erwinia spp., Xanthomonas campestris, and Ralstonia solanacearum [reviewed


by Galán & Collmer (32) and by Hueck (46)]. Type III systems are not found on
nonpathogenic members of these species. Some pathogens have two distinct type
III systems; for example, Salmonella has complete type III systems on both the
SPI-1 and SPI-2 PAIs [reviewed by Groisman et al (33); see also Galan (31)], and
Yersinia enterocolitica has a chromosomally encoded type III system in addition
to the plasmid-encoded system (39). For most of these PAIs, genes encoding the
type III secretion system and genes encoding the proteins secreted by the type III
system comprise the great majority of the genes contained on the PAI. Although
the sequence and host cell activities of the secreted proteins differ according to the
individual pathogen, the majority of the >20 proteins that comprise the type III
secretion apparatus are highly conserved among the different pathogens [reviewed
by Hueck (46)]. Greater divergence is seen among the proteins secreted by the
type III systems, which are characterized by the lack of a cleavable amino-terminal
signal peptide.
The majority of the proteins comprising the type III secretion apparatus are
located in the inner membrane, but the functions of many of the type III secretion
apparatus proteins are not known. Members of the YscN family (e.g. InvC of S.
typhimurium, Spa47 of S. flexneri, and EscN of EPEC/EHEC) are ATPases that
are believed to provide energy for the secretion/assembly process (46). Proteins
of the LcrD family (e.g. InvA of S. typhimurium, MxiA of S. flexneri, and EscV
of EPEC/EHEC) and proteins of the YscC family (e.g. InvG of S. typhimurium,
MxiD of S. flexneri, and EscC of EPEC/EHEC) are believed to form channels in
the inner and outer membranes, respectively (112). Some type III systems, such as
those of S. typhimurium (62), P. syringae (91), and EPEC (59), have filamentous
surface structures that are distinct from flagella and are presumed to function as
channels for translocation of effector proteins into the host cell. Type III systems
have been proposed to act like a needle and syringe, injecting effector proteins into
the host cell.
Thus, PAIs of several mammalian and plant pathogens encode type III secretion
systems that are essential for virulence. In many cases, the PAIs are self-contained
units containing all genes encoding secreted proteins, the secretion apparatus, the
chaperones, etc. Many PAIs also contain genes encoding regulators of the type III
secretion system, but in most, if not all, PAIs the PAI-encoded regulators are in turn
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

652 HACKER ¥ KAPER

regulated by regulatory systems encoded outside the PAI (see below). In at least
one case, the LEE PAI of EPEC, the cloned PAI confers all known PAI-associated
phenotypes on E. coli K-12 (70), but in other cases, such as the closely related
LEE of EHEC O157:H7, the cloned PAI does not confer the expected phenotype
on K-12 (25). Some proteins secreted by a type III system are encoded outside
the PAI but still use the secretion system encoded in the PAI. For example, the
SopB and SopE proteins, first identified in S. enterica serovar Dublin, are secreted
via the type III secretion system encoded on the Salmonella SPI-1 PAI that is
located at 63 min on the S. enterica serovar Typhimurium chromosome. SopB is
encoded on the SPI-5 PAI, located at 20 min on the chromosome (114), and SopE
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

is encoded on a bacteriophage inserted at 61 min (76). It is unknown whether the


sopB and sopE genes were originally located on the SPI-1 island and subsequently
relocated or they were introduced into Salmonella spp. after SPI-1 was introduced
and somehow adapted to use the preexisting type III secretion system.

Type IV Secretion Systems


Like type III systems, type IV secretion systems have also been closely as-
sociated with PAIs. Type IV systems have been found to be essential for full
virulence of a variety of pathogens, including H. pylori, Bordetella pertussis,
L. pneumophila, Agrobacterium tumefaciens, and Brucella suis [reviewed by Burns
(9)]. In H. pylori, the type IV system encoded on the Cag PAI has recently been
shown to mediate the translocation of the CagA protein to host cells (95). CagA
plays an important role in the induction of host cellular growth changes induced
by H. pylori. In B. pertussis, the type IV secretion system (also called the Ptl
transporter) is essential for secretion of pertussis toxin to mammalian cells (9). A
type IV secretion system in B. suis was recently shown to be necessary for full
virulence of this organism in an in vitro infection model (81a).
The type IV secretion system (which has also been called a type V secretion
system, a term that we reserve for autotransporting proteins as discussed below)
was first described in the plant pathogen A. tumefaciens, wherein it mediates trans-
fer of DNA into plant cells. The type IV system in this species is encoded on the
∼200-kb Ti plasmid, which is essential for crown gall tumorigenesis of higher
plants [reviewed by Winans et al (113)]. Part of the Ti plasmid, called T-DNA,
is transferred into plant cells, where it is integrated into the host genome. The
transferred Agrobacterium genes are expressed in the plant cell, which causes the
infected cells to proliferate, ultimately resulting in crown gall tumors. Approxi-
mately 20 vir genes are responsible for this transfer of T-DNA, and many of the
Vir proteins show sequence similarity to Cag proteins of H. pylori and Ptl pro-
teins of B. pertussis (e.g. VirB4 is similar to Cag3 and PtlC, VirB7 is similar to
CagT and PtlI, etc.). In A. tumefaciens, the VirB4, VirB7, VirB9, VirB10, and
VirB11 proteins assemble to form the core of the transporter while VirB2 forms
a pilus. Little information is available on the functions of the type IV proteins of
the mammalian pathogens.
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 653

In L. pneumophila, a cluster of genes called dot (107) or icm (96, 97) was shown
to be essential for intracellular replication in host cells. The proteins encoded by
these genes show sequence similarity to the VirB proteins and are also essential for
transfer of plasmid DNA from one bacterial cell to another. The role of this type IV
secretion system in the pathogenesis of disease due to L. pneumophila is assumed to
involve transfer of protein virulence factors, rather than DNA, into eukaryotic cells,
but the translocated proteins have not yet been identified. Recently another gene
cluster, specific for a second type IV secretion system termed Lvh (for Legionella
vir homologs), was detected on a putative PAI of the L. pneumophila genome; the
function of this system, however, has yet to be elucidated (97).
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

Other Secretion Systems


Type III or type IV secretion systems are invariably associated with PAIs and are
nearly always essential for full virulence of a pathogen. Other protein secretion
systems of gram-negative bacteria have also been found to be encoded on PAIs,
but the association of these other systems with PAIs is not yet as strong as it is for
type III or type IV systems. As more genomic sequences become available and
the evolution of these secretion systems is further characterized, it is possible that
most, if not all, representatives of these specialized protein secretion systems will
be recognized as being part of PAIs or other genomic islands.
The type I secretion system was first described in the E. coli α-hemolysin, which
is encoded on PAIs in most UPEC strains and on the pO157 plasmid of EHEC
O157:H7. This secretion system requires three proteins: an inner-membrane
ATPase that provides energy for the system (HlyB for E. coli hemolysin), a mem-
brane fusion protein that spans the periplasm (HlyD), and the TolC outer-membrane
protein [for a review see Dobrindt & Hacker (22)]. Other examples of proteins se-
creted by type I systems include the B. pertussis adenylate cyclase, the Pasteurella
haemolytica leukotoxin, and proteases of P. aeruginosa and Erwinia chrysanthemi.
Type II secretion systems are exemplified by the pullulanase secretion system
of Klebsiella oxytoca, which requires 14 proteins, most of which are located in
the inner membrane [reviewed by Lory (65) and by Pugsley et al (85, 86)]. The
outer-membrane protein PulD forms a pore in the outer membrane, through which
the type II-secreted proteins are believed to pass. PulD is a homolog of the YscC
family of proteins, which are involved in type III secretion, and homologs of
other components of the pullulanase system are involved in secretion of type 4
pili of V. cholerae and other organisms. Type II systems are primarily involved
in extracellular secretion of degradative enzymes, although some toxins are also
secreted via this mechanism. Proteins secreted via type II systems include cel-
lulase and pectinase enzymes of Erwinia, elastase, phospholipase C, and toxin A
of P. aeruginosa, amylase and protease enzymes of Aeromonas hydrophila, and
chitinase, protease, and cholera toxin of V. cholerae. Although the genes encoding
the type II secretion apparatus are usually clustered, the genes encoding proteins
secreted by the apparatus are frequently unlinked to these clusters and in one case
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

654 HACKER ¥ KAPER

may not even be on the same chromosome. In V. cholerae, the Eps type II secre-
tion system is encoded on the large chromosome while a protein secreted by this
system, the Hap protease, is encoded on the small chromosome (105). Type II
systems can be found on mobile genetic elements as well as on the core genomes
of nonpathogenic members of the same species. For example, the pO157 viru-
lence plasmid of EHEC encodes an apparently intact type II system (8), but the
proteins secreted by this system and the contribution of this system to virulence
of EHEC are unknown. A cryptic yet apparently intact type II system is also
encoded in the E. coli K-12 chromosome (27), but the proteins secreted by this
system and the environmental conditions necessary for this system to function are
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

not known.
The type V secretion system encompasses a group of proteins known as auto-
transporters [reviewed by Henderson et al (42)]. This system has also been called
type IV in some publications. Examples of the autotransporters include the im-
munoglobulin A (IgA) protease of N. gonorrhoeae, pertactin of B. pertussis, the
vacuolating cytotoxin (Vac) of H. pylori, SepA of S. flexneri, and EspC of EPEC.
Rather than a cluster of genes encoding a multicomponent secretion apparatus, the
secreted protein and the secretion apparatus are encoded in a single open reading
frame. The autotransporters are exported from the cytoplasm via the sec pathway
with cleavage of an amino-terminal signal peptide. The C-terminal portion of the
protein then forms a β-barrel structure which inserts into the outer membrane and
serves as a pore for the passage of the mature protein. Some autotransporters, such
as pertactin, remain attached to the bacterial cell surface, while other members of
this class, such as IgA protease, are cleaved, thereby leaving the C-terminal por-
tion in the outer membrane and releasing the mature protein into the extracellular
milieu. Examples of autotransporters which are encoded on PAIs include EspC
of EPEC (52) and the IgA protease of N. gonorrhoeae (84). The SHI-1 (formerly
she) PAI of S. flexneri contains genes for two different yet related autotransporters,
SigA and Pic (formerly ShMu), within a 12-kb region (41, 87).

Invasins, Modulins, and Effectors


Some of the best characterized PAIs provide the host bacterium with the ability
to invade epithelial cells and/or modulate host cell activities. At least five PAIs
(named SPI-1 through SPI-5; Table 2), encoding various virulence phenotypes,
including epithelial cell invasion and macrophage apoptosis (SPI-1), intracellular
proliferation and systemic spread (SPI-2), intramacrophage survival (SPI-3 and
SPI-4), and intestinal fluid secretion and inflammation (SPI-5), have been de-
scribed for Salmonella [reviewed by Groisman et al (33)]. The best-characterized
Salmonella PAI, SPI-1, encodes a type III secretion system and several proteins
that are translocated into host cells via this type III system [reviewed by Galán
(31), Galán & Collmer (32), and Groisman et al (33)]. The SipB and SipC proteins
form part of the protein translocation system, allowing translocation of effector
proteins into epithelial cells. The SipA protein binds cellular actin and inhibits
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 655

depolymerization of actin filaments, thereby promoting localized actin cytoskele-


ton reorganization. The SopE protein, which is encoded outside SPI-I but secreted
via the SPI-1 type III secretion system (see above), initiates the invasion process
by activating small GTPases such as Rac and Cdc42, thereby leading to activation
of mitogen-activated protein kinase pathways and membrane ruffling. Another
translocated protein, SptP, inactivates Rac and Cdc42, thereby restoring normal
cell architecture after bacterial internalization (30). The SPI-2 island also encodes
a type III secretion system, but the proteins secreted by this system are not as well
defined as those secreted by the SPI-1 system. One protein translocated into host
cells by the SPI-2 secretion system, SpiC, was recently shown to inhibit cellular
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

trafficking in macrophages (106).


Two PAIs located on plasmids in Shigella and Yersinia spp. also encode proteins
that mediate internalization and other host cell responses. The type III secretion
system encoded on the Shigella virulence plasmid is essential for invasion of host
epithelial cells by this pathogen (73, 82, 104). Several Ipa proteins secreted by this
type III system are essential for invasion, although the exact functions of these
proteins are unknown. IpaB and IpaC are inserted into host cell membranes (3),
and IpaC has been specifically implicated in induction of actin polymerization and
filopodia formation (103). The IpaA protein binds to the focal adhesion protein
vinculin, resulting in F-actin depolymerization (5). The Yersinia Yop virulon pro-
tects the organism from macrophages by disrupting the phagocytic and signaling
functions of these cells and ultimately inducing apoptosis (15, 16, 48). This region
encodes a type III secretion system that translocates a number of effector proteins
into host cells. The YopH protein has a tyrosine phosphatase activity, and it in-
hibits phagocytosis and disrupts focal adhesions of HeLa cells. The YopE protein,
for which no enzymatic activity has been described, also inhibits phagocytosis and
disrupts the cytoskeleton of HeLa cells. The YpkA protein of Yersinia pestis and
Y. pseudotuberculosis, also known as YopO in Y. enterocolitica, is a serine/threonine
kinase whose target protein is as yet unidentified. The YopT protein induces a cy-
totoxic effect in cells, leading to disruption of the actin filaments and alteration
of the cell cytoskeleton, while mutants defective for production of YopM exhibit
reduced virulence in the mouse by an as-yet-unknown mechanism.
The LEE PAI of EPEC and EHEC encodes a type III secretion system that is es-
sential for formation of the attaching-and-effacing histopathology seen with these
pathogens [reviewed by Frankel et al (29) and by Kaper & colleagues (52, 81)].
The EspA, EspB, and EspD proteins are secreted via the type III system and are
believed to be part of a protein translocation complex with EspA, forming a fil-
amentous structure on the surface of the bacterium, and with EspB and EspD,
perhaps forming a pore in the host cell membrane (29, 109). The Tir protein is
translocated into host cell membranes via the type III system and serves as a recep-
tor for the LEE-encoded intimin adhesin (58). Tir may also serve to nucleate host
cell actin and to transmit signals to the host cell following intimin-Tir binding.
The human pathogen L. monocytogenes and the animal pathogen L. ivanovii
have the capacity to survive and replicate intracellularly in eukaryotic cells (61).
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

656 HACKER ¥ KAPER

The internalization of L. monocytogenes as well as of L. ivanovii is triggered by


membrane proteins termed internalins (InlA and InlB). The genes encoding the L.
monocytogenes-specific large internalins InlA and InlB are located in a particular
chromosomal region, but their expression depends on the presence of the PrfA
regulator, which is part of the PAI-like Prf virulence gene cluster present in the
genomes of both species. In addition to the large internalins InlA and InlB, Listeria
strains express so-called small internalins (InlC to InlI), which are secreted into
the medium and which also contribute to internalization and host cell specificity.
The genes encoding the small internalins of L. ivanovii are part of two PAIs termed
LIPI2 and the inlCD gene cluster. Whereas the latter is only 5 kb in size and is
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

linked to a tRNA cluster, LIPI2 is 18 kb and represents an unstable DNA region


exclusively located on the L. ivanovii genome.

Toxins
Toxin Genes on Mobile Genetic Elements
Many bacterial pathogens harbor plasmids or bacteriophages which encode impor-
tant toxins. Thus, the heat-labile and heat-stable enterotoxins of enterotoxigenic
E. coli, as well as pore-forming toxins of various enterobacteria, cytolysins of
enterococci, enterotoxins of S. aureus with superantigen activity, and neurotoxins
of pathogenic clostridia and Bacillus anthracis, are plasmid encoded [for a review
see Dobrindt & Hacker (22)]. In addition, the cholera toxin genes, the genes en-
coding diphtheria toxin, the Shiga toxin genes of pathogenic enterobacteria, the
C. botulinum neurotoxin, and various enterotoxins of S. aureus and Streptococcus
pyogenes with superantigen specificity are located on phages. In addition, unortho-
dox toxins, factors which modulate host signaling or which may act as effector
molecules in bacterial host-cell interactions, are encoded by plasmids and phages.
Because so many bacterial toxin genes are located on plasmids and bacteriophages,
it is not surprising that PAIs also often carry toxin-encoding genes.

Pore-Forming Toxins
The prototypes of pore-forming PAI-encoded toxins are α-hemolysins of UPEC
termed HlyA (22). These toxins are transported by a type I secretion system and
have the capacity to lyse erythrocytes and other eukaryotic cells following inser-
tion into the eukaryotic cell membrane. The genes encoding the type I transport
system, hlyB and hlyD, as well as a the hlyC gene encoding an HlyA-modifying
enzyme, are clustered on PAIs of UPEC and, to a minor extent, on bacterial plas-
mids. On PAIs, but not on plasmids, the hly gene cluster is often colocalized with
P-fimbrial genes and loci encoding cytotoxic necrotizing factor 1. The importance
of the hemolysin for UPEC is corroborated by the fact that particular strains often
carry two PAIs with two hly determinants on their genomes. Interestingly, the large
virulence plasmid of EHEC carries a hly gene cluster similar to the PAI-located hly
determinant of UPEC (8, 35). Another example of a pore-forming toxin encoded
by a PAI or PAI-like structures is listeriolysin O of pathogenic Listeria species.
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 657

Listeriolysin O is able to lyse erythrocytes and other cells in a cholesterol-dependent


manner but is also necessary for lysis of the vacuoles of eukaryotic cells before
L. monocytogenes spreads through the cytoplasm. The gene encoding listeriolysin
O is part of the so-called PrfA virulence gene cluster of L. monocytogenes and
L. ivanovii (also termed LIPI1), which carries virulence genes and the prf-specific
regulatory region (61). The prf locus exhibits some characteristics of PAIs but is
considered a chromosomal DNA segment rather than a typical PAI.

Proteases, Lipases, and Enterotoxins


Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org

Various bacteria of pathogenic and even nonpathogenic species and subtypes pro-
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

duce enzymes with proteolytic activities, many of which play a role in pathogenic-
ity. The corresponding genes are often located on PAIs or on PAI-like structures
of gram-positive bacteria. Thus, the prf virulence gene cluster of L. monocy-
togenes and L. ivanovii (LIPI1) carries the genes for two phospholipases ( plcA
and plcB) and a locus encoding a metalloprotease (mpl). In L. ivanovii, but not in
L. monocytogenes, another PAI (termed LIPI2) encodes a sphingomyelinase
(SmcL) (61). Furthermore, enterotoxigenic Bacteroides fragilis strains produce
another metalloprotease, termed fragilysin. This enterotoxin causes fluid accumu-
lation in lamb ligated ileal loops (115). The corresponding gene, bft, is inserted
into the B. fragilis chromosome on a small (6-kb) DNA segment, termed the BfPAI,
that appears to be of plasmid origin, suggesting a potential mechanism for intro-
duction into this species (28). In addition, several other proteolytic proteins with
enterotoxic activity have been found to be encoded on PAIs. The EspC protein of
EPEC, encoded within a 15-kb PAI that is present in some but not all lineages of
EPEC, is an autotransporter that increases the short-circuit current in rat jejunal
tissue (52). Related proteins with similar functions are the EspP protein, encoded
on the EHEC virulence plasmid, and the Pic protease/mucinase of S. flexneri SHI-
1, which degrades gelatin and mucin. It is interesting that another enterotoxin
of Shigella, ShET1, which causes fluid accumulation in rabbit ileal loops, is also
encoded in the SHI1 PAI of S. flexneri by two open reading frames that are on the
DNA strand opposite that encodes the Pic protease/mucinase (26, 87).

Toxins with Other Enzymatic Activities


Many toxins have the capacity to modify host cell components such as small G
proteins or elongation factors. While some of these toxins (e.g. cholera toxin
and diphtheria toxin) are encoded by bacteriophages, others are known to be
encoded by PAIs. Thus, the gene encoding cytotoxic necrotizing factor 1 is part
of PAIs of UPEC, where it is often located next to hly and pap gene clusters.
cytotoxic necrotizing factor 1 has the capacity to modify the RhoA protein, a small
GTPase, by deamidation. Interestingly, the cnf2 gene, encoding a toxin of similar
function, is part of a plasmid of enterotoxigenic E. coli (22, 35). Furthermore,
the genes encoding the enterotoxins of C. difficile, TcdA and TcdB, which exhibit
glycosyltransferase activity and also modify GTPases, are part of the pathogenicity
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

658 HACKER ¥ KAPER

locus, a DNA segment of 19 kb of the C. difficile genome which exhibits some, but
not all, features of PAIs (23). In addition, the pertussis toxin gene cluster ptx of
B. pertussis has features of PAIs. ptx encodes a very complex toxin which ADP-
ribosylates small G proteins. Another toxin with enzymatic activity is the Shiga
toxin (encoded by stx), which specifically cleaves the 28S eukaryotic ribosomal-
RNA molecule. Whereas in pathogenic E. coli and other enterobacterial species
the stx genes are part of phage genomes, in Shigella dysenteriae the stx locus is
located on the chromosome. The stx region exhibits features of integrated phages
which in turn could be considered as PAIs or PAI-like structures (71).
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

Superantigens
Superantigens have the capacity to bind to the T-cell receptor molecule and in turn
activate these cells in a nonspecific manner. Many of these toxins are produced
by S. pyogenes and S. aureus, in which the corresponding genes are often located
on bacteriophages or plasmids. The toxic shock syndrome toxin (TSST)-1 toxin
of S. aureus, however, is part of a 15-kb PAI (SaPI) which represents a defective
bacteriophage. The SaPI can be mobilized by helper phages and transferred to
other strains (64). Similar PAIs have been found in different S. aureus isolates,
indicating that the occurrence of SaPIs is a general phenomenon in S. aureus.

Iron Uptake Systems


Survival and multiplication of microbes in certain ecological niches depend on
the ability of these organisms to scavenge essential nutrients such as iron. For
pathogenic bacteria the acquisition of iron is a prerequisite for the infectious pro-
cess, but nonpathogenic organisms also need sufficient iron ions (74). At least two
different strategies are used by bacteria to meet their iron needs: the expression of
receptors for iron carriers (e.g. heme, hemoglobin, lactoferrin, and transferrin) and
the synthesis and secretion of siderophores, which are low-molecular-weight, high-
affinity iron-binding compounds. The gene clusters for two different siderophore
systems, yersiniabactin and aerobactin, are part of PAIs, while a hemin uptake
system of enterobacteria is part of an islet.
The siderophore system of the phenolate type termed yersiniabactin (Ybt) is
encoded by the so-called HPI first described for the pathogenic Yersinia spp.
Y. pestis, Y. pseudotuberculosis, and Y. enterocolitica (7). The Ybt system, how-
ever, is also encoded by nonpathogenic E. coli, by defined E. coli pathotypes
such as enteroaggregative E. coli (94), non-O157 EHEC (57), and extraintestinal
E. coli, as well as by other enterobacteria such as Citrobacter spp. and S. enterica
serotype IIIb (94; T Ölschläger, T Zhang, E Carniel, J Heesemann, W Rabsch, H
Tschaepe, H Karch, J Hacker, unpublished data). The HPI consists of a core ele-
ment, common to all HPI-positive species, comprising 12 yersiniabactin-encoding
genes and additional loci at the right end which are specific for particular species
and pathotypes. Y. pestis and Y. pseudotuberculosis also carry hemin storage locus
genes, which are located next to the HPI sequences but which are not part of the
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 659

island. It is interesting that the HPI is located next to one of three asparagine tRNA
loci. In Y. pseudotuberculosis the island has the capacity to move from one tRNA
locus to the other (7).
Aerobactin has long been known as an important hydroxamate iron uptake
system of pathogenic and nonpathogenic E. coli, in which the corresponding gene
cluster (termed aer or iut) may be part of large virulence plasmids which also
encode the colicin ColV. In S. flexneri, however, the aer (iut) locus is part of a 23-
to 30-kb PAI which is located next to the selC locus. The island, termed Shigella
PAI 2 (SHI-2), is unstable, and in species others than S. flexneri (e.g. S. boydii and
E. coli) the aerobactin PAI seems to be located at a chromosomal site different
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

from that of selC (80, 108). The finding that the aer determinant of the Shigella
PAI is linked to sequences with homology to ColV-specific genes confirms the
view that the PAI may be generated following integration of plasmids encoding
the aerobactin system as well as colicin biosynthesis. Whereas the two siderophore
systems yersiniabactin and aerobactin are encoded by PAI genes, the hemin uptake
system chu (for E. coli hemin uptake) or shu (for Shigella hemin uptake) is encoded
by a 5-kb DNA fragment located at position 78 on the E. coli linkage map. This
region is considered an islet rather an island, but nevertheless the chu (shu) gene
products seem to be important for the pathogenesis of EHEC and Shigella (116).
Iron uptake systems are encoded by pathogenic as well as nonpathogenic mem-
bers of the same species; however, they are more prevalent in pathogenic strains.
This supports the hypothesis that iron uptake systems, as part of genomic islands in
nonpathogenic strains, may contribute to the fitness of these organisms as well as
their adaptability to particular environments, whereas in pathogenic bacteria these
systems contribute to virulence. Accordingly, in nonpathogenic strains such DNA
fragments are considered fitness islands while in pathogenic strains they represent
PAIs.

Other Virulence-Associated Genes


O Antigen Synthesis
Although most novel pathogens arising by transfer of a PAI are thought to have
emerged hundreds, thousands, or millions of years ago, one novel pathogen with
a newly acquired (or at least newly recognized) PAI has emerged within the
past decade. Historically, V. cholerae strains associated with epidemic cholera
exclusively possessed the O1 lipopolysaccharide antigen. In 1992, epidemic
V. cholerae with a different O antigen, O139, emerged in India and Bangladesh.
The O139 clone arose from an O1 El Tor strain when a 22-kb region encoding the
O1 lipopolysaccharide antigen was replaced by a 35.8-kb region containing the wbf
genes, which are responsible for synthesis of the O139 polysaccharide [reviewed
by Karaolis & Kaper (55)]. Like PAIs, the O139 wbf region has a G+C content
(average, 42%) lower than that of the genome (47.5%), it contains an IS, and it
is inserted at a specific chromosomal site (99). The substitution of the O139 wbf
genes for the O1 genes had a dramatic effect on the virulence of V. cholerae and the
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

660 HACKER ¥ KAPER

epidemiology of cholera in India and Bangladesh. Before 1992, the majority of


cholera cases were in individuals <15 years of age, because substantial immunity
is acquired by adulthood in countries in which V. cholerae is endemic. When the
O139 strain emerged, the majority of cases were in individuals >15 years old whose
immunity against O1 strains apparently did not protect against O139 strains (53).

Serum Resistance
PAI-encoded proteins have also been implicated in mediating serum resistance.
The sac-4 gene, encoding serum resistance to N. gonorrhoeae, was recently re-
ported to be contained on a PAI that is found preferentially in isolates from persons
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

with disseminated gonococcal infections rather than those from cases of uncompli-
cated gonorrhea (JP Dillard & HS Seifert, submitted for publication). This island
is not present in strain FA1090, the strain for which the N. gonorrhoeae genomic
sequence was recently obtained. The Pic protein of S. flexneri and enteroaggrega-
tive E. coli, which is encoded on the SHI-1 PAI of S. flexneri, confers serum
resistance on E. coli K-12 strains expressing this protein (41).

Immunoglobulin A Proteases
Proteases of nonenterobacterial species with homology to the degradating pro-
teinase toxins of the Pic family (see section on toxins above) have the capacity
to cleave IgA1 molecules. For the enterobacterial IgA-like protease family (Tsh,
EspC, EspP, and Pic), an IgA-degrading activity has not been demonstrated so far
(41, 42). Other pathogenic organisms, such as Haemophilus influenzae, N. gon-
orrhoeae, Neisseria meningitidis, and Streptococcus pneumoniae, synthesize IgA
proteases, while commensal strains of these species do not show this activity. The
corresponding genes, however, have not formally been described as PAIs.

Apoptosis
Several PAI-encoded proteins that are secreted via type III secretion systems
have been shown to induce apoptosis in host cells. The SipB protein, encoded on
the Salmonella SPI-1 island, and the IpaB protein, encoded on the Shigella viru-
lence plasmid, induce apoptosis in macrophages by binding to caspase-1 (43, 44).
The Y. pseudotuberculosis YopJ and Y. enterocolitica YopP proteins, which are
homologous proteins encoded on the Yersinia virulence plasmids, also induce
apoptosis in macrophages (75, 77). These Yops exhibit a high degree of similarity
to AvrRxv from the plant pathogen X. campestris. AvrRxv, which mediates a
programmed cell death pathway in plant cells, is secreted by the type III secretion
system encoded in the hrp gene cluster (92), a PAI of plant pathogens.

Capsule Synthesis
Many pathogenic bacteria have the capacity to synthesize capsules; so far, how-
ever, the genomic locations of the corresponding genetic determinants have not
been carefully described. The capsule synthesis locus of extraintestinal E. coli
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 661

represents an exception, because in one particular E. coli K1 strain the 20-kb seg-
ment could be mapped next to the pheV tRNA locus. It is still uncertain whether
the kps gene cluster itself forms a PAI as suggested recently (13). A similar ob-
servation was made for the locus encoding the exopolysaccharide PAI of a biofilm-
positive subgroup of S. epidermidis. The corresponding genes, termed ica, are also
located on a particular DNA segment of >100 kb which is absent in biofilm-
negative strains. Further studies will be neccessary to answer the question of
whether the ica region can be considered a PAI (118; I Lösner, J Hacker,
W Ziebuhr, unpublished data).
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

Plant Tumorigenesis
The Ti virulence plasmid of A. tumefaciens encodes the ability to cause crown
gall tumors in higher plants. As noted above, the Ti plasmid encodes a type IV
secretion system (encoded by the virBDE regions) that transfers one, two, or three
fragments of DNA into host plant cells [reviewed by Winans et al (113)]. The
transferred DNA, which possesses characteristics more typical of plant DNA than
of bacterial DNA, encodes proteins that produce plant hormones such as auxin
that cause neoplastic growth of transformed plant cells. Another group of proteins
encoded by transferred DNA directs the synthesis by plant cells of opines, which
are in turn used by the bacterium for nutrition. The bacterial proteins required for
the uptake, degradation, and utilization of opines are encoded by other genes on
the Ti plasmid (e.g. occ, mop, and aga).

Genes and Gene Products of Unknown Function


Among the emerging DNA sequence data for partly or entirely sequenced PAIs
of various bacteria, many intact open reading frames which may encode putative
new virulence factors have been detected. It has been suggested that a functional
analysis of these genes and their products will certainly lead to the discovery of
new virulence strategies used by PAI-positive bacterial pathogens (35, 83). Ad-
ditionally, in the genomic islands of the gram-negative pathogen Dichelobacter
nodosus have been identified the vap and vpl regions, which exhibit many features
of PAIs. The contribution of the vap and vpl regions and their products to virulence
of the bacteria, however, is not yet clear. The DNA sequences of particular open
reading frames identified on the D. nodosus genomic islands show strong homolo-
gies to toxin, adhesin, and enzyme gene clusters of other pathogenic organisms,
and it has been suggested that the corresponding products may play a role in the
pathogenesis of this important veterinary pathogen (12).

REGULATION OF PATHOGENICITY ISLANDS

PAIs frequently contain genes encoding regulators of virulence factor genes located
on the same island. However, various arrangements of regulators and regulatory
factors are possible, including PAI-encoded regulators that are specific for PAI
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

662 HACKER ¥ KAPER

genes, PAI-encoded regulators that also regulate genes located outside the PAI,
and regulators encoded outside the PAI that regulate genes encoded on the PAI.
The last class includes regulators, present only in pathogens, which regulate
primarily virulence factors genes as well as regulators, present in both pathogenic
and nonpathogenic members of the species, that may also regulate housekeep-
ing genes in addition to virulence factors. The best-characterized regulatory sys-
tems of PAIs feature a regulatory cascade in which PAI-encoded regulators of
PAI-encoded virulence genes are themselves regulated by systems encoded out-
side the PAI.
Two major classes of regulators that are encoded on PAIs or that specifically
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

regulate PAI genes are AraC-like proteins and two-component response regulators.
Other classes of regulators include alternate sigma factors and histone-like proteins.
A few of the better-characterized PAI regulators are reviewed to illustrate the
spectrum of regulatory systems, but for many PAIs, little information concerning
regulation is currently available.
The VPI of V. cholerae contains at least three regulatory genes, toxT, tcpP, and
tcpH. ToxT is a member of the AraC family of transcriptional activators (21) and
activates transcription of the tcp genes, which code for the major virulence factor
encoded on the VPI, TCP. ToxT also regulates the ctx genes, encoding cholera
toxin, which are located on a filamentous phage inserted outside the VPI. ToxT
is itself regulated by TcpP and TcpH, which are cytoplasmic membrane proteins
encoded on the VPI (40). Transcription of toxT is also regulated by the ToxR and
ToxS proteins, which are encoded outside the VPI. ToxR and ToxS are present
in nonpathogenic strains of V. cholerae, as well as in other Vibrio spp., and they
regulate the OmpU and OmpT outer-membrane proteins of V. cholerae, which
are the major porins of this species. Thus, the ToxRS regulon, which controls
expression of housekeeping genes in V. cholerae, has been adapted by the toxT
regulator encoded on the VPI. [Expression of ctx and tcp is also regulated by other
factors encoded outside the VPI, such as cAMP receptor protein (CRP) (98)]. It is
interesting that TCP and ToxRS are encoded on the larger of the two V. cholerae
O1 chromosomes, whereas in the classical biotype, ctx genes are contained on both
the larger and the smaller chromosomes (105), an unusual example of prokaryotic
trans-chromosomal regulation.
Another well-characterized example of PAI genes regulated by proteins en-
coded within and outside of the PAI is the SPI-1 island of Salmonella [reviewed
by Cotter & Miller (17), Groisman et al (33), and Hueck (46)]. The HilA and
InvF proteins are encoded on SPI-1 and belong to the OmpR/ToxR and AraC
families of transcriptional activators, respectively. HilA regulates InvF, which in
turn regulates expression of the sip genes, encoding proteins secreted by the type
III secretion system, located on SPI-1 (19). HilA also directly regulates expres-
sion of the components of the type III system apparatus. The hilA gene is itself
regulated by proteins encoded both inside and outside SPI-1. The HilC and HilD
proteins, encoded on SPI-1, are members of the AraC family of regulators that
derepress expression of hilA (93). The PhoP and SirA proteins are encoded outside
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 663

SPI-1 and also modulate expression of hilA. PhoP is the response regulator of the
PhoP/PhoQ two-component regulatory system which responds to environmental
Mg2+ ion concentrations and acts as a negative regulator of HilA. SirA is the
response protein of another two-component system for which the cognate sensor
protein has not yet been identified. In contrast to the negative effect of PhoP, SirA
acts as a positive regulator of HilA expression. Environmental signals such as
pH, osmolarity, oxygen, and Mg2+ concentration are detected by such global reg-
ulatory systems, which then regulate expression of the SPI-1-encoded virulence
factors through HilA, supercoiling, or other mechanisms which have yet to be
characterized (17, 33).
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

The plasmid-encoded Yop virulon of pathogenic Yersinia spp. is regulated by


a global regulator called VirF (LcrF in Y. pestis and Y. pseudotuberculosis), which
belongs to the AraC family [reviewed by Cornelis et al (16)]. VirF directly binds
to the promoter regions of the several operons in the Yop virulon and is itself
strongly regulated by temperature, being expressed at 37◦ C but not at 26◦ C. This
temperature regulation involves a chromosomally encoded protein called YmoA
which resembles histone-like proteins and acts as a negative regulator of virF.
An additional level of control may involve feedback inhibition by a repressor(s)
(LrcQ in Y. pseudotuberculosis and YscM1 and YscM2 in Y. enterocolitica) that
is normally expelled via the type III secretion system and is similar to the secreted
anti-sigma factor involved in regulation of flagellum synthesis. In addition, another
regulator of pathogenic yersiniae, YbtA, is encoded by the HPI. YbtA belongs to
the AraC class of regulatory elements and seems to be involved in the expression
of both HPI-specific genes and determinants located outside the HPI (88).
The LEE PAIs of EPEC and EHEC encode a transcriptional activator of the
H-NS family called Ler which controls expression of the type III secretion system
and the Tir/intimin receptor/adhesin (72). Ler also controls expression of fimbriae
encoded outside the LEE (24) and is itself regulated by integration host factor (IHF)
and quorum sensing (100). The quorum-sensing regulatory system is encoded in
pathogenic as well as nonpathogenic E. coli strains, but the only E. coli genes so
far known to be regulated by quorum sensing are the LEE genes. EHEC is notable
for the low infectious dose required for disease (∼100 organisms), and it has been
proposed that the quorum-sensing system of EHEC allows the organism, in the
presence of high concentrations of coliforms in the colon, to induce expression of
the intimin and Tir proteins necessary for intestinal colonization (100). Regulation
of the EPEC LEE is not identical to that of the EHEC LEE, and variations include
the presence of an AraC homolog (PerA), encoded on the large virulence plasmid
of EPEC, which regulates expression of the EPEC, but not the EHEC, Ler and the
transcription of some LEE genes of EHEC, but not EPEC, by the RpoS alternate
sigma factor (100). UPEC strains carry fimbrial adhesin determinants that consist
of structural genes and two regulatory loci, which in the case of P fimbriae were
termed PapB and PapC. The Prf fimbrial operon located on PAI II of strain 536
also encodes such regulators which activate the S-fimbrial determinant (sfa) in
addition to the P-fimbrial genes. The sfa determinant is located on another PAI.
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

664 HACKER ¥ KAPER

The process in which one group of PAI-located genes is regulated by regulators


encoded by another PAI has been termed cross-talk activation (79).
An alternate sigma factor is also involved in regulation of the hrp gene cluster in
the plant pathogen P. syringae which appears to be the plant pathogen equivalent of
the PAIs found in mammalian pathogens [reviewed by Hutcheson (47)]. The hrp
cluster encodes a type III secretion system that is regulated by four proteins encoded
on the hrp PAI. HrpL is an alternate sigma factor that is essential for transcription
of all genes of the hrp regulon except hrpR, hrpS, and hrpL. The HrpL sigma factor
is similar to σ 28, which controls expression of genes involved in biosynthesis of
flagella. Transcription of hrpL itself is dependent on the σ 54 alternate sigma factor
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

and is positively regulated by the HrpR and HrpS proteins. The HrpR and HrpS
proteins both exhibit similarity to response regulators of two-component systems
such as NtrC but they do not appear to be typical two-component regulators,
because they lack the N-terminal domain that functions in typical two-component
regulators to modulate activity through phosphorylation. A fourth protein, HrpV,
negatively regulates genes in the hrp regulon by an unknown method, perhaps via
HrpR and HrpS. Homologs of the regulatory components HrpL, HrpS, and HrpV
have also been implicated in regulation of type III secretion systems on PAIs in
Erwinia strains that are pathogenic for plants.
The examples presented above represent only a sampling of the various regu-
lators and regulatory mechanisms found in PAIs. Similar regulatory mechanisms
are also seen with genes that are not encoded in PAIs. However, one recently
described regulatory mechanism, involving translation of rare tRNAs, may be
specific for regulation of genes encoded on PAIs. This mechanism, defined by the
so-called minor codon hypothesis, is described below in the section on the role of
tRNAs.

JUNCTION SITES OF PATHOGENICITY ISLANDS

PAIs represent distinct DNA segments which differ from the core genome by
several features (see above). Therefore, the boundary regions of the majority of
PAIs have a specific DNA composition, preferentially of DR DNA sequences.
As indicated in Figure 1, one of the two PAI junction sites is very often part of
the 30 end of tRNA genes or of similar gene loci encoding the small regulatory
RNAs (e.g. ssrA for D. nodosus) (12) or the gene glr, encoding glutamate racemase
(e.g. Cag PAI of H. pylori) (10). The length of the DRs ranges from 9 bp (in UPEC
CFT073) to 135 bp (PAI in UPEC J96 and LEE PAI in particular EHEC strains).
In most cases the DRs comprise 16 to 20 bp (35).
The DR DNA sequences were probably generated following insertion of pre-
PAI elements such as bacteriophages or plasmids into the core genomes of the host
organisms. It should be mentioned here that the 30 ends of tRNA loci very often
act as attachment sites for the integration of bacteriophages. Thus, the strong as-
sociation between tRNA genes and PAIs may argue for a development of PAIs
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 665

from former bacteriophages (89; see also section on mobility genes on PAIs
below). In addition, the DR segments may act as target sequences for the ac-
tion of integrases/excisionases in deletion processes, since many PAIs have the
tendency to delete (see section on transfer and deletion of PAIs below). In most
cases these deletion processes are RecA independent, and it seems that particular
integrases, preferentially those encoded by PAIs, may be involved in the deletion
processes. IS elements, in contrast, flank PAIs in very few cases (e.g. the PAI-like
element of the stx gene in S. dysenteriae) (71). IS elements or portions thereof
are, however, very often part of PAI segments. This is also true for Rhs sequences
and other repeated DNA segments which do not form junction sites for PAIs but
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

often act as targets for partial deletions of PAI-specific sequences (7).

THE ROLE OF tRNA LOCI

About 75% of the PAIs identified so far are associated with tRNA loci (see Table 2).
It has been known for many years that tRNA genes may act as landmarks for
the integration of foreign DNA, either of plasmids and phages or, in the case
of eukaryotes, of retroviruses (45, 89). It is interesting that extrachromosomal
genetic elements themselves often carry tRNA genes or parts of them. This is true
for plasmids of streptomycetes but also for bacteriophages of enterobacteria, such
as the T4 phage or the phage carrying Shiga toxin genes. In addition, in eukaryotes,
mitochondrial as well as plastidic DNA molecules harbor tRNA loci. It is therefore
suggested that the extrachromosomal elements integrate into the chromosomes of
their host organisms by homologous recombination between tRNA genes of the
chromosome and their extrachromosomal counterparts, thereby keeping the tRNA
loci intact. It seems that the 30 ends of the tRNA genes may preferentially act as
targets for integration. DNA sequence data for many PAIs and their associated
tRNA genes show that overlaps of 15–25 nucleotides exist between the 30 ends of
the tRNA loci and PAI-specific sequences (45).
Although in principle many tRNA genes can be used as integration sites for PAIs
and other extrachromosomal elements, in praxi particular preferences exist. The
tRNA genes frequently associated with PAIs (see Table 2) also quite often harbor
attachment sites for bacteriophages, indicative of the development of PAIs from
former bacteriophage genomes. One of the tRNA loci most frequently associated
with PAIs is the selC gene, encoding the selenocysteine-specific tRNAsec. In
enterobacteria the bacteriophage φR73 and five different PAI elements can be
located next to selC (35). Three different E. coli-specific PAIs, leading to different
E. coli pathotypes, as well as PAIs of S. enterica and S. flexneri are associated with
the selC locus. In addition, the two identical phenylalanine-specific tRNA genes
pheV and pheU (also termed pheR) often act as targets for PAI formation. The idea
that PAIs may represent a particular subtype of genomic island is corroborated by
the fact that not only three different PAIs but also two genomic islands, encoding
an alternative sucrose uptake system in S. enterica and the nitrogenase system in
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

666 HACKER ¥ KAPER

Mesorhizobium loti, are located next to the pheR and pheU loci (37; see Tables
2 and 3). In addition, tRNA-associated PAIs also have the capacity to be deleted
from the genomes of their respective pathogens, leading to a truncation of the
tRNA-specific genes. In the case of UPEC strain 536, the tRNA genes leuX and
selC, located next to PAI I and PAI II, became truncated at their 30 ends after
deletion of the two PAIs, which consequently leads to tRNA-specific mutations. It
therefore seems that only tRNA genes with another identical locus in the genome
(e.g. phenylalanine-specific tRNAs), genes which encode tRNAs with wobble
capacity (e.g. leuX ), or nonessential tRNAs (e.g. selC ) are frequently used as
integration sites for PAIs.
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

Many authors have suggested that the presence of particular tRNAs that are
less frequently used than others (so-called minor tRNAs) may have a modulatory
effect on the translational efficiency of particular target genes (the minor codon
hypothesis). The evolutionary advantage of extrachromosomal genetic elements
carrying additional tRNA genes may be a more effective expression of genes with
a larger amount of the corresponding codon located on the respective element.
Thus, the genes present on PAI II of UPEC strain 536 that are located next to
the leuX tRNA have a significantly larger number of leuX-specific codons than
genes located on the rest of the core genome. Therefore, it was not surprising
that leuX mutants were less virulent than their leuX-positive counterparts, because
many genes located on PAI II, such as that encoding α hemolysin, and also other
genes were less frequently expressed (90). For type I fimbriae, whose expression

TABLE 3 Examples of genomic islands


Organism Property Type of island Genetic feature Size (kb)

Mesorhizobium loti Nitrogen fixation Symbiosis Integrated plasmid 500


Pseudomonas putida Chlorocatechol Degradation Integrated plasmid 105
degradation
Salmonella senftenberg Sucrose uptake Metabolism Conjugative 100
transposon
Staphylococcus aureus MecA protein Resistance Location on 52
chromosome
Salmonella Multiresistance Resistance Location on 14
typhimurium DT 104 chromosome
Various bacteria Type III secretion Secretion Location on Variable
Type IV secretion chromosome
or plasmid
Various bacteria Iron uptake Fitness Location on Variable
chromosome
or plasmid
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 667

is also affected by the presence of leuX, it was shown that the translation of the
fimB-specific positive regulator gene, which harbors an unusually large number of
leuX codons, is strongly affected by the presence of the leuX-specific tRNA5Leu.
It is interesting that the leuX gene itself is regulated by global regulators such as
the alternative sigma factors RpoS and RpoH, providing additional support for
the minor codon hypothesis (U Dobrindt & J Hacker, unpublished data). Besides
leuX, other minor tRNA genes, such as argU and leuZ, influence the expression of
particular virulence-associated genes such as the fimbrial genes of S. enterica.
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

MOBILITY GENES ON PATHOGENICITY ISLANDS


A variety of mobility genes are found on PAIs, particularly genes encoding phage
integrases and ISs/transposons. However, the presence and functionality of these
mobility genes can differ greatly among PAIs, from genes that encode intact and
functional integrases to mutated genes that encode nonfunctional integrases to PAIs
that contain no apparent integrase genes. These mobility genes and the protein
products they encode can lead to the formation, integration, deletion, and mobility
of PAIs as described below in the section on transfer and deletion of PAIs.
Several PAIs contain sequences that show homology to genes encoding in-
tegrases of bacteriophage P4 and retronphage φ73. P4 integrase sequences are
present in the HPI of Yersinia, a PAI which is also found in enteroaggregative
E. coli (94), some Shiga toxin-producing E. coli (STEC) serotypes (57), and UPEC
(PAI IV536) (94). This integrase appears to be intact and functional in the majority
of strains, in contrast to the P4 integrases encoded in PAI II536 of UPEC and in
E. coli K-12, both of which appear to be defective due to the presence of premature
stop codons. Integrases that are more closely related to the integrase of φ73 are
found in PAI I536 of UPEC, the SHI-2 PAI of Shigella species (108), and in the vap
(for virulence-associated protein) regions of D. nodosus (12, 35). The integrase
present in the V. cholerae VPI most closely resembles that of the cryptic bacterio-
phage CP4-57 (53% amino acid identity) but also exhibits substantial homology
with the P4 integrase (32% identity) and the D. nodosus vap-encoded integrase
(34% identity). The substantial homology among integrases from different genera
suggests that these sequences could be transferred among a wide range of bacterial
species.
The integrases and other bacteriophage proteins encoded on PAIs are usually
assumed to have played a role in the original mobilization of the PAI, but there
are some phages that appear to have been inserted into the PAI after the origi-
nal mobilization of the PAI into the chromosome. One such phage is the 933L
prophage of the EHEC O157:H7 LEE, which is absent from the closely related
LEE of EPEC E2348/69 (83). Similar phages may have nevertheless played a role
in the mobilization of other PAIs, because an integrase gene nearly identical to
that of 933L is also present in the Shigella SHI-2 PAI (108).
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

668 HACKER ¥ KAPER

Numerous IS elements and transposon sequences can be found in PAIs. In


some cases, such elements may have played a role in the original mobilization
or subsequent rearrangement of the PAI, or they may have inserted afterward and
played little or no role in PAI formation. The insertion of IS605 into the cag PAI of
H. pylori appears to have played no role in the original mobilization of this PAI
but has played a major role in its subsequent evolution (18). The SHI-2 PAI of
S. flexneri, which encodes the aerobactin iron transport system, contains numer-
ous partial and complete IS elements, including IS3, IS629, IS600, IS2, and IS1
(80, 108). Comparison of SHI-2 islands from S. flexneri strains SA100 (serotype
2a) and M90T (serotype 5a) revealed essentially identical insertion sites, aerobactin
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

genes, and IS elements on the left side of the PAI but substantial differences in
IS elements and other sequences on the right side of the island. The SHI-2 PAIs
in Shigella sonnei and Shigella boydii have the same insertion site and aerobactin
genes but exhibit major differences in the IS elements on their left sides.
In addition to mediating rearrangements of PAIs over long time frames (macro-
evolution), IS elements can also mediate important changes over very short time
frames, as is seen in the phase variation of the polysaccharide intercellular adhesin
of Staphylococcus epidermidis, which is a microevolutionary process. Alternating
insertion and excision of IS256 into different sites of the S. epidermidis ica gene
cluster controls expression of the polysaccharide intercellular adhesin, which is a
major component of staphylococcal biofilms (118).

TRANSFER AND DELETION


OF PATHOGENICITY ISLANDS
The VPI of V. cholerae was recently shown to be mobilizable from one strain of
V. cholerae to another by transduction (56). This island encodes several proteins
whose predicted sequences are similar to those of proteins of bacteriophages or
eukaryotic viruses as well as a protein that is highly homologous to the E. coli
cryptic prophage (CP4-57) integrase (60). Furthermore, the VPI is flanked by
20-bp DR sequences that resemble phage att sites (56). Culture supernatants of
VPI-positive V. cholerae strains contained sequences present on the VPI, as shown
by polymerase chain reaction, but did not contain sequences of other chromosomal
genes located outside the VPI (56). Cell-free phage preparations from a strain
marked by insertion of a kanamycin resistance gene into the VPI were found to be
capable of transferring the marked VPI into a previously VPI-negative recipient
strain. These results suggest that transfer of the VPI via phage transduction could
play an important role in the evolution of pathogenic V. cholerae strains from
nonpathogenic strains.
The staphyloccocal PAIs (SaPIs) encode the TSST, a property which has been
previously shown to be transferable. It became clear from studies by Novick &
coworkers that the SaPI is part of a defective bacteriophage of 15 kb which has the
capacity to be excised and circulated by helper phages such as φ13 and φ80α (64).
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 669

After excision, the islands are transduced to other staphylococcal strains with high
frequencies. Thus, the SaPI belongs to the group of mobile PAIs that may have
been derived from bacteriophages.
Phage transfer has been implicated in the transfer of other PAIs and PAI-like
structures. The genes responsible for the serotype 1a O antigen of S. flexneri
are encoded on a 5.8-kb region that is flanked by 45-bp att sequences and IS
elements (1). The overall G+C content of this area (40%) is significantly below
the 50% Shigella genome content. On one side of this region is a gene whose
predicted protein product shows significant homology to integrases of P22 and
other bacteriophages. This region is inserted at the thrW tRNA gene, and in fact
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

the 30 end of this gene is identical to the attR sequence. One final example is the
SopE protein of Salmonella, which is secreted via the type III secretion system
encoded on the SPI-1 PAI. The SopE protein is not encoded on SPI-I but instead
is encoded within a temperate bacteriophage belonging to the P2 family (76).
Certain PAIs do not have the capacity to move from one bacterial strain to
another but are, however, able to jump from one site to another in the genome of
a bacterial pathogen. The HPIs of pathogenic enterobacteria, encoding the iron
uptake system yersiniabactin in Y. pseudotuberculosis and Y. pestis, carry intact
integrase genes and are flanked by DRs of 17 kb (88). The HPIs in Yersinia are
usually located in the asparagine-specific tRNA gene asnT. In particular strains of
Y. pseudotuberculosis, however, the HPI first detected in asnT, over a short time
period, has also been found to be located in two other asparagine-specific tRNA
genes, asnW and asnV, indicating a transfer of the element from one target site to
another (7). It has been suggested that such mobility events of PAIs may also occur
during infections and may represent examples of PAI-induced microevolutionary
processes.
In addition, certain PAIs have a tendency for deletion of particular PAI-specific
sequences or of the whole genetic element. On one hand, such deletions may occur
over longer time intervals in order to optimize the structure of the PAI elements and
to reduce the genetic burden by eliminating genes whose products are not further
used (see also below and Figure 2). On the other hand, in contrast to the processes
of macroevolution, in many organisms PAIs show deletions in a short time range.
Such processes of microevolution have been detected for the Cag PAI of H. pylori,
which has a tendency to decrease in size over short time intervals (10). In addition,
the HPI and also the hemin storage locus of Y. pestis carry many Rhs elements
which may act as landmarks for frequent deletion processes (7). Furthermore,
in other organisms, such as UPEC, whole PAI elements can be deleted. In these
cases recombinases/integrases located on the corresponding PAI may be involved
in the deletion processes (4, 35). For PAI I and PAI II of E. coli 536, however,
the PAI-encoded recombinases are not intact, thereby suggesting that integrases
of other PAIs or encoded by the rest of the genome are probably involved in the
deletion processes. It is suggested that the deletion processes may play a role in the
adaptation of pathogenic microbes during certain stages of infection. Therefore,
genetic flexibility of pathogenic microbes may create selective advantages over
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

670 HACKER ¥ KAPER


Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

Figure 2 Evolutional stages of pathogenicity island formation. The vertical line represents
the evolutionary progress; the horizontal line represents a time scale. The thin line represents
parts of the core genome; the double line represents pathogenicity island-specific DNA. The
boxes indicate genes, and the asterisks indicate mutations. Abbreviations: tRNA, transfer RNA
genes; int, integrase gene; vir, virulence-associated gene; mob, mobility gene (e.g. gene encoding
transposase).

other, less-flexible organisms and may finally result in proper replication in host
organisms or other ecological niches.

CONCLUSION: Pathogenicity Islands, Genomic Islands,


and the Evolution of Microbes
The key processes of Darwinian evolution can be described by the terms genetic
variability, phenotype formation, and natural selection. There is no doubt that the
permanent generation of new genetic variants represents the main prerequisite for
evolution. The term macroevolution describes evolutionary processes which oc-
cur within a longer period of time and which lead to the formation of new species
or pathotypes (78). In contrast, microevolution, which generates new variants of
given species or pathotypes, takes only days or weeks. Both processes, macro- and
microevolution, are also involved in the generation and adaptation of pathogenic
microbes.
The majority of pathogenic bacteria show evidence of horizontal gene transfer,
which together with the generation of point mutations and genetic rearrangements
represents one major source for the creation of new genetic variants. The analysis
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 671

of the structure of PAIs revealed that these genetic elements have been generated
after lateral gene transfer processes. Thus, PAIs contributed to the development of
pathogenic variants, preferentially in macroevolution. The ongoing processes of
rearrangements, deletions, and transfer of PAIs, however, also have a strong impact
on microevolution and adaptation of pathogenic microbes during acute infectious
processes.
As indicated in Figure 2, PAIs can be considered genetic fossils owing to their
particular stages in the evolutionary process of pathogens. Integrated newly ac-
quired DNAs seem to represent, from the evolutionary point of view, the youngest
types of PAIs. The complete or incomplete phages which are identical to S. aureus
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

PAIs and the V. cholerae VPI encoding the TCP may represent PAIs at the begin-
ning of their career as evolutionary chronometers (56, 64). If a PAI significantly
contributes to the fitness and probably also the pathogenic potency of its host
organism the mobility genes involved in transfer, deletion, or excision will be
inactivated or even deleted. In other words, selection for stability and homing of
the formerly acquired DNA takes place. The HPIs of Yersinia, the PAIs of UPEC,
and the Cag island of H. pylori may represent such “middle-aged,” adolescent-
type PAIs (18, 88). Over time, for example periods of thousands of years, the sizes
of successful PAIs tend to be reduced to include just the important genes required
for their key functions, and the PAIs become more and more condensed (e.g. the
LEE island of EPEC) (52). Thus, the Salmonella SPI-1, which was introduced
into the genome of the common ancestor of S. enterica and Salmonella bongori
100 million years ago, is already part of the core genome of both organisms (33).
It is our assumption that there exist genetic segments which are former PAI regions
but already have been adapted to the core genome of the host and which are no
longer detectable as PAIs. In these cases the evolutionary progress has terminated.
Over the past few years, research on PAIs has attracted much attention. How-
ever, important questions concerning the development and selection of PAIs remain
unanswered. One major question is: Where do the PAI-specific DNA segments
come from? From our point of view it is very difficult to answer this question.
The overall number of prokaryotes in the soils, fresh waters, and oceans has been
estimated to comprise 150,000–200,000 different species; however, only ∼4000
have been described so far. The majority of the bacterial species are not culturable
in the laboratory, and therefore analyses of their genomes have been largely im-
possible. It has been suggested that the enormous amount of genetic information
present in the genomes of species not yet described may be a source for many ge-
netic elements, including PAIs. In the case of the Yersinia HPI it is suggested that
members of the Pseudomonadae may be donors for this DNA; however, this has
not yet been verified. The further development of microbial ecology will certainly
open new avenues to approach the important question of the source of PAI-specific
DNA in the future.
Another question is: What are the selective forces for the development of PAIs?
Pathogenic organisms as well as nonpathogenic microbes have to overcome envi-
ronmental restrictions for survival and replication and have to compete successfully
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

672 HACKER ¥ KAPER

in their ecological niches. These ecological processes, more so than the processes
of disease formation, are of evolutionary relevance. The occurrence of infectious
diseases does not necessarily contribute to the evolutionary fitness of microbes.
It is therefore suggested that from an evolutionary view point, many PAIs rep-
resent fitness islands because they were selected in the natural environment of
microbes to stimulate their survival and replication. Thus, P fimbriae of UPEC,
whose genes are components of PAIs, contribute to adherence of these microbes
in the gut (14). Iron uptake systems, of course, are necessary for the survival
of microbes in various niches. As the ecological niches and reservoirs of many
pathogens (e.g. pathogenic Shigella) are not yet known, it is not possible now to
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

fully answer the question on the selective pressures leading to PAI generation.
The third question of interest is: What are the host equivalents of PAIs? The
acquisition of new genetic material by the microbes, of course, may generate new
phenotypes which may interact with host structures. If the acquisition and genera-
tion of PAIs were to generate more-aggressive microbes, they would destroy their
hosts and would therefore be unsuccessful in evolutionary terms. On the other
hand, the generation of new relevant microbial phenotypes for host interaction
should also select for new host structures such as receptor variants or alterations
in signal transduction pathways. Such changes in host genes will be important
for overcoming hyperpathogenicity and for development of new strategies for an
appeasement of hosts and microbes. These host strategies are largely unknown
to date, and the study of host-pathogen coevolution at the genetic level will be a
major topic in the years to come. The development of DNA chips which represent
whole host genomes, including the human genome, will certainly help to address
these important questions in the future.
The accumulating sequence data for genomes of pathogenic as well as non-
pathogenic bacteria have shown that genetic structures similar to PAIs are also
part of the genomes of many nonpathogenic bacteria. These regions were termed
genomic islands because they were found to possess many features of PAIs, often
differing from the rest of the core genome in G+C content and coding usage,
and they often carry mobility genes (integrases and transposases). As with PAIs,
genomic islands are often be linked to tRNA genes and may be flanked by re-
peated DNA at their ends. Last, but not least, genomic islands may represent
genome regions of high instability. Many genomic islands, of course, do not
contribute to pathogenicity, and their gene products are rather involved in symbio-
sis, in degradation of xenobiotic compounds, in the generation of new metabolic
properties, or in the expression of antibiotic resistance phenotypes (6, 37, 49; for
details see Table 3). As already mentioned, iron uptake systems and bacterial
secretion pathways can be expressed either by pathogenic or by nonpathogenic
bacteria. In both cases they seem to contribute to fitness and adaptation of the
strains. It therefore appears that PAIs represent a genomic-island subgroup whose
members are, by definition, restricted to pathogenic bacteria. The analysis of
PAIs, however, not only opens new insights into the evolution of pathogens but also
contributes to the formulation of evolutional principles of prokaryotes in general.
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 673

ACKNOWLEDGMENTS
We thank Ute Hentschel (Würzburg) for helpful comments and Claudia Borde
(Würzburg) for editorial assistance. We apologize to the many colleagues whose
work we could not specifically cite due to space limitations. Our own work related
to this topic was supported by the Deutsche Forschungsgemeinschaft (SFB479,
Projekt A1) and the Fond der Chemischen Industrie as well as by the National
Institutes of Health (grants AI21657, AI19716, and AI41325 to JB Kaper).

Visit the Annual Reviews home page at www.AnnualReviews.org


Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

LITERATURE CITED
1. Adhikari P, Allison G, Whittle B, Verma NK. inserted into any of the three chromosomal
1999. Serotype 1a O-antigen modification: asn tRNA genes. Mol. Microbiol. 30:965–
molecular characterization of the genes in- 78
volved and their novel organization in the 8. Burland V, Shao Y, Perna NT, Plunkett
Shigella flexneri chromosome. J. Bacteriol. G, Sofia HJ, Blattner FR. 1998. The com-
181:4711–18 plete DNA sequence and analysis of the
2. Anderson DM, Schneewind O. 1997. A large virulence plasmid of Escherichia coli
mRNA signal for the type III secretion of O157:H7. Nucleic Acids Res. 26:4196–204
Yop proteins by Yersinia enterocolitica. Sci- 9. Burns DL. 1999. Biochemistry of type
ence 278:1140–43 IV secretion. Curr. Opin. Microbiol. 2:25–
3. Blocker A, Gounon P, Larquet E, Niebuhr 29
K, Cabiaux V, et al. 1999. The tripartite type 10. Cesini S, Lange C, Xiang Z, Crabtree JE,
III secreton of Shigella flexneri inserts IpaB Ghiara P, et al. 1996. cag, a pathogenic-
and IpaC into host membranes. J. Cell Biol. ity island of Helicobacter pylori, encodes
147:683–93 type-I specific and disease-associated vir-
4. Blum G, Ott M, Lischewski A, Ritter A, Im- ulence factors. Proc. Natl. Acad. Sci. USA
rich H, et al. 1994. Excision of large DNA 93:14648–53
regions termed pathogenicity islands from 11. Cheetham BF, Katz ME. 1995. A role for
tRNA-specific loci in the chromosome of an bacteriophages in the evolution and trans-
Escherichia coli wild-type pathogen. Infect. fer of bacterial virulence determinants.
Immun. 62:606–14 Mol. Microbiol. 18:201–8
5. Bourdet-Sicard R, Rudiger M, Jockusch 12. Cheetham BF, Whittle G, Katz ME. 1999.
BM, Gounon P, Sansonetti PJ, Tran VN. Are the vap regions of Dichelobacter no-
1999. Binding of the shigella protein IpaA to dosus pathogenicity islands? See Ref. 51a,
vinculin induces F-actin depolymerization. pp. 203–18
EMBO J. 18:5853–62 13. Cieslewicz M, Vimr E. 1997. Reduced
6. Briggs CE, Fratamico PM. 1999. Molecular polysialic acid capsule expression in Es-
characterization of an antibiotic resistance cherichia coli K1 mutants with chromo-
gene cluster of Salmonella typhimurium somal defects in kpsF. Mol. Microbiol.
DT104. Antimicrob. Agents Chemother. 26:237–49
43:846–49 14. Connell H, Hedlund M, Agace W, Svan-
7. Buchrieser C, Brosch R, Bach S, Guiyoule borg C. 1997. Bacterial attachment to uro-
A, Carniel E. 1998. The high-pathogenicity epithelial cells: mechanisms and conse-
island of Yersinia pseudotuberculosis can be quences. Adv. Dent. Res. 11:50–58
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

674 HACKER ¥ KAPER

15. Cornelis GR, Boland A, Boyd AP, Geui- 26. Fasano A, Noriega FR, Maneval DR Jr,
jen C, Iriarte M, et al. 1998. The virulence Chanasongcram S, Russell R, et al. 1995.
plasmid of Yersinia, an antihost genome. Shigella enterotoxin 1: an enterotoxin of
Microbiol. Mol. Biol. Rev. 62:1315–52 Shigella flexneri 2a active in rabbit small
16. Cornelis GR, Wolf-Watz H. 1997. The intestine in vivo and in vitro. J. Clin. In-
Yersinia Yop virulon: a bacterial system vest. 95:2853–61
for subverting eukaryotic cells. Mol. Mi- 27. Francetic O, Pugsley AP. 1996. The cryptic
crobiol. 23:861–67 general secretory pathway ( gsp) operon of
17. Cotter PA, Miller JF. 1998. In vivo and ex Escherichia coli K-12 encodes functional
vivo regulation of bacterial virulence gene proteins. J. Bacteriol. 178:3544–49
expression. Curr. Opin. Microbiol. 1:17– 28. Franco AA, Cheng RK, Chung GT, Wu S,
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

26 Oh HB, Sears CL. 1999. Molecular evo-


18. Covacci A, Rappuoli R. 1999. cag, the lution of the pathogenicity island of en-
pathogenicity island of Helicobacter py- terotoxigenic Bacteroides fragilis strains.
lori, triggers host response. See Ref. 51a, J. Bacteriol. 181:6623–33
pp. 189–202 29. Frankel G, Phillips AD, Rosenshine I,
19. Darwin KH, Miller VL. 1999. InvF is re- Dougan G, Kaper JB, Knutton S. 1998.
quired for expression of genes encoding Enteropathogenic and enterohaemorrhagic
proteins secreted by the SPI1 type III secre- Escherichia coli: more subversive ele-
tion apparatus in Salmonella typhimurium. ments. Mol. Microbiol. 30:911–21
J. Bacteriol. 181:4949–54 30. Fu Y, Galan JE. 1999. A salmonella protein
20. Deleted in proof antagonizes Rac-1 and Cdc42 to mediate
21. DiRita VJ. 1992. Co-ordinate expression host-cell recovery after bacterial invasion.
of virulence genes by ToxR in Vibrio Nature 401:293–97
cholerae. Mol. Microbiol. 6:451–58 31. Galán JE. 1999. Interaction of Salmonella
22. Dobrindt U, Hacker J. 1999. Plasmids, with host cells through the centisome 63
phages and pathogenicity islands: lessons type III secretion system. Curr. Opin. Mi-
on the evolution of bacterial toxins. In The crobiol. 2:46–50
Comprehensive Sourcebook of Bacterial 32. Galán JE, Collmer A. 1999. Type III se-
Protein Toxins, ed. J Alouf, J Freer, pp. cretion machines: bacterial devices for
3–23. New York: Academic protein delivery into host cells. Science
23. Eichel-Streiber von C, Braun V. 1999. 284:1322–28
Virulence-associated mobile elements in 33. Groisman EA, Blanc-Portard AB, Uchiya
bacilli and clostridia. See Ref. 51a, pp. K. 1999. Pathogenicity islands and the evo-
233–64 lution of Salmonella virulence. See Ref.
24. Elliott SJ, Hutcheson SW, Dubois MS, 51a, pp. 127–50
Mellies JL, Wainwright LA, et al. 1999. 34. Hacker J, Bender L, Ott M, Wingender J,
Identification of CesT, a chaperone for Lund B, et al. 1990. Deletions of chro-
the type III secretion of Tir in en- mosomal regions coding for fimbriae and
teropathogenic Escherichia coli. Mol. Mi- hemolysins occur in vivo and in vitro in
crobiol. 33:1176–89 various extraintestinal Escherichia coli iso-
25. Elliott SJ, Yu J, Kaper JB. 1999. The lates. Microb. Pathog. 8:213–25
cloned locus of enterocyte effacement 35. Hacker J, Blum-Oehler G, Janke B, Nagy
from enterohemorrhagic Escherichia coli G, Goebel W. 1999. Pathogenicity islands
O157:H7 is unable to confer the attaching of extraintestinal Escherichia coli. See Ref.
and effacing phenotype upon E. coli K-12. 51a, pp. 59–76
Infect. Immun. 67:4260–63 36. Hacker J, Blum-Oehler G, Mühldorfer I,
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 675

Tschäpe H. 1997. Pathogenicity islands of mals and plants. Microbiol. Mol. Biol.
virulent bacteria: structure, function and Rev. 62:379–433
impact on microbial evolution. Mol. Mi- 47. Hutcheson SW. 1999. The hrp cluster of
crobiol. 23:1089–97 Pseudomonas syringae: a pathogenicity
37. Hacker J, Kaper JB. 1999. The concept of island encoding a type III protein translo-
pathogenicity islands. See Ref. 51a, pp. 1– cation complex? See Ref. 51a, pp. 309–
11 29
38. Hacker J, Knapp S, Goebel W. 1983. Spon- 48. Iriarte M, Cornelis GR. 1999. The 70-
taneous deletions and flanking regions of kilobase virulence plasmid of Yersiniae.
the chromosomal inherited hemolysin de- See Ref. 51a, pp. 91–126
terminant of an Escherichia coli O6 strain. 49. Ito T, Katayama Y, Hiramatsu K. 1999.
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

J. Bacteriol. 154:1145–52 Cloning and nucleotide sequence de-


39. Haller JC, Carlson SJ, Pierson DE. 1999. A termination of the entire mec DNA
chromosomally-encoded type III secretion of pre-Methicillin-resistant Staphylococ-
apparatus of Yersinia enterocolitica affects cus aureus N315. Antimicrob. Agents
virulence in the mouse model of infection. Chemother. 43:1449–58
Abstr. ASM Conf. Cell Biol. Approach Mi- 50. Jackson RW, Athanassopoulos GT, Tsi-
crob. Pathog., Portland, OR, p. 25. Wash- amis G, Mansfield JW, Sesma A, et al.
ington, DC: Am. Soc. Microbiol. 1999. Identification of a pathogenicity is-
40. Häse CC, Mekalanos JJ. 1998. TcpP pro- land, which contains genes for virulence
tein is a positive regulator of virulence gene and avirulence, on a large native plasmid
expression in Vibrio cholerae. Proc. Natl. in the bean pathogen Pseudomonas sy-
Acad. Sci. USA 95:730–34 ringae pathovar phaseolicola. Proc. Natl.
41. Henderson IR, Czeczulin J, Eslava C, Nor- Acad. Sci. USA 96:10875–80
iega F, Nataro JP. 1999. Characterization of 51. Kaper JB, Gansheroff LJ, Wachtel WR,
Pic, a secreted protease of Shigella flexneri O’Brien AD. 1998. Intimin-mediated ad-
and enteroaggregative Escherichia coli. In- herence of Shiga toxin-producing Es-
fect. Immun. 67:5587–96 cherichia coli and attaching-and-effacing
42. Henderson IR, Navarro-Garcia F, Nataro pathogens. In Escherichia coli O157:H7
JP. 1998. The great escape: structure and and Other Shiga Toxin-Producing E. coli
function of the autotransporter proteins. Strains, ed. JB Kaper, AD O’Brien, pp.
Trends Microbiol. 6:370–78 148–56. Washington, DC: Am. Soc. Mi-
43. Hersh D, Monack DM, Smith MR, crobiol.
Ghori N, Falkow S, Zychlinsky A. 51a. Kaper JB, Hacker J, eds. 1999.
1999. The Salmonella invasin SipB in- Pathogenicity Islands and Other Mobile
duces macrophage apoptosis by binding Virulence Elements. Washington, DC:
to caspase-1. Proc. Natl. Acad. Sci. USA Am. Soc. Microbiol.
96:2396–401 52. Kaper JB, Mellies JL, Nataro JP. 1999.
44. Hilbi H, Moss JE, Hersh D, Chen Y, Aron- Pathogenicity islands and other mobile
del J, et al. 1998. Shigella-induced apopto- genetic elements of diarrheagenic Es-
sis is dependent on caspase-1 which binds cherichia coli. See Ref. 51a, pp. 33–58
to IpaB. J. Biol. Chem. 273:32895–900 53. Kaper JB, Morris JG Jr, Levine MM.
45. Hou YM. 1999. Transfer RNAs and 1995. Cholera. Clin. Microbiol. Rev.
pathogenicity islands. Trends Biochem. 8:48–86
Sci. 24:295–98 54. Karaolis DKR, Johnson JA, Bailey CC,
46. Hueck CJ. 1998. Type III protein secretion Boedeker EC, Kaper JB, Reeves PR.
systems in bacterial pathogens of ani- 1998. A Vibrio cholerae pathogenicity
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

676 HACKER ¥ KAPER

island associated with epidemic and pan- 64. Lindsay JA, Ruzin A, Ross HF, Kurepina
demic strains. Proc. Natl. Acad. Sci. USA N, Novick RP. 1998. The gene for toxic
95:3134–39 shock toxin is carried by a family of mo-
55. Karaolis DKR, Kaper JB. 1999. Patho- bile pathogenicity islands in Staphylococ-
genicity islands and other mobile virulence cus aureus. Mol. Microbiol. 29:527–43
elements of Vibrio cholerae. See Ref. 51a, 65. Lory S. 1998. Secretion of proteins and
pp. 167–87 assembly of bacterial surface organelles:
56. Karaolis DKR, Somara S, Maneval DR shared pathways of extracellular protein
Jr, Johnson JA, Kaper JB. 1999. A bacte- targeting. Curr. Opin. Microbiol. 1:27–35
riophage encoding a pathogenicity island, 66. Low D, David V, Lark D, Schoolnik
a type-IV pilus and a phage receptor in G, Falkow S. 1984. Gene clusters gov-
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

cholera bacteria. Nature 399:375–79 erning the production of hemolysin and


57. Karch H, Schubert S, Zhang D, Zhang W, mannose-resistant hemagglutination are
Schmidt H, et al. 1999. A genomic is- closely linked in Escherichia coli serotype
land, termed high-pathogenicity island, is O4 and O6 isolates from urinary tract in-
present in certain non-O157 Shiga toxin- fections. Infect. Immun. 43:353–58
producing Escherichia coli clonal lineages. 67. Marsh JW, Taylor RK. 1999. Genetic
Infect. Immun. 67:5994–6001 and transcriptional analyses of the Vib-
58. Kenny B, DeVinney R, Stein M, Rein- rio cholerae mannose-sensitive hemagglu-
scheid DJ, Frey EA, Finlay BB. 1997. En- tinin type 4 pilus gene locus. J. Bacteriol.
teropathogenic E. coli (EPEC) transfers its 181:1110–17
receptor for intimate adherence into mam- 68. Mazel D, Dychinco B, Webb VA, Davis J.
malian cells. Cell 91:511–20 1998. A distinctive class of integron in the
59. Knutton S, Rosenshine I, Pallen MJ, Vibrio cholerae genome. Science 280:605–
Nisan L, Neves BC, et al. 1998. A novel 8
EspA-associated surface organelle of en- 69. McDaniel TK, Jarvis KG, Donnersberg
teropathogenic Escherichia coli involved MS, Kaper JB. 1995. A genetic locus of
in protein translocation into epithelial cells. enterocyte effacement conserved among
EMBO J. 17:2166–76 diverse enterobacterial pathogens. Proc.
60. Kovach ME, Shaffer MD, Peterson KM. Natl. Acad. Sci. USA 92:1664–68
1996. A putative integrase gene defines 70. McDaniel TK, Kaper JB. 1997. A
the distal end of a large cluster of ToxR- cloned pathogenicity island from en-
regulated colonization genes in Vibrio teropathogenic Escherichia coli confers
cholerae. Microbiology 142:2165–74 the attaching and effacing phenotype on
61. Kreft J, Vazquez-Boland JA, Ng E, Goebel E. coli K-12. Mol. Microbiol. 23:399–407
W. 1999. Virulence gene clusters and puta- 71. McDonough MA, Butterton JR. 1999.
tive pathogenicity islands in Listeriae. See Spontaneous tandem amplification and
Ref. 51a, pp. 219–32 deletion of the Shiga toxin operon in
62. Kubori T, Matsushima Y, Nakamura Shigella dysenteriae 1. Mol. Microbiol.
D, Uralil J, Lara-Tejero M, et al. 34:1058–69
1998. Supramolecular structure of the 72. Mellies JL, Elliott SJ, Sperandio V, Don-
Salmonella typhimurium type III protein nenberg MS, Kaper JB. 1999. The Per reg-
secretion system. Science 280:602–5 ulon of enteropathogenic Escherichia coli:
63. Lawrence JG, Ochman H. 1998. Molec- identification of a regulatory cascade and a
ular archaeology of the Escherichia coli novel transcriptional activator, the locus of
genome. Proc. Natl. Acad. Sci. USA enterocyte effacement (LEE)-encoded reg-
95:9413–17 ulator (Ler). Mol. Microbiol. 33:296–306
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 677

73. Ménard R, Sansonetti P, Parsot C, Vasselon 81a. O’Callaghan D, Cazevielle C, Allardet-


T. 1994. Extracellular association and cy- Servent A, Boschiroli ML, Bourg G, et al.
toplasmic partitioning of the IpaB and 1999. A homologue of the Agrobacterium
IpaC invasins of S. flexneri. Cell 79:515– tumefaciens VirB and Bordetella pertus-
25 sis Ptl type IV secretion systems is essen-
74. Mietzner TA, Morse SA. 1994. The role tial for intracellular survival of Brucella
of iron-binding proteins in the survival suis. Mol. Microbiol. 33:1210–20
of pathogenic bacteria. Annu. Rev. Nutr. 82. Parsot C, Sansonetti PJ. 1999. The
14:471–93 virulence plasmid of Shigellae: an
75. Mills SD, Boland A, Sory MP, van der archipelago of pathogenicity islands? See
Missen P, Kerbourch C, et al. 1997. Ref. 51a, pp. 151–65
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

Yersinia enterocolitica induces apoptosis 83. Perna NT, Mayhew GF, PósfaI G, Elliott
in macrophages by a process requiring S, Donnenberg MS, et al. 1998. Molec-
functional type III secretion and translo- ular evolution of a pathogenicity island
cation mechanisms and involving YopP, from enterohemorrhagic Escherichia coli
presumably acting as an effector protein. O157:H7. Infect. Immun. 66:3810–17
Proc. Natl. Acad. Sci. USA 94:12638– 84. Perrin A, Nassif X, Tinsley C. 1999. Iden-
43 tification of regions of the chromosome
76. Mirold S, Rabsch W, Rohde M, Stender of Neisseria meningitidis and Neisseria
S, Tschäpe H, et al. 1999. Isolation of a gonorrhoeae which are specific to the
temperate bacteriophage encoding the type pathogenic Neisseria species. Infect. Im-
III effector protein SopE from an epidemic mun. 67:6119–29
Salmonella typhimurium strain. Proc. Natl. 85. Pugsley AP. 1993. The complete general
Acad. Sci. USA 96:9845–50 secretory pathway in gram-negative bac-
77. Monack DM, Mecsas J, Ghori N, Falkow teria. Microbiol. Rev. 57:50–108
S. 1997. Yersinia signals macrophages to 86. Pugsley AP, Francetic O, Possot OM,
undergo apoptosis and YopJ is necessary Sauvonnet N, Hardie KR. 1997. Recent
for this cell death. Proc. Natl. Acad. Sci. progress and future directions in studies
USA 94:10385–90 of the main terminal branch of the gen-
78. Morschhäuser J, Köhler G, Ziebuhr W, eral secretory pathway in gram-negative
Blum-Oehler G, Dobrindt U, Hacker J. bacteria: a review. Gene 192:13–19
2000. Evolution of microbial pathogens. 87. Rajakumar K, Sasakawa C, Adler B.
Phil. Trans. R. Soc. London B 355:695– 1997. Use of a novel approach, termed
704 island probing, identifies the Shigella
79. Morschhäuser J, Vetter V, Emödy L, flexneri she pathogenicity island which
Hacker J. 1994. Adhesin regulatory genes encodes a homolog of the immunoglob-
within large, unstable DNA regions of ulin A protease-like family of proteins.
pathogenic Escherichia coli: cross-talk be- Infect. Immun. 65:4606–14
tween different adhesin gene clusters. Mol. 88. Rakin A, Schubert S, Pelludat C, Brem
Microbiol. 11:555–66 D, Heesemann J. 1999. The high-
80. Moss JE, Cardozo TJ, Zychlinsky A, Gro- pathogenicity island of yersiniae. See Ref.
isman EA. 1999. The selC-associated SHI- 51a, pp. 77–90
2 pathogenicity island of Shigella flexneri. 89. Reiter W, Palm DP, Yeats S. 1989. Trans-
Mol. Microbiol. 33:74–83 fer RNA genes frequently serve as inte-
81. Nataro JP, Kaper JB. 1998. Diarrheagenic gration sites for prokaryotic genetic ele-
Escherichia coli. Clin. Microbiol. Rev. ments. Nucleic Acids Res. 17:1907–14
11:142–201 90. Ritter A, Gally DL, Olsen PB, Dobrindt
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

678 HACKER ¥ KAPER

U, Friedrich A, et al. 1997. The PAI- regulate the coordinate expression of


associated leuX specific tRNA5Leu affects cholera toxin and toxin-coregulated pilus
type 1 fimbriation in pathogenic Esch- in Vibrio cholerae. Proc. Natl. Acad. Sci.
erichia coli by control of FimB recombi- USA 94:265–70
nase expression. Mol. Microbiol. 25:871– 99. Sozhamannan S, Deng YK, Li M, Su-
82 lakvelidze A, Kaper JB, et al. 1999.
91. Roine E, Wei W, Yuan J, Nurmiaho-Lassila Cloning and sequencing of the genes
E-L, Kalkkinen N, et al. 1997. Hrp pilus: downstream of the wbf gene cluster of Vib-
an hrp-dependent bacterial surface ap- rio cholerae serogroup O139 and analysis
pendage produced by Pseudomonas sy- of the junction genes in other serogroups.
ringae pv. tomato DC3000. Proc. Natl. Infect. Immun. 67:5033–40
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

Acad. Sci. USA 94:3459–64 100. Sperandio V, Mellies JL, Nguyen W, Shin
92. Rossier O, Wengelnik K, Hahn K, Bonas U. S, Kaper JB. 1999. Quorum sensing con-
1999. The Xanthomonas Hrp type III sys- trols expression of the type III secretion
tem secretes proteins from plant and mam- gene transcription and protein secretion in
malian bacterial pathogens. Proc. Natl. enterohemorrhagic and enteropathogenic
Acad. Sci. USA 96:9368–73 Escherichia coli. Proc. Natl. Acad. Sci.
93. Schechter LM, Damrauer SM, Lee CA. USA 96:15196–201
1999. Two AraC/XylS family members can 101. Stein MA, Leung KY, Zwick M, del Por-
independently counteract the effect of re- tillo FG, Finlay BB. 1996. Identification
pressing sequences upstream of the hilA of a Salmonella virulence gene required
promoter. Mol. Microbiol. 32:629–42 for formation of filamentous structures
94. Schubert S, Rakin A, Karch H, Carniel E, containing lysosomal membrane glyco-
Heeseman J. 1998. Prevalence of the high proteins within epithelial cells. Mol. Mi-
pathogenicity island of Yersinia species crobiol. 20:151–64
among Escherichia coli strains that are 102. Tacket CO, Taylor RK, Losonsky G, Lim
pathogenic to humans. Infect. Immun. Y, Nataro JP, et al. 1998. Investigation
66:480–85 of the roles of toxin-coregulated pili and
95. Segal ED, Cha J, Lo J, Falkow S, Tomp- mannose-sensitive hemagglutinin pili in
kins LS. 1999. Altered states: involvement the pathogenesis of Vibrio cholerae O139
of phosphorylated CagA in the induction infection. Infect. Immun. 66:692–95
of host cellular growth changes by Heli- 103. Tran Van Nhieu G, Caron E, Hall A,
cobacter pylori. Proc. Natl. Acad. Sci. USA Sansonetti PJ. 1999. IpaC induces actin
96:14559–64 polymerization and filopodia formation
96. Segal G, Purcell M, Shuman HA. 1998. during Shigella entry into epithelial cells.
Host cell killing and bacterial conjugation EMBO J. 18:3249–62
require overlapping sets of genes within 104. Tran Van Nhieu G, Sansonetti PJ. 1999.
a 22-kb region of the Legionella pneu- Mechanism of Shigella entry into epithe-
mophila genome. Proc. Natl. Acad. Sci. lial cells. Curr. Opin. Microbiol. 2:51–
USA 95:1669–74 55
97. Segal G, Russo JJ, Shuman HA. 1999. 105. Trucksis M, Michalski J, Deng YK,
Relationships between a new type IV se- Kaper JB. 1998. The Vibrio cholerae
cretion system and the icm/dot virulence genome contains two unique circular
system of Legionella pneumophila. Mol. chromosomes. Proc. Natl. Acad. Sci. USA
Microbiol. 34:799–809 95:14464–69
98. Skorupski K, Taylor RK. 1997. Cyclic 106. Uchiya K, Barbieri MA, Funatao K, Shah
AMP and its receptor protein negatively AH, Stahl PD, Groisman EA. 1999. A
P1: FRD/FQU P2: FQP
August 11, 2000 11:13 Annual Reviews AR110-19

PATHOGENICITY ISLANDS 679

Salmonella virulence protein that inhibits 114. Wood MW, Jones MA, Watson PR,
cellular trafficking. EMBO J. 18:3924–33 Hedges S, Wallis TS, Galyov EE. 1998.
107. Vogel JP, Andrews HL, Wong SK, Is- Identification of a pathogenicity is-
berg RR. 1998. Conjugative transfer by land required for Salmonella entero-
the virulence system of Legionella pneu- pathogenicity. Mol. Microbiol. 29:883–
mophila. Science 279:873–76 91
108. Vokes SA, Torres AG, Reeves SA, Payne 115. Wu S, Lim KC, Huang J, Saidi RF, Sears
SM. 1999. The aerobactin iron trans- CL. 1998. Bacteroides fragilis entero-
port system genes in Shigella flexneri toxin cleaves the zonula adherens protein,
are present within a pathogenicity island. E-cadherin. Proc. Natl. Acad. Sci. USA
Mol. Microbiol. 33:63–73 95:14979–84
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

109. Wachter C, Beinke C, Mattes M, Schmidt 116. Wyckoff EE, Duncan D, Torres AG, Mills
MA. 1999. Insertion of EspD into epithe- M, Maase K, Payne SM. 1998. Structure
lial target cell membranes by infecting en- of the Shigella dysenteriae heaem trans-
teropathogenic Escherichia coli. Mol. Mi- port locus and its phylogenetic distribu-
crobiol. 31:1695–707 tion in enteric bacteria. Mol. Microbiol.
110. Waldor MK, Mekalanos JJ. 1996. Lyso- 28:1139–52
genic conversion by a filamentous 117. Yildiz FH, Schoolnik GK. 1999. Vib-
phage encoding cholera toxin. Science rio cholerae O1 El Tor: identification
272:1910–14 of a gene cluster required for the ru-
111. Watnick PI, Fullner KJ, Kolter R. 1999. gose colony type, exopolysaccharide pro-
A role for the mannose-sensitive hemag- duction, chlorine resistance, and biofilm
glutinin in biofilm formation by Vibrio formation. Proc. Natl. Acad. Sci. USA
cholerae El Tor. J. Bacteriol. 181:3606–9 96:4028–33
112. Wattiau P, Woestyn S, Cornelis G. 118. Ziebuhr W, Krimmer V, Rachid S, Loss-
1996. Customized secretion chaperones ner I, Götz F, Hacker J. 1999. A novel
in pathogenic bacteria. Mol. Microbiol. mechanism of phase variation of vir-
20:255–62 ulence in Staphylococcus epidermidis:
113. Winans SC, Kalogeraki V, Jafri S, evidence for control of the polysaccha-
Akakura R, Xia Q. 1999. Diverse roles ride intercellular adhesin synthesis by
of Agrobacterium Ti plasmid-borne genes alternating insertion and excision of the
in the formation and colonization of plant insertion sequence element IS256. Mol.
tumors. See Ref. 51a, pp. 289–307 Microbiol. 32:345–56
Annual Review of Microbiology
Volume 54, 2000

CONTENTS
THE LIFE AND TIMES OF A CLINICAL MICROBIOLOGIST, Albert
Balows 1
ROLE OF CYTOTOXIC T LYMPHOCYTES IN EPSTEIN-BARR
VIRUS-ASSOCIATED DISEASES, Rajiv Khanna, Scott R. Burrows 19
BIOFILM FORMATION AS MICROBIAL DEVELOPMENT, George
O'Toole, Heidi B. Kaplan, Roberto Kolter 49
MICROBIOLOGICAL SAFETY OF DRINKING WATER, U. Szewzyk,
R. Szewzyk, W. Manz, K.-H. Schleifer 81
THE ADAPTATIVE MECHANISMS OF TRYPANOSOMA BRUCEI
FOR STEROL HOMEOSTASIS IN ITS DIFFERENT LIFE-CYCLE
ENVIRONMENTS, I. Coppens, P. J. Courtoy 129
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.

THE DEVELOPMENT OF GENETIC TOOLS FOR DISSECTING THE


BIOLOGY OF MALARIA PARASITES, Tania F. de Koning-Ward,
Chris J. Janse, Andrew P. Waters 157
NUCLEIC ACID TRANSPORT IN PLANT-MICROBE
INTERACTIONS: The Molecules That Walk Through the Walls, Tzvi
Tzfira, Yoon Rhee, Min-Huei Chen, Talya Kunik, Vitaly Citovsky
187
PHYTOPLASMA: Phytopathogenic Mollicutes, Ing-Ming Lee, Robert E.
Davis, Dawn E. Gundersen-Rindal 221
ROOT NODULATION AND INFECTION FACTORS PRODUCED BY
RHIZOBIAL BACTERIA, Herman P. Spaink 257
ALGINATE LYASE: Review of Major Sources and Enzyme
Characteristics, Structure-Function Analysis, Biological Roles, and
Applications, Thiang Yian Wong, Lori A. Preston, Neal L. Schiller 289
INTERIM REPORT ON GENOMICS OF ESCHERICHIA COLI, M.
Riley, M. H. Serres 341
ORAL MICROBIAL COMMUNITIES: Biofilms, Interactions, and
Genetic Systems, Paul E. Kolenbrander 413
ROLES OF THE GLUTATHIONE- AND THIOREDOXIN-
DEPENDENT REDUCTION SYSTEMS IN THE ESCHERICHIA COLI
AND SACCHAROMYCES CEREVISIAE RESPONSES TO OXIDATIVE
STRESS, Orna Carmel-Harel, Gisela Storz 439
RECENT DEVELOPMENTS IN MOLECULAR GENETICS OF
CANDIDA ALBICANS, Marianne D. De Backer, Paul T. Magee, Jesus
Pla 463
FUNCTIONAL MODULATION OF ESCHERICHIA COLI RNA
POLYMERASE, Akira Ishihama 499
BACTERIAL VIRULENCE GENE REGULATION: An Evolutionary
Perspective, Peggy A. Cotter, Victor J. DiRita 519
LEGIONELLA PNEUMOPHILA PATHOGENESIS: A Fateful Journey
from Amoebae to Macrophages, M. S. Swanson, B. K. Hammer
567
THE DISEASE SPECTRUM OF HELICOBACTER PYLORI : The
Immunopathogenesis of Gastroduodenal Ulcer and Gastric Cancer, Peter
B. Ernst, Benjamin D. Gold 615
PATHOGENICITY ISLANDS AND THE EVOLUTION OF
MICROBES, Jörg Hacker, James B. Kaper 641
DNA SEGREGATION IN BACTERIA, Gideon Scott Gordon, Andrew
Wright 681
POLYPHOSPHATE AND PHOSPHATE PUMP, I. Kulaev, T.
Kulakovskaya 709
ASSEMBLY AND FUNCTION OF TYPE III SECRETORY SYSTEMS,
Guy R. Cornelis, Frédérique Van Gijsegem 735
PROTEINS SHARED BY THE TRANSCRIPTION AND
TRANSLATION MACHINES, Catherine L. Squires, Dmitry Zaporojets
775
HOLINS: The Protein Clocks of Bacteriophage Infections, Ing-Nang
Wang, David L. Smith, Ry Young 799
OXYGEN RESPIRATION BY DESULFOVIBRIO SPECIES, Heribert
Cypionka 827
REGULATION OF CARBON CATABOLISM IN BACILLUS
SPECIES, J. Stülke, W. Hillen 849
IRON METABOLISM IN PATHOGENIC BACTERIA, Colin Ratledge,
Lynn G Dover 881
Annu. Rev. Microbiol. 2000.54:641-679. Downloaded from arjournals.annualreviews.org
by Secretaria de Ciencia y Tecnologia de Argentina on 06/21/05. For personal use only.
Genetic context of ISCR1 elements found in association with antibiotic resistance genes as part of complex class 1 integrons.
ISCRs are represented by yellow boxes. The qac/sul 3′ ends of the class 1 complex integrons are denoted by gray and black boxes,
respectively. The blue box denoting the intgene (encoding the integrase) represents the 5′ end of the class 1 integron. Gene
cassettes forming parts of class 1 integrons are represented by white boxes with aqua vertical lines. Genes that are not part of
cassettes have been given their own individual color as indicated by the key in Fig. 4. Open reading frames are indicated with
open boxes and the direction of their transcription indicated with arrows. See the text for a detailed description of class 1
integrons. (A) Complex class 1 integrons In6 and In7, in which ISCR1 elements were first observed. (B to F) Genetic context of
ISCR1 associated with trimethoprim resistance dfr genes, ciprofloxacin resistance qnrA genes, aminoglycoside resistance genes,
class C β-lactamase genes, and class A β-lactamase genes, respectively. Accession numbers of the nucleotide sequences of the
various gene arrays are included on the right. Microbiol. Mol. Biol. Rev. June 2006 vol. 70 no. 2 296-316
Table 1. Putative and proved integrative and conjugative elements.a,b

Size Integrase
Elements Genus or species (kb) Other carried functions type Integration sites References

TCrERLa Bacteroides fragilis 80 Tetr ND Few sites Salyers et al. (1995b)


CTnDOTa Bacteroides 65 Tetr Ermr Tyr Few sites (GTTNNTTTGG) Cheng et al. (2001)
Symbiosis islanda Mezorhizobium loti R7A 502 Symbiosis Tyr 3¢ of a gene encoding tRNAPhe gene Sullivan et al. (2002)
CTnscr94 Salmonella enterica 100 Sucrose utilization ND 3¢ of two genes encoding a tRNAPhe Hochhut et al. (1997)
SXTa Vibrio cholerae 99.5 Sufr Trmr Camr Strr Tyr 5¢ of a gene encoding RF3 Hochhut and Waldor (1999)
R391a Providencia rettgeri 94 Kanr Hgr Tyr 5¢ of a gene encoding RF3 Murphy and Pembroke (1999)
clc Pseudomonas sp. B13 105 Chlorocatechol degradation Tyr 3¢ of two genes encoding a tRNAGly Ravatn et al. (1998b)
bph–sal Pseudomonas putida 90 Biphenyl and salicylate degradation ND ND Nishi et al. (2000)
pSAM2a Streptomyces ambofaciens 10.9 None identified Tyr 3¢ of a gene encoding a tRNAPro Pernodet et al. (1984)
pSE101a Saccharopolyspora erythraea 11.3 None identified Tyr 3¢ of a gene encoding a tRNAThr Brown et al. (1994)
Streptomyces lividans c Numerous sites
Tn1549 a Enterococcus sp. 34 Vanr Tyr A+T rich Garnier et al. (2000)
Tn916 a Enterococcus faecalis 18 Tetr Tyr A+T rich Clewell et al. (1995)

© 2002 Blackwell Publishing Ltd, Molecular Microbiology, 46, 601–610


Clostridium difficile One preferential site Wang et al. (2000a)
Tn5397 a C. difficile 21 Tetr Ser Single site Wang et al. (2000b)
Bacillus subtilis c Numerous sites
Tn5252 a Streptococcus pneumoniae 47 Camr Tyr Single intergenic site Vijayakumar and Ayalew (1993)
Tn5276 a Lactococcus lactis 70 Sucrose utilization and nisin synthesis Tyr Few sites (TTTTTG) Rauch and de Vos (1992)
pRS01/sex factor L. lactis 48.4 Tellurium resistance ND Single site Gasson et al. (1995)
EfaC1a E. faecalis 25.3 ND Tyr 3¢ end of a gene encoding a ThrtRNA Burrus et al. (2002)
EfaC2a E. faecalis 32.7 ND Tyr 3¢ of a gene encoding a GMP-synthase Burrus et al. (2002)
EfaD2a E. faecalis ND ND Tyr ND Burrus et al. (2002)
Tn5801a Staphylococcus aureus 25.8 Tetr Tyr 3¢ of a gene encoding a GMP-synthase Kuroda et al. (2001)
CW459tet(M)a Clostridium perfringens ND Tetr Tyr 3¢ of a gene encoding a GMP-synthase Roberts et al. (2001)
ICELm1a Listeria monocytogenes 21.3 Putative Cd2+ resistance Tyr 3¢ of a gene encoding a GMP-synthase Burrus et al. (2002)
ICESt1a Streptococcus thermophilus 34.7 Restriction-modification Sth368I Tyr 3¢ end of a gene encoding a Burrus et al. (2002)
fructose-1,6-diphosphate aldolase

a. Other related integrative and conjugative elements have not been indicated in this table.
b. The names of the proven ICEs are not underlined. The names of the putative ICEs are underlined.
c. pSE101 and Tn5397 were not found in S. lividans and B. subtilis respectively.
Cam, chloramphenicol; Erm, erythromycin; Hg, mercury; Kan, kanamycin; ND, not determined; Ser, serine recombinase; Str, streptomycin; Suf, sulphamethoxazole; Tet, tetracycline; Trm,
trimethoprim; Tyr, tyrosine recombinase; Van, vancomycin;
Conjugative transposons 603
Tetracycline
A 1 kb Conjugation resistance RegulationRecombination

Tn5397
Group II
intron

88% 88% 84%

Tn916

67%

Tn1549

Conjugation Vancomycin resistance Recombination


Regulation
B
1 kb
Recombination Conjugation

EfaC1

0%
20%
40%
70%
100%
% identity of proteins

> 90% nt identity with pAD1


EfaC2
606 V. Burrus, G. Pavlovic, B. Decaris and G. Guédon

Conjugation Recombination

C
1 kb Recombination Regulation Conjugation
EfaD2

0%
20%
40%
70%
100%
% identity of proteins

pAM373

Conjugation Replication

Fig. 1. Comparison of the gene organization of some putative or proven conjugative elements.
A. Conjugative transposons Tn5397, Tn916 and Tn1549.
B. Putative site-specific integrative and conjugative elements EfaC1 and EfaC2.
C. Putative integrative conjugative element/conjugative transposon EfaD2 and conjugative plasmid pAM373. Boxes represent the ORFs, with the proposed direction of transcription shown by the
arrow. ORF names beginning with ‘orf ’ (Tn5397, Tn916, Tn1549), ‘EF’ (EfaC1, EfaC2, EfaD2) or ‘EP’ (pAM373) in the GenBank annotation are abbreviated with the corresponding number. The
colour of the boxes depicts the putative function of each ORF deduced from functional analysis or from BLASTP comparison: Blue, conjugative transfer; grey, antibiotic resistance; green, putative
transcriptional regulator related to those of Tn916; orange, serine recombinase; red, excisionase and tyrosine recombinase; black, plasmid replication functions; white, other putative functions
or no putative function.
In (A), the ORFs are connected by grey shading when significant nucleotide identity (given between brackets) is detected; the ORFs are connected by light yellow shading when the predicted
proteins share significant sequence identity (<40% in BLASTP comparison).
In (B) and (C), the ORFs are connected by a yellow/orange shading when the predicted proteins share significant sequence identity; the colour of this shading is linked to the percentage amino
acid identity. The whole sequence of each element is presented, except for EfaD2, as its ends are not known. In Tn5397, the group II intron is depicted by a square pattern. In Tn5397, orf5 is

© 2002 Blackwell Publishing Ltd, Molecular Microbiology, 46, 601–610


not described by Roberts et al. (2001), but is found by sequence comparison with Tn916. In EfaC1, the black line indicates ORFs closely related to those of the conjugative plasmid pAD1 (>90%
identity in nucleotide sequence). Whereas the ORFs EF2546 from EfaC1 and EF0166 from EfaC2 encode distantly related putative integrases (29% amino acid identity), they are not connected
in (B).
© 1988 Nature Publishing Group
© 1988 Nature Publishing Group
© 1988 Nature Publishing Group
© 1988 Nature Publishing Group
Blackwell Science, LtdOxford, UKMMIMolecular Microbiology0950-382XBlackwell Publishing Ltd, 2004? 2004523643650Review ArticleStress-directed mutationsB. E. Wright

Molecular Microbiology (2004) 52(3), 643–650 doi:10.1111/j.1365-2958.2004.04012.x

MicroReview

Stress-directed adaptive mutations and evolution

Barbara E. Wright* question concerns the origin of the variants from which
Division of Biological Sciences, University of Montana, the fittest survive. Dogma has it that these variants arise
Missoula, MT 59812, USA. from pre-existing ‘random’ mutations that are not directed
by the different conditions of stress that select them. In
other words, there would be no specific biochemical
Summary
mechanisms initiated by each stressor that can increase
Comparative biochemistry demonstrates that the mutation rates in those genes related to that stress.
metabolites, complex biochemical networks, However, specific, stress-directed mutagenesis (SDM)
enzymes and regulatory mechanisms essential to all would offer an enormous advantage for evolution and
living cells are conserved in amazing detail through- would be selected. Therefore, if SDM exists it should be
out evolution. Thus, in order to evolve, an organism present today in organisms coping with adverse condi-
must overcome new adverse conditions without cre- tions in their environment. Increased mutation rates would
ating different but equally dangerous alterations in its be directed to specific genes that must mutate to alleviate
ongoing successful metabolic relationship with its the stress, while avoiding random genome-wide damage.
environment. Evidence suggests that stable long- The search for directed mutations has been stymied by
term acquisitive evolution results from minor the problem posed by Delbrück in (1946): if a mutation
increases in mutation rates of genes related to a par- can only be observed under conditions that may cause it,
ticular stress, with minimal disturbance to the bal- as well as select it, how can we tell the difference? Does
anced and resilient metabolism critical for responding the stress cause the mutation before the mutant is
to an unpredictable environment. Microorganisms selected? This conundrum has understandably led to con-
have evolved specific biochemical feedback mecha- fusion, controversy, and, by default, acceptance of the
nisms that direct mutations to genes derepressed by Dogma first articulated more than 100 years ago by Weis-
starvation or other stressors in their environment. mann (1893). Recent advances in this field reveal mech-
Transcription of the activated genes creates localized anisms by which a specific stress does in fact cause
supercoiling and DNA secondary structures with mutations in related genes, thus resolving the issue raised
unpaired bases vulnerable to mutation. The resulting by Delbrück.
mutants provide appropriate variants for selection by This MicroReview attempts to critique and relate three
the stress involved, thus accelerating evolution with different areas of research concerned with mechanisms
minimal random damage to the genome. This model underlying adaptive mutations: (i) acquisitive evolution, in
has successfully predicted mutation frequencies in which gain-of-function mutations confer new and advan-
genes of E. coli and humans. Stressed cells observed tageous capabilities upon organisms; (ii) experimental
in the laboratory over hundreds of generations accu- evolution, observed over hundreds of generations using
mulate mutations that also arise by this mechanism. glucose limited/starved microbial populations, and (iii) bio-
When this occurs in repair-deficient mutator strains chemical mechanisms underlying SDM.
with high rates of random mutation, the specific
stress-directed mutations are also enhanced.
Acquisitive evolution

Investigators in this field have provided beautiful examples


Introduction
of evolving biosynthetic and catabolic pathways respond-
Evolution is defined as the result of environmental stress ing to environmental stress. These mutations modify exist-
that selects the fittest. The yet unresolved and fascinating ing gene-enzyme systems to initiate pathways using a
new carbon source as the previous source becomes lim-
Accepted 7 January, 2004. *For correspondence. E-mail
barbara.wright@mso.umt.edu; Tel. (+1) 406 243 6676; Fax (+1) 406 iting to survival (for reviews see Hegeman and Rosen-
243 4184. berg, 1970; Lin et al., 1976; Clarke, 1984; Mortlock,

© 2004 Blackwell Publishing Ltd


644 B. E. Wright
1984). Early investigations in this field demonstrated that Phe+ mutations found on selective plates from stationary-
cells starved in the presence of a carbon source they phase cells was higher than from exponentially growing
cannot use produced mutants that could use it. The most cells, they concluded that specific factors in starving cells
common mechanism involved is gene derepression result- must facilitate mutations conferring the ability to utilize
ing in the constitutive production of a previously inducible phenol. Other investigators have also observed that
enzyme or an enzyme with altered substrate specificity. mutants arising from SDM in starving bacteria differ from
Many examples exist; some of the earliest are the use of those found during growth, implicating different molecular
altrose-galactoside via b-galactosidase (Lederberg, mechanisms (Prival and Cebula, 1992; Rosenberg et al.,
1951); b-glycerolphosphate via alkaline phosphatase 1994; Foster and Trimarchi, 1995; Notley-McRobb and
(Torriani and Rothman, 1961) and xylitol via ribitol dehy- Ferenci, 1999a; Sung and Yasbin, 2002). Although star-
drogenase (Lerner et al., 1964). As discussed below, such vation and gene derepression are probably the predomi-
derepressed genes are actively transcribed and thereby nant cause of SDM in nature, others include direct effects
become targets for SDM. The metabolic steps required to on supercoiling by variables such as osmolarity, temper-
metabolize a new related substrate are similar to those for ature or anaerobiosis. For example, deletion mutations of
existing pathways. Therefore, relatively minor changes to the stress-induced spvB gene in Salmonella (Massey
a duplicated copy of the existing gene may be required for et al., 1999) only occur at a specific concentration of NaCl
recruitment to serve a new function (reviewed in Wright, that would presumably induce a particular level of super-
2000). Other frequently observed mutations in response coiling, which is known to create DNA secondary structures
to carbon source starvation confer increased permeability containing unpaired bases vulnerable to mutations (Ripley
for the limiting metabolite. Examples of this kind will be and Glickman, 1983; Wright et al., 2002; Wright et al.,
discussed below in the section on experimental evolution. 2003). Supercoiling also plays a critical role in selective
The field of acquisitive evolution has been enhanced by gene transcription (Lefstin and Yamamoto, 1998), for exam-
biotechnologists, who have published many papers on the ple, in sensitive genes such as proU (Higgins et al., 1988)
results of SDM. Genetic engineering is used to achieve and Hg-MerR (Ansari et al., 1992).
strain improvement by increasing the yield of a valuable
product for example, or allowing the metabolism of a
Experimental evolution
xenobiotic compound (van der Meer et al., 1992). Although
genetically engineered microbial strains are less compet- A number of laboratories study mutations that arise over
itive in natural environments than those that originally many generations in microbial populations limited or
evolved and were selected under those conditions, they starved for a carbon source (for example, Dykhuizen and
have provided valuable insight into mechanisms by which Hartl, 1983; Death et al., 1993; Notley-McRobb and Fer-
new catabolic and biosynthetic pathways arise during evo- enci, 1999a, b; Funchain et al., 2000; Giraud et al., 2001;
lution. Of particular relevance are studies that are similar Notley-McRobb et al., 2003). The consequences of glu-
to investigations by evolutionary biologists analysing cose starvation (in contrast to less ‘global’ nutrients such
mechanisms of acquisitive evolution. Single point muta- as phosphate or an amino acid) are extremely widespread
tions have been shown to extend the substrate range for and difficult to sort out, as all areas of metabolism are
the degradation of many organic compounds, for example, affected directly or, eventually, indirectly. A multitude of
xylene monooxygenase (Abril et al., 1989) and catachol genes are derepressed and activated as a result of the
dioxygenase (Ramos et al., 1987). Gain-of-function muta- stringent response (Cashel et al., 1996), for example, relA
tions for metabolizing a new substrate typically require and spotT which regulate the accumulation of ppGpp that
starvation conditions and long exposure times to that sub- is required for the accumulation of rpoS-encoded ss. This
strate as the sole carbon source (e.g. Nochur et al., 1990). central regulator then initiates a cascade derepression of
Some examples of specific SDM in starving bacteria starvation-responsive genes including those protecting
are particularly well-documented and pertinent to this the cells from additional stressors such as heat, osmotic
review. Kasak et al. (1997) described the accumulation of shock, anaerobiosis, and oxidative damage (Gentry et al.,
phenol-utilizing mutants (Phe+) in starving cultures of a 1993; Hengge-Aronis, 1996). The cAMP-CRP complex
promoterless phenol degradation operon of P. putida in accumulates and activates about two-thirds of the carbon
the presence, but not the absence, of phenol (the possibility starvation responsive genes. Derepressed genes include
that the absence of phenol eliminates Phe+ mutants from those improving outer membrane permeability and glu-
starving populations was excluded). Sequence analysis of cose transport, and those encoding enzymes using alter-
these mutants revealed base substitutions, deletions and nate carbon sources, such as lactose, maltose, galactose
insertions that in effect created a promoter. These muta- and ribose (Saier et al., 1996). A burst of supercoiling
tions differed in type and frequency in starving as com- accompanies this widespread derepression and a 30-fold
pared to growing populations. Because the frequency of higher mutation rate is seen in severely glucose-limited

© 2004 Blackwell Publishing Ltd, Molecular Microbiology, 52, 643–650


Stress-directed mutations 645
cultures with maximally derepressed genes than in mildly The most fascinating results were seen in the analysis
limited cultures (Notley-McRobb and Ferenci, 1999a). As of 14 point mutations of malT, encoding the specific MalT
discussed below, such derepressed genes are not only activator protein (Dardonville and Raibaud, 1990; Notley-
activated and selected in starving cells, but also become McRobb and Ferenci, 1999a) which regulates at least five
targets for SDM that may provide beneficial mutations and ‘maltose box’ promoters and LamB glycoporin expression
increased fitness for the stressed cells. The majority of critical to outer membrane permeability (Death et al.,
mutations found in experimental evolution experiments do 1993). These mutations are observed only in response to
not appear to be random, but occur repeatedly in these severe glucose limitation, and result in constitutive expres-
newly derepressed genes and in replicate cultures. sion of the regulon. Quite unexpectedly, they were seen
The most frequently identified mutants in glucose- to share the same characteristics found in the mutable
starved cultures show improved uptake of the limiting nutri- bases of a model system of the immune response (Wright
ent (Dykhuizen and Hartl, 1983). A literature search for et al., 2004). Ninety per cent of these malT point muta-
large data-bases of sequenced mutations from such cul- tions occurred at G:C base pairs; they were rarely
tures was found in the work of Dardonville and Raibaud unpaired and located at the ends of SLSs (unpublished
(1990) and Notley-McRobb and Ferenci (1999a,b). The data). The most striking characteristic linking these muta-
majority of mutations (17, see Table 2 in Notley-McRobb ble bases to those in the immune system is that the rarely
and Ferenci, 1999b) occurred in the mgl operator and unpaired bases are preferentially influenced by transcrip-
showed greater rates of glucose transport than the mglD tion. There is an inverse correlation (P = 0.03) between
null mutations inactivating a repressor that binds to the the extent to which mutable malT bases are unpaired and
operator. These authors believe that the mglO mutations, the increase in stability of their SLSs as a function of
located within a 14 bp sequence, were non-random and increased levels of transcription. This correlation suggests
somehow directed to the operator. The regulatory muta- a mechanism by which transcription could determine the
tions increasing glucose transport were analysed using a order and intensity of base mutability (Wright et al., 2004).
new computer program, mfg, to determine if SDM were The gain-of-function malT mutations are clustered in two
responsible for their appearance (Wright et al., 1999; small regions of the gene, and they all result in the same
2002; 2003; 2004). This program (see below) has suc- discrete and well-defined change in MalT conformation.
cessfully predicted mutation frequencies in E. coli and The altered conformation confers constitutivity on this key
humans, based on the extent to which a mutable base is activator protein which binds to specific nucleotide
unpaired during transcription and upon the stability of the sequences in the promoters it activates (Dardonville and
DNA secondary structure in which it is unpaired. The eight Raibaud, 1990). Adaptations enhancing antibody affinity
mglO point mutations were analysed and six were found for antigens during the immune response also involve
to be similar to those described in amino acid auxotrophs alterations in protein conformation. We speculate that
of E. coli (Wright et al., 2003) and in human p53 cancers such adaptations required the evolution of localized
(Wright et al., 2002). The mutable sites were located in groups of unpaired mutable bases allowing coordinated
small loops very near or between stems of DNA stem-loop trials with simultaneous mutations at many sites to
structures (SLSs) with high stability, and their predicted achieve a best fit for binding to their target. In any event,
mutability was high, as they were unpaired and exposed it appears that mutations occurring in derepressed genes
most of the time (averaging 94% of their folds) during of glucose-starved cultures are not random, but share the
transcription (unpublished data). Of the nine mglD- mechanisms of SDM elicited by many kinds of stressors
encoded null repressor mutants available for analysis most in the environment (see below).
were similar to the mglO mutable bases but were less Compared to wild type, the frequency of mismatch
frequently unpaired and had lower predicted mutabilities. repair defective mutator strains is higher in experimental
Investigations with experimental evolution in glucose- evolution experiments where strong selection is applied
limited cultures are not designed to determine mutation (Cox and Gibson, 1974; Notley-McRobb et al., 2003).
frequencies. However, we have reported reversion rates Funchain et al. (2000) have monitored the function of
of a derepressed lacZ auxotroph which serves as a model more than 700 genes (~15% of the genome) in repair-
for the analysis of ‘forward’ mutations in derepressed deficient mutants after 1000 generations in rich media and
genes. In this study relative reversion rates of each base documented the presence of extensive damage not seen
in a lacZ stop codon were determined, following glucose in wild type under the same conditions. As discussed
starvation and derepression of this gene (including its above, the magnitude and variety of mutations expected
defective codon). There was a good correlation between in nutritionally stressed mutators is expected to be much
the relative number of mutations in the three bases and greater than in cells grown in rich media. Distinguishing
mutation frequencies predicted by the mfg program the multitude of specific, transcription-induced mutations
(Wright et al., 2003; and see below). expected in glucose-starved mutator cells from the

© 2004 Blackwell Publishing Ltd, Molecular Microbiology, 52, 643–650


646 B. E. Wright
genome-wide random mutations resulting from deficien- related to the stress. Examples include phosphate starva-
cies in repair would be impossible. Moreover, the magni- tion (Antelmann et al., 2000; Ishige et al., 2003), amino
tude of specific SDM will be amplified in strains deficient acid starvation (Rudner et al., 1999; Wright et al., 1999),
in repair (see below). superoxide stress (Pomposiello et al., 2001), and osmotic
stress (Wolf et al., 2003).
Reversions of amino acid auxotrophs are convenient
Mechanisms of SDM
models for analysing the specific consequences of
During cell growth, there are many DNA-destabilizing starvation on mutations in related genes (Wright et al.,
events that will increase mutation rates: recombination, 1999; 2003). For example, Fig. 1. depicts mechanisms by
transcription, error-prone DNA polymerases, proofreading which gene derepression can initiate a series of events
errors during DNA replication, and repair-deficient muta- resulting in higher mutation rates in trpA of the trp
tors. In stressed cells, however, when growth is absent or operon. As the absence of tryptophan removes gene
minimized, replication-associated events may not be major repression, transcription is activated in all the biosyn-
sources of mutations compared with SDM. As mentioned thetic trp genes, including the defective trpA– mutant
earlier, the most efficient process for overcoming stress allele 23 (A-to-G) (Wright et al., 2003). Transcription
and accelerating evolution would be the initiation of spe- drives supercoiling (Liu and Wang, 1987), and the DNA
cific feedback mechanisms by which each kind of stress secondary stem-loop structures (SLSs) created by super-
targets hypermutation to those genes that must mutate to coiling can be quantitatively measured (Dayn et al.,
overcome the stress. Although there are a number of 1992). Stem-loop structures probably occur in the wake
examples of specifically directed mutations caused by of the transcription bubble, as the bubble itself contains
artificially induced transcription in growing cells (reviewed too little ssDNA to form predicted structures. Stem-loop
in Wright, 2000), evolution requires mutations directed by structures are sequence-dependent; that is, they form in
stress. To my knowledge, the first example of SDM was supercoiled DNA because of the proximity of inverted
reported by Lipschutz et al. (1965), who examined the complements such as the two 5 nt segments forming the
effect of phosphate starvation on reversion rates of an stem in Fig. 1. Templated mutations are perhaps the
alkaline phosphatase mutant in derepressed compared most convincing evidence for the location of mutable
with repressed genes. Reversion rates in the derepressed bases in SLSs (Ripley and Glickman, 1983). A recent
gene (in the absence of orthophosphate) were 15–20-fold example is the simultaneous mutation of three contigu-
higher than in the repressed gene indicating . . . ‘that the ous bases templated by three unpaired bases in the
susceptibility of . . . bases to chemical changes increases opposing strand of a SLS in the lacZ gene (Wright et al.,
during synthesis of mRNA . . . from an opening of the DNA 2003).
helix or from an uncovering of the gene during derepres- Supercoiling and SLSs are highly localized to the vicin-
sion’. With the advent of gene arrays we now know that ity of each RNA polymerase complex (Rahmouni and
each kind of stress is limited to about 1% of the genome Wells, 1992) and the secondary structures are character-
and specifically derepresses and activates those genes ized by unpaired bases located in a loop or at the base

+
mRNA
RNAP +

_
_
Transcription
A-
G
C

-A
-G-

-
G-C
A-T
G C-G
mutation G-C
T -A
T C trpA allele 23
A C
C T
C

Fig. 1. A model depicting the effect of transcription on mutations. Transcription drives supercoiling which creates and stabilizes SLSs containing
unpaired bases vulnerable to mutation.

© 2004 Blackwell Publishing Ltd, Molecular Microbiology, 52, 643–650


Stress-directed mutations 647
Fig. 2. Mutation frequencies in genes of E. coli
and humans were predicted successfully by the
mfg program.
A. Predicted compared with experimentally
determined reversion rates in the derepressed
E. coli auxotroph, trpA and the non-induced lacI
repressor.
B. Predicted compared with observed mutation
frequencies in hypermutable bases of p53 and
in control bases in nearby codons.

of a stem. Increasing levels of transcription will increase 2003). This correlation is shown for a series of trpA–
supercoiling, the lengths of ssDNA, the size and stability mutant alleles in Fig. 2A and compared with 35 back-
of SLSs (Balke and Gralla, 1987; Figueroa and Bossi, ground mutable bases in the constitutively expressed con-
1988), and thereby increase mutation rates. Unpaired trol gene, lacI.
bases in these secondary structures are vulnerable to As demonstrated by ‘The Unity in Biochemistry’ [first
mutation because of their intrinsic thermodynamic insta- recognized by Kluyver and Donker (1926)], the process of
bility (especially Gs and Cs) and their availability to evolution has bestowed a fundamental unity of biochemi-
nucleotide-altering enzymes (Lindahl, 1993). The inci- cal behaviour on all forms of life: the same metabolites,
dence of mutations in such unpaired bases [including, biosynthetic and catabolic pathways, enzymes and regu-
perhaps, ‘contingency’ loci (Moxon et al., 1994)] would of latory mechanisms. Mutagenesis in microbes and
course be amplified in mutator strains lacking mismatch humans, then, should have much in common. Moreover,
repair (see above). higher rates of transcription are associated with hypermu-
Because extensive evidence indicates that mutable tation in both cancer (Ghosh and Mitchell, 1999) and the
bases in actively transcribed genes of stressed cells are immune response (Bachl et al., 2001), both of which have
unpaired and located in SLSs (reviewed in Wright, 2000), been analysed with the mfg program (Wright et al., 2002;
their mutation rates could be predicted knowing (i) how 2004). The mfg program was used to predict mutation
frequently a base is unpaired in the successive SLSs that frequencies in an unprecedented database of 14 000
contain it during transcription, and (ii) which of those SLSs hypermutable bases of the human p53 tumor suppressor
is the most stable, as that is the one in which the base gene. As seen in Fig. 2B, an excellent correlation was
would be maximally exposed and most likely to mutate. A found for the hypermutable bases as compared to control
new computer algorithm named mfg (http://biol- bases located in nearby codons. Plotting the number of
ogy.dbs.umt.edu/wright/upload/mfg.html) was therefore mutations against –DG alone did not give a statistically
developed to provide this information and to calculate a significant correlation, i.e. both variables in the above
Mutability Index (MI) for each base, defined as the abso- equation were necessary to predict mutation frequencies
lute value of [(percentage of folds in which the base is successfully (Wright et al., 2002). The p53 data are com-
unpaired) (highest –DG of all folds in which the base is pelling in demonstrating that the assumptions underlying
unpaired)] (Wright et al., 2003). Thus, primary importance the mfg program are essentially correct. We conclude that
is given to the most stable SLS in which a given base is mutable bases in stressed cells are in fact unpaired and
unpaired and exposed for the longest period of time; this located in SLSs caused by supercoiling in the wake of the
should be the structure in which the base has the highest transcription bubble. The mutability of such a base will be
probability of mutation. affected by the: (i) per cent of total folds in which the base
The mfg program was used to predict reversion fre- is unpaired during transcription; (ii) stability of the most
quencies in derepressed genes of 15 auxotrophs, and the stable SLS in which the base is unpaired; (iii) intrinsic
correlation was good between MIs and relative mutation thermodynamic instability of each base, and (iv) level of
frequencies determined experimentally (Wright et al., transcription.

© 2004 Blackwell Publishing Ltd, Molecular Microbiology, 52, 643–650


648 B. E. Wright
As DNA sequence and thermodynamics determine the tive control of transcription by Hg-MerR. Nature 355: 87–
first three variables, the two extrinsic variables that can 89.
influence mutability are transcription and supercoiling. As Antelmann, H., Scharf, C., and Hecker, M. (2000) Phosphate
starvation-inducible proteins of Bacillus subtilis: proteomics
discussed above, various environmental stressors can
and transcriptional analysis. J Bacteriol 182: 4478–4490.
enhance mutations directly via transcription and/or super- Bachl, J., Carlson, C., Gray-Schopfer, V., Dessing, M., and
coiling, and epigenetic factors increasing gene expression Olsson, C. (2001) Increased transcription levels induce
are also clearly associated with several tumour models higher mutation rates in a hypermutating cell line. J Immu-
(Ghosh and Mitchell, 1999). The data suggest that, if nol 166: 5051–5057.
stressors cause direct damage to the hypermutable bases Balke, V.L., and Gralla, J.D. (1987) Changes in the linking
analysed in these studies, the resulting increase in muta- number of supercoiled DNA accompany growth transitions
in Escherichia coli. J Bacteriol 169: 4499–4506.
tion frequencies must depend upon, and reflect, the intrin-
Cashel, M., Gentry, D.R., Hernandez, V.J., and Vinella, D.
sic mutabilities (MIs) of these bases. (1996) The stringent response. In Escherichia Coli and
Salmonella: Cellular and Molecular Biology, Vol. 1.
Neidhardt, F.C., Curtis, R. III, Ingraham, J.L., Lin, E.C.C.,
Conclusions
Low, K.B., Magasanik, B., et al., (eds). Washington, DC:
In nature, where cell division must often be negligible as American Society for Microbiology, pp. 1458–1496.
a result of multiple adverse conditions, beneficial muta- Clarke, P.H. (1984) The evolution of degradative pathways.
In Microbial Degradation of Organic Compounds. Gibson,
tions for evolution can arise in specific response to stres-
D.T, (ed.). New York: Marcel Dekker, pp. 11–27.
sors that target related genes for derepression. Specific Cox, E.C., and Gibson, T.C. (1974) Selection for high muta-
transcription of these genes then results in localized DNA tion rates in chemostats. Genetics 77: 169–184.
secondary structures containing unpaired bases vulnera- Dardonville, B., and Raibaud, O. (1990) Characterization of
ble to mutation. Many environmental stressors can also malT mutants that constitutively activate the maltose reg-
affect supercoiling and SDM directly. ulon of Escherichia coli. J Bacteriol 172: 1846–1852.
Background mutation rates in nature remain within a Dayn, A., Malkhosyan, S., and Mirkin, S.M. (1992) Transcrip-
tionally driven cruciform formation in vivo. Nucleic Acids
very narrow range: sufficiently high to adapt to environ-
Res 20: 5991–5997.
mental stress, yet sufficiently low to avoid random damage Death, A., Notley, L., and Ferenci, T. (1993) Derepression of
to the genome (Kimura, 1967; Drake, 1991). This sensi- LamB protein facilitates outer membrane permeation of
tivity is consistent with the low incidence of mutators in carbohydrates into Escherichia coli under conditions of
wild-type populations and with the conclusion that optimal nutrient stress. J Bacteriol 175: 1475–1483.
conditions for stable, long-term evolutionary advances in Delbruck, M., and Bailey, W.T. Jr (1946) Induced mutations
response to stress generally involve minor increases in in bacterial viruses. Cold Spring Harb Symp Quant Biol 11:
33–37.
mutation frequency. The resulting advantage to the organ-
Drake, J.W. (1991) A constant rate of spontaneous mutation
ism may necessarily be minor, in order to avoid rocking in DNA-based microbes. Proc Natl Acad Sci USA 88:
the metabolic boat of complex and finely tuned interac- 7160–7164.
tions that have been selected in the past and must be Dykhuizen, D.E., and Hartl, D.L. (1983) Selection in chemo-
maintained during further evolution. stats. Microbiol Rev 47: 150–168.
Figueroa, N., and Bossi, L. (1988) Transcription induces gyra-
tion of the DNA template in Escherichia coli. Proc Natl
Acknowledgements Acad Sci USA 85: 9416–9420.
Foster, P.L., and Trimarchi, J.M. (1995) Adaptive reversion of
I thank Arnold Demain, Scott Samuels, Frank Rosenzweig,
an episomal frameshift mutation in Escherichia coli
Jacqueline Reimers and Karen Schmidt for valuable discus-
requires conjugal functions but not actual conjugation. Proc
sions and suggestions, and Maryann Hubbard for manuscript
Natl Acad Sci USA 92: 5487–5490.
preparation. This work was supported by National Institutes
Funchain, P., Yeung, A., Stewart, J.L., Lin, R., Slupska, M.M.,
of Health grants R15CA88893 and R55CA99242 and the
and Miller, J.H. (2000) The consequences of growth of a
Stella Duncan Memorial Research Institute.
mutator strain of Escherichia coli as measured by loss of
function among multiple gene targets and loss of fitness.
References Genetics 154: 959–970.
Gentry, D.R., Hernandez, V.J., Nguyen, L.H., Jensen, D.B.,
Abril, M.A., Michan, C., Timmis, K.N., and Ramos, J.L. (1989) and Cashel, M. (1993) Synthesis of the stationary-phase
Regulator and enzyme specificities of the TOL plasmid- sigma factor ss is positively regulated by ppGpp. J Bacteriol
encoded upper pathway for degradation of aromatic 175: 7982–7989.
hydrocarbons and expansion of the substrate range of the Ghosh, R., and Mitchell, D.L. (1999) Effect of oxidative DNA
pathway. J Bacteriol 171: 6782–6790. damage in promoter elements on transcription factor bind-
Ansari, A.Z., Chael, M.L., and O’Halloran, T.V. (1992) ing. Nucleic Acids Res 27: 3213–3218.
Allosteric underwinding of DNA is a critical step in posi- Giraud, A., Matic, I., Tenaillon, O., Clara, A., Radman, M.,

© 2004 Blackwell Publishing Ltd, Molecular Microbiology, 52, 643–650


Stress-directed mutations 649
Fons, M., and Taddei, F. (2001) Costs and benefits of high Notley-McRobb, L., and Ferenci, T. (1999a) The generation
mutation rates: adaptive evolution of bacteria in the mouse of multiple co-existing mal-regulatory mutations through
gut. Science 291: 2606–2608. polygenic evolution in glucose-limited populations of
Hegeman, G.D., and Rosenberg, S.L. (1970) The evolution Escherichia coli. Environ Microbiol 1: 45–52.
of bacterial enzyme systems. Annu Rev Microbiol 24: 429– Notley-McRobb, L., and Ferenci, T. (1999b) Adaptive mgl-
462. regulatory mutations and genetic diversity evolving in
Hengge-Aronis, R. (1996) Regulation of gene expression glucose-limited Escherichia coli populations. Environ
during entry into stationary phase. In Escherichia Coli and Microbiol 1: 33–43.
Salmonella: Cellular and Molecular Biology, Vol. 1. Notley-McRobb, L., Seeto, S., and Ferenci, T. (2003) The
Neidhardt, F.C., Curtis, R. III, Ingraham, J.L., Lin, E.C.C., influence of cellular physiology on the initiation of muta-
Low, K.B., Magasanik, B., et al., (eds). Washington, DC: tional pathways in Escherichia coli populations. Proc R Soc
American Society for Microbiology. Lond B Biol Sci 270: 843–848.
Higgins, C.F., Dorman, C.J., Stirling, D.A., Waddell, L., Pomposiello, P.J., Bennik, M.H., and Demple, B. (2001)
Booth, I.R., May, G., and Bremer, E. (1988) A physiological Genome-wide transcriptional profiling of the Escherichia
role for DNA supercoiling in the osmotic regulation of gene coli responses to superoxide stress and sodium salicylate.
expression in S. typhimurium and E. coli. Cell 52: 569–584. J Bacteriol 183: 3890–3902.
Ishige, T., Krause, M., Bott, M., Wendisch, V.F., and Sahm, Prival, M.J., and Cebula, T.A. (1992) Sequence analysis of
H. (2003) The phosphate starvation stimulon of Coryne- mutations arising during prolonged starvation of Salmo-
bacterium glutamicum determined by DNA microarray nella typhimurium. Genetics 132: 303–310.
analyses. J Bacteriol 185: 4519–4529. Rahmouni, A.R., and Wells, R.D. (1992) Direct evidence for
Kasak, L., Horak, R., and Kivisaar, M. (1997) Promoter-cre- the effect of transcription on local DNA supercoiling in vivo.
ating mutations in Pseudomonas putida: a model system J Mol Biol 223: 131–144.
for the study of mutation in starving bacteria. Proc Natl Ramos, J.L., Wasserfallen, A., Rose, K., and Timmis, K.N.
Acad Sci USA 94: 3134–3139. (1987) Redesigning metabolic routes: manipulation of TOL
Kimura, M. (1967) On the evolutionary adjustment of spon- plasmid pathway for catabolism of alkylbenzoates. Science
taneous mutation rates. Genet Res Camb 9: 23–43. 235: 593–596.
Kluyver, A.J., and Donker, H.J.L. (1926) Die Einheit in der Ripley, L.S., and Glickman, B.W. (1983) Unique self-
Biochemie. Chem d Zelle u Gew 13: 134–190. complementarity of palindromic sequences provides DNA
Lederberg, J. (1951) In Genetics in the 20th Century. Dunn, structural intermediates for mutation. Cold Spring Harb
L.C., (ed.). New York: Macmillan, pp. 263. Symp Quant Biol 47 Part 2: 851–861.
Lefstin, J.A., and Yamamoto, K.R. (1998) Allosteric effects of Rosenberg, S.M., Longerich, S., Gee, P., and Harris, R.S.
DNA on transcriptional regulators. Nature 392: 885–888. (1994) Adaptive mutation by deletions in small mononucle-
Lerner, S.A., Wu, T.T., and Lin, E.C.C. (1964) Evolution of a otide repeats. Science 265: 405–407.
catabolic pathway in bacteria. Science 146: 1313–1315. Rudner, R., Murray, A., and Huda, N. (1999) Is there a link
Lin, E.C.C., Hacking, A.J., and Aguilar, J. (1976) Experimen- between mutation rates and the stringent response in
tal models of acquisitive evolution. Bioscience 26: 548– Bacillus subtilis? Ann N Y Acad Sci 870: 418–422.
555. Saier, M.H., Ramseier, T.M., and Reizer, J. (1996) Regula-
Lindahl, T. (1993) Instability and decay of the primary struc- tion of carbon utilization. In Escherichia Coli and Salmo-
ture of DNA. Nature 362: 709–715. nella: Cellular and Molecular Biology, Vol. 1. Neidhardt,
Lipschutz, R., Falk, R., and Avigad, G. (1965) Mutation rates F.C., Curtis, R. III, Ingraham, J.L., Lin, E.C.C., Low, K.B.,
of the induced and repressed locus of alkaline phos- Magasanik, B., et al. (eds). Washington, DC: American
phatase in Escherichia coli. Israel J Med Sci 1: 323–324. Society for Microbiology, pp. 1325–1343.
Liu, L.F., and Wang, J.C. (1987) Supercoiling of the DNA Sung, H.M., and Yasbin, R.E. (2002) Adaptive, or
template during transcription. Proc Natl Acad Sci USA 84: stationary-phase, mutagenesis, a component of bacterial
7024–7027. differentiation in Bacillus subtilis. J Bacteriol 184: 5641–
Massey, R.C., Rainey, P.B., Sheehan, B.J., Keane, O.M., 5653.
and Dorman, C.J. (1999) Environmentally constrained Torriani, A., and Rothman, F. (1961) Mutants of Escherichia
mutation and adaptive evolution in Salmonella. Curr Biol 9: coli gene essential for survival in stationary phase. J Bac-
1477–1480. teriol 81: 835–840.
van der Meer, J.R., de Vos, W.M., Harayama, S., and Weismann, A. (1893) Uber die Vererbung. Jena, Germany:
Zehnder, A.J. (1992) Molecular mechanisms of genetic Gustav Fischer.
adaptation to xenobiotic compounds. Microbiol Rev 56: Wolf, A., Kramer, R., and Morbach, S. (2003) Three pathways
677–694. for trehalose metabolism in Corynebacterium glutamicum
Mortlock, R.P. (1984) Microorganisms as Model Systems for ATCC13032 and their significance in response to osmotic
Studying Evolution. New York, USA: Plenum Press. stress. Mol Microbiol 49: 1119–1134.
Moxon, E.R., Rainey, P.B., Nowak, M.A., and Lenski, R.E. Wright, B.E. (2000) A biochemical mechanism for nonrandom
(1994) Adaptive evolution of highly mutable loci in patho- mutations and evolution. J Bacteriol 182: 2993–3001.
genic bacteria. Curr Biol 4: 24–33. Wright, B.E., Longacre, A., and Reimers, J.M. (1999) Hyper-
Nochur, S.V., Roberts, M.F., and Demain, A.L. (1990) Muta- mutation in derepressed operons of Escherichia coli K-12.
tion of Clostridium thermocellum in the presence of certain Proc Natl Acad Sci USA 96: 5089–5094.
carbon sources. FEMS Microbiol Lett 71: 199–204. Wright, B.E., Reimers, J.M., Schmidt, K.H., and Reschke,

© 2004 Blackwell Publishing Ltd, Molecular Microbiology, 52, 643–650


650 B. E. Wright
D.K. (2002) Hypermutable bases in the p53 cancer gene stem-loop structures of derepressed genes: implications
are at vulnerable positions in DNA secondary structures. for evolution. Mol Microbiol 48: 429–441.
Cancer Res 62: 5641–5644. Wright, B.E., Schmidt, K.H., and Minnick, M.F. (2004) Mech-
Wright, B.E., Reschke, D.K., Schmidt, K.H., Reimers, J.M., anisms by which transcription can regulate somatic hyper-
and Knight, W. (2003) Predicting mutation frequencies in mutation. Genes Immun 5 (in press).

© 2004 Blackwell Publishing Ltd, Molecular Microbiology, 52, 643–650


REPORTS
References and Notes 11. J. W. Allen, R. M. Martin, Phys. Rev. Lett. 49, 1106 19. P. Monthoux, G. G. Lonzarich, in preparation (avail-
1. N. D. Mathur et al., Nature 394, 39 (1998). (1982). able at http://xxx.lanl.gov/abs/cond-mat/0309551).
2. R. Movshovich et al., Phys. Rev. B 53, 8241 (1996). 12. A. Onodera et al., Solid State Commun. 123, 113 20. O. Stockert et al., Acta Phys. Pol. B 34, 963 (2003).
3. H. Hegger et al., Phys. Rev. Lett. 84, 4986 (2000). (2002). 21. We thank P. Gegenwart, G. G. Lonzarich, K. Miyake, P.
4. F. Steglich et al., Phys. Rev. Lett. 43, 1892 (1979). 13. Materials and methods are available as supporting Monthoux, J. A. Mydosh, O. Stockert, and H. Wilhelm
5. P. Gegenwart et al., Phys. Rev. Lett. 81, 1501 (1998). material on Science Online. for helpful discussions and S. Mederle for assistance
6. F. Thomas, J. Thomasson, C. Ayache, C. Geibel, F. 14. H. Q. Yuan, unpublished data. in some aspects of the high-pressure work.
Steglich, Physica B 186 –188, 303 (1993). 15. E. Vargoz, D. Jaccard, J. Y. Genoud, J.-P. Brison, J.
7. D. Jaccard, H. Wilhelm, K. Alami-Yadri, E. Vargoz, Flouquet, Solid State Commun. 106, 631 (1998). Supporting Online Material
Physica B 259 –261, 1 (1999). 16. J. Thomasson, Y. Okayama, I. Sheikin, J.-P. Brison, www.sciencemag.org/cgi/content/full/302/5653/2104/
8. A. T. Holmes, A. Demuer, D. Jaccard, Acta Phys. Pol. B. D. Braithwaite, Solid State Commun. 106, 637 DC1
34, 567 (2003). (1998). Materials and Methods
9. B. Bellarbi, A. Benoit, D. Jaccard, J. M. Mignot, H. F. 17. Y. Onishi, K. Miyake, J. Phys. Soc. Jap. 69, 3955 (2001). References and Notes
Braun, Phys. Rev. B 30, 1182 (1984). 18. A. Holmes, D. Jaccard, K. Miyake, in preparation (avail-
10. H. Q. Yuan et al., Acta Phys. Pol. B 34, 533 (2003). able at http://xxx.lanl.gov/abs/cond-mat/0306054). 18 September 2003; accepted 18 November 2003

Adaptation Limits Diversification niche 1 (I ⬎ 0.5) to diversify (i.e., for a


mutant specializing in niche 2 to be able to

of Experimental Bacterial invade and be maintained). For a variety of


different functional forms of Wi, we find that

Downloaded from http://science.sciencemag.org/ on April 12, 2017


the better adapted the population to niche 1
Populations (increasing I), the greater the phenotypic ef-
fect of a mutation required for invasion of
Angus Buckling,1* Matthew A. Wills,1 Nick Colegrave2 niche 2 (Fig. 1). If the probability of a muta-
tion is inversely proportional to its phenotyp-
Adaptation to a specific niche theoretically constrains a population’s ability to ic effect, then the specialization will make
subsequently diversify into other niches. We tested this theory using the diversification less likely.
bacterium Pseudomonas fluorescens, which diversifies into niche specialists We tested the hypothesis that adaptation
when propagated in laboratory microcosms. Numerically dominant genotypes within an existing niche in rugged fitness land-
were allowed to diversify in isolation. As predicted, populations increased in scapes constrains diversification using the com-
fitness through time but showed a greatly decreased ability to diversify. Sub- mon plant-colonizing bacterium Pseudomonas
sequent experiments demonstrated that niche generalists and reductions in fluorescens (8). Rejection of the hypothesis
intrinsic evolvability were not responsible for our data. These results show that would imply that the landscape is not very rug-
niche specialization may come with a cost of reduced potential to diversify. ged relative to the magnitude of mutational ef-
fect. When propagated in spatially structured
Despite much theoretical (1–6) and empirical fect, will be required for mutants to be com- environments (a static glass bottle containing
(7–14) work investigating the conditions pro- petitive when ascending an alternative peak, nutrient-rich medium), isogenic P. fluorescens
moting the evolution of sympatric (within- reducing the potential to diversify. populations rapidly diversify, generating numer-
population) diversity, little is known about Consider an environment with two avail- ous niche specialist genotypes that are readily
the constraints. Here, we demonstrate that able niches (1 and 2) with the population distinguished by their (heritable) colony mor-
adaptation itself is likely to limit a popula- being regulated at the level of the niche, such phologies on agar plates (8). The genotypes can
tion’s ability to diversify, when evolution that half the population derives from each be grouped into three distinct classes on the
occurs in “rugged fitness landscapes.” Fitness niche. The fitness of an individual in each basis of colony morphology and niche occupa-
landscapes are created by plotting fitness niche (Wi) depends on the value of a pheno- tion. Smooth (SM) morphs resemble the ances-
against all possible genotypes within the pop- typic trait (I), which can vary between 0 and tral genotype and largely inhabit the same, liq-
ulation (4–6, 15, 16). Assuming large popu- 1, and there is a trade-off between fitness in uid-phase, niche; wrinkly-spreader (WS)
lations, selection drives populations uphill each niche such that W1 ⫽ f (I ) and W2 ⫽ morphs form a mat at the air-broth interface, and
toward fitness peaks (adaptation). If several f (1 ⫺ I ). Using Levene’s classic model of the much less frequent fuzzy-spreader (FS)
ecological niches exist and organisms show species coexistence (1), we can consider the morphs colonize the harsher, less aerobic bot-
fitness trade-offs between niches (niche spe- size of the phenotypic (mutational) change tom of the vials. Fitness trade-offs between
cialization), or genetic interactions (epistasis) required for a population partially adapted to niches have been demonstrated (8, 17) by the
are a major determinant of fitness, the fitness
landscape may be rugged, with multiple Fig. 1. Magnitude of mutational ef-
peaks and valleys (4–6, 15, 16). When a fect required for adaptation to a
population is poorly adapted and in a fitness new niche, hence diversification, as
valley, multiple peaks can be ascended, hence a function of adaptation to niche 1
the potential for high rates of diversification. (population phenotype). Different
lines show different relationships
However, as the population ascends a peak, between phenotype (I) and fit-
mutations of increasingly large effect, or a ness in niche 1 (W1): W1 ⫽ I (F);
number of concurrent mutations of small ef- W1 ⫽ I 2 (‘); W1 ⫽ I 0.5 (E).
Fitness functions in niche 2 (W2) are
the same as in niche 1, but with I
1
Department of Biology and Biochemistry, University replaced with 1 ⫺ I.
of Bath, Bath BA2 7AY, UK. 2Institute of Cell, Animal
and Population Biology, University of Edinburgh,
King’s Buildings, Edinburgh EH9 3JT, UK.
*To whom correspondence should be addressed. E-
mail: bssagjb@bath.ac.uk

www.sciencemag.org SCIENCE VOL 302 19 DECEMBER 2003 2107


REPORTS
operation of negative frequency-dependent se- suites of novel genotypes when they were al- ferring 1% of the complete culture to fresh
lection; genotypes have a fitness advantage lowed to diversify in replicate (22), suggesting media, rather than individual genotypes. We
when rare because there is less intense compe- that the reduction in diversification is due, to found no evidence of diversity decreasing
tition within their niche (18, 19). some extent, to a deterministic loss in accessi- through time, refuting generalist evolution as
To investigate whether adaptation within ble genotypes during the climbing of a partic- the main explanation for our data (sign test
existing niches constrained diversifications, we ular adaptive peak. of correlations between number of genotypes
initiated six P. fluorescens populations with a Our proposed explanation for this de- and transfer number for the six lines:
single clone [SBW25 (20)] and propagated creasing ability to diversify (with increasing P ⬎ 0.2).
them for a week in spatially heterogeneous transfer number) is niche-specific adaptation. A second explanation is an intrinsic re-
environments. Diversity was determined from We tested this hypothesis by competing all duction in evolvability (for example, reduced
colony morphologies on agar plates (8, 10, 17, genotypes from all time points with a genet- mutation rates) through time, possibly as a
21), and the numerically most dominant SM- ically marked isogenic mutant of the ances- result of genetic drift caused by our selection
like genotype in each population was isolated tral genotype in the selective environment regime. Such intrinsic reductions are likely to
and used to initiate a single subsequent culture, (22). Fitness of genotypes did indeed increase be apparent in all environments; hence, the
which was propagated as above. This procedure through time in all replicates (Fig. 2B) same relationships between time spent evolv-
was repeated five times for each of the six (paired t test of ancestral versus transfer 6 ing and diversification should be apparent in
replicate lines, resulting in one genotype from selection rate constant: t ⫽ 15.27, P ⬍ a novel environment. Conversely, if ascend-
each of six transfers for each line (a total of 36 0.0001; sign test of correlations between fit- ing local fitness peaks are responsible for the

Downloaded from http://science.sciencemag.org/ on April 12, 2017


evolved clones) (22). Consistent with the ness and transfer number for the six lines: inability to diversify (our hypothesis), diver-
evolved genotypes occupying the SM niche, the P ⫽ 0.03). Furthermore, there was an overall sification in a novel environment should be
six clones from the final transfer were able to negative correlation between fitness and abil- independent of adaptation to the selective
competitively exclude ancestral SM (22). In all ity to diversify (sign test of correlations be- environment: All genotypes are equally un-
lines, diversification after six transfers was tween fitness and number of genotypes for likely to be approaching fitness peaks in the
much less than ancestral diversification, and the six lines: P ⫽ 0.03). fitness landscape created by the novel envi-
there was an overall negative correlation be- It is possible that the inability to diversify ronment. To examine this hypothesis, we
tween transfer number and ability to diversify is the result of types evolving that are supe- measured the ability of each of the genotypes
(Fig. 2A) (sign test of correlations between rior in all niches (23). If this were the case, to diversify in two novel environments: in
number of genotypes and transfer number for we would expect diversity to decrease (as a spatially homogeneous environments (shaken
the six lines: P ⫽ 0.03). Different colony types result of the evolution of generalists) under microcosms) and in spatially heterogeneous
dominated in different populations, suggesting conditions in which the selection regime in- environments (static microcosms) with low
the ascent of different adaptive peaks. Howev- volves propagation of a sample of the whole nutrients (22). Diversification of the ancestral
er, individual evolved genotypes (that showed population. We evolved six populations un- genotype was considerably less in both envi-
little diversification) produced almost identical der identical conditions as above, but trans- ronments than in the original environment

Fig. 2. Diversification and fit-


ness through time. (A) The
ability of genotypes to diver-
sify and (B) their fitness
(selection rate constant)
against a standard competi-
tor, where a value of zero in-
dicates equal performance of
the two competing geno-
types, after 1 week of growth
in static KB media micro-
cosms, as a function of trans-
fer number. Different lines
and symbols represent the six
replicate lines.

Fig. 3. Diversification in novel


environments. The ability of
genotypes to diversify in two
novel environments: (A) shak-
en KB media microcosms and
(B) static minimal media mi-
crocosms as a function of
transfer number. Different
lines and symbols represent
the six replicate lines.

2108 19 DECEMBER 2003 VOL 302 SCIENCE www.sciencemag.org


REPORTS
(compare Figs. 2A and 3). Nevertheless, in 25. D. Schluter, The Ecology of Adaptive Radiations (Ox- Supporting Online Material
both environments there was no relation be- ford Univ. Press, Oxford, 2000). www.sciencemag.org/cgi/content/full/302/5653/2107/DC1
26. We thank L. Hurst and three anonymous referees for Materials and Methods
tween ability to diversify and time spent comments on the manuscript. This work was funded References
evolving in the original high-nutrient media by the Royal Society and Natural Environment Re-
(Fig. 3) (sign tests of correlations between search Council (UK). 7 July 2003; accepted 14 October 2003
number of genotypes and transfer number for
the six lines: P ⬎ 0.2 in both cases).
Adaptation can limit the ability of bac-
terial genotypes to diversify genetically. Melt Inclusions in Veins: Linking
This was not the result of generalist evolu-
tion or the evolution of an intrinsic reduc- Magmas and Porphyry Cu Deposits
tion in evolvability, but was caused by
environment-specific adaptation. Given the Anthony C. Harris,1* Vadim S. Kamenetsky,1 Noel C. White,1
strong empirical support for both the im- Esmé van Achterbergh,2 Chris G. Ryan2
portance of environmental heterogeneity in
diversification (7–14) and epistasis (6, 24), At a porphyry copper-gold deposit in Bajo de la Alumbrera, Argentina, silicate-
it is likely that rugged fitness landscapes, a melt inclusions coexist with hypersaline liquid- and vapor-rich inclusions in the
requirement for the observed effects, are earliest magmatic-hydrothermal quartz veins. Copper concentrations of the

Downloaded from http://science.sciencemag.org/ on April 12, 2017


common. These results are therefore likely hypersaline liquid and vapor inclusions reached maxima of 10.0 weight % (wt
to be generally relevant and may help to %) and 4.5 wt %, respectively. These unusually copper-rich inclusions are
explain patterns of diversity over both mi- considered to be the most primitive ore fluid found thus far. Their preservation
cro- and macroevolutionary time scales. with coexisting melt allows for the direct quantification of important ore-
Consistent with recent interpretations of forming processes, including determination of bulk partition coefficients of
macroevolutionary adaptive radiations metals from magma into ore-forming magmatic volatile phases.
(25), we predict that in environments that
can potentially support similar levels of In porphyry ore deposits, metals are con- Bajo de la Alumbrera is an Au-rich porphyry
diversity, diversification is more likely to centrated by large volumes of magmatic Cu deposit where potassic alteration assemblag-
occur immediately following colonization volatiles exsolved from crystallizing upper es overprint several phases of porphyritic dacite
of the environment than through expansion crustal magma bodies (1). Whether an ore intrusions and are associated with the bulk of the
into new niches within the environment deposit forms depends on the availability of disseminated Cu-Fe sulfides and Au. High tem-
after an extinction event. metals in the magma, the partitioning of perature (maximum of 750°C) and saline fluid
those metals into the volatile phase, and the (⬎35 wt % equivalent NaCl) of magmatic ori-
References and Notes history of the fluid after release from the gin (as inferred from the calculated ␦18O and ␦D
1. H. Levene, Am. Nat. 87, 331 (1953). magma (2– 4 ). Ores are typically associated compositions) formed these alteration assem-
2. P. W. Hedrick, M. E. Ginevan, E. P. Ewing, Annu. Rev. with hydrothermal mineral assemblages blages (8). Some of the earliest Cu-Fe sulfides
Ecol. Syst. 7, 1 (1976).
3. P. W. Hedrick, Annu. Rev. Ecol. Syst. 17, 535 (1986).
produced by the interaction of magmatic occur in diffuse quartz veins, which are textur-
4. T. Dobzhansky, Genetics and the Origin of Species fluids with wall rocks (5). The earliest ally similar to those described as A veins (9);
(Columbia Univ. Press, New York, 1937). formed hydrothermal alteration of wall however, the presence of silicate-melt inclusions
5. M. C. Whitlock, P. C. Phillips, F. B. G. Moore, S. J. rocks is potassic (biotite-K-feldspar-quartz warrants a new vein subclass. Hereafter we refer
Tonsor, Annu. Rev. Ecol. Syst. 26, 601 (1995).
6. M. J. Wade, in Epistasis and the Evolutionary Process, with or without magnetite assemblage) and to them as P veins, reflecting their primitive role
J. B. Wolf, E. D. Brodie, M. J. Wade, Eds. (Oxford Univ. is caused by high temperature (350° to in the evolution of the magmatic-hydrothermal
Press, Oxford, 2000), pp. 213–231. 800°C) and saline (up to 70 weight % system. Typically, they consist of sugary quartz,
7. R. Korona, C. H. Nakatsu, L. J. Forney, R. E. Lenski,
Proc. Natl. Acad. Sci. U.S.A. 91, 9037 (1994).
equivalent NaCl) fluids (6, 7 ). Discovery with lesser amounts of K-feldspar and with or
8. P. B. Rainey, M. Travisano, Nature 394, 69 (1998). of silicate-melt inclusions coexisting with without hornblende-biotite-magnetite-chalcopy-
9. D. E. Rozen, R. E. Lenski, Am. Nat. 155, 24 (2000). fluid inclusions in magmatic-hydrothermal rite (CuFeS2) and pyrite.
10. A. Buckling, P. B. Rainey, Nature 420, 496 (2002). quartz veins unambiguously links devola- The P veins contain silicate-melt inclusions
11. D. Schluter, Am. Nat. 157 (suppl.), S4 (2000).
12. S. M. Vamosi, D. Schluter, Proc. R. Soc. London Ser. B tilization of the magma with the associated that consist of silicate crystals, vapor bubbles,
269, 923 (2002). porphyry ore deposit. This occurrence pre- salt crystals, and opaque oxide and sulfide crys-
13. R. F. Rosenzweig, R. R. Sharp, D. S. Treves, J. Adams, serves the most primitive magmatic vola- tals (Fig. 1). These inclusions are similar to
Genetics 137, 903 (1994).
14. R. Kassen, J. Evol. Biol. 15, 173 (2002).
tiles and the melt from which these were those in magmatic quartz phenocrysts in the
15. S. Wright, Genetics 16, 97 (1931). derived and, with the use of advanced mi- mineralized intrusions (10). Heating experi-
16. H. J. Muller, Biol. Rev. Camb. Philos. Soc. 14, 261 croanalysis techniques, allows chemical ments (11) on silicate-melt inclusions revealed
(1939).
changes to be traced through the evolution consistent phase transformations: The dissolu-
17. A. Buckling, R. Kassen, G. Bell, P. B. Rainey, Nature
408, 961. (2000). of the hydrothermal system. Moreover, we tion of the salt phases (e.g., halite and sylvite)
18. F. J. Ayala, C. A. Campbell, Annu. Rev. Ecol. Syst. 5, are able to use those data to quantify the occurs between 105° and 560°C; dissolution of
115 (1974). magmatic-hydrothermal processes that lead crystalline silicate phases occurs between 650°
19. M. L. Rosenzweig, Species Diversity in Space and Time
(Cambridge Univ. Press, Cambridge, 1995). to the formation of porphyry Cu deposits. and 765°C. After heating the host quartz crys-
20. P. B. Rainey, M. J. Bailey, Mol. Microbiol. 19, 521 (1996). tals to 800°C at 1 atm external pressure for
21. R. Kassen, A. Buckling, G. Bell, P. B. Rainey, Nature 1
Centre for Ore Deposit Research, University of several hours, the sample was quenched to pro-
406, 508 (2000). Tasmania, Private Bag 79, Hobart, Tasmania 7001,
22. Materials and methods are available as supporting
duce a silicate glass, which is extremely rich in
Australia. 2Commonwealth Scientific and Industrial
material on Science Online. Research Organization Exploration and Mining, Post
K2O compared to whole rock analyses and has
23. G. Bell, Selection: The Mechanism of Evolution (Chap- Office Box 136, North Ryde, New South Wales a composition similar to K-feldspar (table
man & Hall, London, 1997). 1670, Australia. S2). This composition may represent a che-
24. A. R. Templeton, in Epistasis and the Evolutionary
Process, J. B. Wolf, E. D. Brodie, M. J. Wade, Eds. *To whom correspondence should be addressed. E- mically modified melt trapped in a dominantly
(Oxford Univ. Press, Oxford, 2000). mail: A.Harris@utas.edu.au aqueous environment or may contain ex-

www.sciencemag.org SCIENCE VOL 302 19 DECEMBER 2003 2109


Adaptation Limits Diversification of Experimental Bacterial
Populations
Angus Buckling, Matthew A. Wills and Nick Colegrave (December 18,
2003)
Science 302 (5653), 2107-2109. [doi: 10.1126/science.1088848]

Editor's Summary

This copy is for your personal, non-commercial use only.

Downloaded from http://science.sciencemag.org/ on April 12, 2017


Article Tools Visit the online version of this article to access the personalization and article
tools:
http://science.sciencemag.org/content/302/5653/2107

Permissions Obtain information about reproducing this article:


http://www.sciencemag.org/about/permissions.dtl

Science (print ISSN 0036-8075; online ISSN 1095-9203) is published weekly, except the last week in
December, by the American Association for the Advancement of Science, 1200 New York Avenue NW,
Washington, DC 20005. Copyright 2016 by the American Association for the Advancement of Science;
all rights reserved. The title Science is a registered trademark of AAAS.
REVIEW ARTICLE

The role of variable DNA tandem repeats in bacterial


adaptation
Kai Zhou, Abram Aertsen & Chris W. Michiels
Department of Microbial and Molecular Systems (M²S), Faculty of Bioscience Engineering, Laboratory of Food Microbiology and Leuven Food
Science and Nutrition Research Centre (LFoRCe), KU Leuven, Leuven, Belgium

Correspondence: Chris W. Michiels, Abstract


Laboratory of Food Microbiology, Kasteelpark
Arenberg 23, B-3001 Leuven, Belgium. DNA tandem repeats (TRs), also designated as satellite DNA, are inter- or
Tel.: +32 16 321578; intragenic nucleotide sequences that are repeated two or more times in a head-
fax: +32 16 321960; to-tail manner. Because TR tracts are prone to strand-slippage replication and
e-mail: chris.michiels@biw.kuleuven.be recombination events that cause the TR copy number to increase or decrease,
loci containing TRs are hypermutable. An increasing number of examples illus-
Received 17 April 2013; revised 13 July
trate that bacteria can exploit this instability of TRs to reversibly shut down or
2013; accepted 26 July 2013. Final version
published online 28 August 2013.
modulate the function of specific genes, allowing them to adapt to changing
environments on short evolutionary time scales without an increased overall
DOI: 10.1111/1574-6976.12036 mutation rate. In this review, we discuss the prevalence and distribution of
inter- and intragenic TRs in bacteria and the mechanisms of their instability.
Editor: Grzegorz Wegrzyn In addition, we review evidence demonstrating a role of TR variations in bacte-
rial adaptation strategies, ranging from immune evasion and tissue tropism to
Keywords the modulation of environmental stress tolerance. Nevertheless, while bioinfor-
polymorphic DNA; contingency loci; host–
matic analysis reveals that most bacterial genomes contain a few up to several
pathogen interaction; stress tolerance;
dozens of intra- and intergenic TRs, only a small fraction of these have been
evolution; phase variation.
functionally studied to date.
MICROBIOLOGY REVIEWS

the latter strategy is that random mutations are more


Introduction
often deleterious than beneficial.
To cope with rapidly changing environmental conditions Interestingly, neither the type nor the frequency of
and ensure their survival, unicellular organisms have mutational events is randomly distributed over the
evolved a plethora of adaptation strategies (Aertsen & genome, and some DNA sequences have evolved to be
Michiels, 2004, 2005). Most of these strategies are based mutational hotspots that drive the variability of genes
on transient alterations in gene expression in response to whose activity can impact the adaptive potential of their
stressful conditions, and well-known examples include the host. One type of such special sequences that is very
SOS response (controlled by RecA and LexA), the general abundant in prokaryotic and eukaryotic genomes is
stress response (regulated by the sigma factor RpoS), the known as tandem repeats (TRs), a major class of direct
stringent response (mediated by pppGpp and ppGpp), DNA repeats. While at first TRs were considered to be
and the heat shock response (mainly controlled by RpoH; junk DNA without any biological function, studies of the
Massey & Buckling, 2002; Foster, 2005; Saint-Ruf & human genome have revealed some of these repetitive
Matic, 2006; Foster, 2007; Jolivet-Gougeon et al., 2011). sequences to be hypermutable and the cause of diseases
In addition, adaptation can also stem from the acquisi- such as fragile X syndrome, spinobulbar muscular atro-
tion of stochastic mutations that alter the genotype, phy, and huntington disease (Hannan, 2010). In addition,
which become positively selected and fixed in a popula- accumulating evidence points out the potential role of
tion if they coincide with a beneficial phenotype (Rando TRs as engines of genetic variability and bacterial
& Verstrepen, 2007). However, an important drawback of adaptation. In this review, we therefore focus on the

FEMS Microbiol Rev 38 (2014) 119–141 ª 2013 Federation of European Microbiological Societies.
Published by John Wiley & Sons Ltd. All rights reserved
120 K. Zhou et al.

identification and distribution of TRs in bacteria, the their evolution. A variety of algorithms have been devel-
mechanisms behind their variability, and their biological oped for detecting TRs, but it is important to be aware
significance in bacterial adaptation. that these may differ in their ability to detect different
types of TRs (Merkel & Gemmell, 2008; Treangen et al.,
2009; Kajava, 2012). Hence, the choice of search tool
Identification and distribution of TRs in
should be determined by the TR type of interest, or the
bacterial genomes
parallel use of several algorithms is advisable when a wide
screen for TRs is performed. Furthermore, parameter set-
Definition of TRs
tings (i.e. alignment weights, definition of repeats, and
TRs are nucleotide sequences that are directly repeated in threshold scores) can also strongly affect outcome in terms
a head-to-tail manner. According to the conservation of of number and consensus motif of TRs (Lim et al., 2013).
the repeated sequence, TRs are classified as identical/per- In particular, problems are still commonly encountered in
fect TRs or degenerated/imperfect TRs, respectively detecting imperfect TRs (Leclercq et al., 2007; Schaper
(Fig. 1). Furthermore, TRs are commonly classified into et al., 2012). Several algorithms are freely available
three categories according to their repeat unit size, online, such as Tandem Repeat Finder (Benson, 1999) and
although there is no consensus definition of these catego- IMEx (Mudunuri & Nagarajaram, 2007). In addition,
ries (Richard et al., 2008). Repeats with unit size varying several databases of annotated TRs in prokaryotes have
from 1 to 9, 10 to 100, and > 100 bp are termed micro- been established, such as TRs DB (http://minisatellites.
satellites, minisatellites, and macrosatellites, respectively u-psud.fr), PSSRDb (http://pssrdb.cdfd.org.in) and
(Lopes et al., 2006). The term ‘satellite DNA’ originally MICAS (http://180.149.48.108/micas/index.php). In the
refers to the very large arrays of tandemly repeated non- next section, we will review the major findings from some
coding DNA (often hundreds of copies) that are charac- recent in silico studies of the genomic distribution of TRs
teristic of large eukaryotic genomes, but, in the context of in bacteria.
bacterial genomes, is also used to include small and intra-
genic TRs.
The distribution of TRs in bacterial genomes
The analysis of TRs in bacterial genomes so far has
In silico identification of TRs
mainly focused on microsatellites with unit size 1–6 bp,
The increasing availability of genome sequences and also termed ‘simple sequence repeats (SSRs)’. A number
specialized bioinformatics software greatly facilitates the of general observations regarding the distribution of SSRs
search and identification of TR loci on a genomewide can be stated. First of all, the abundance of SSRs in bacte-
scale, which obviously is a prerequisite for understanding ria is lower than that in eukaryotes (Schlotterer et al.,
their distribution, predicting their function, and tracking 2006). Nevertheless, the number of SSRs is orders of

(a)
IdenƟcal / Perfect TRs AGCTG AGCTG AGCTG AGCTG AGCTG
(unit sequence = 100% conserved)

Degenerated / Imperfect TRs AGCTG TGCTG AGGTG AGCTG AGCTC


(unit sequence < 100% conserved)

(b) Microsatellite
(unit size 1-9 bp)

Minisatellite
(unit size 10-100 bp)

Macrosatellite
(unit size > 100 bp)

Fig. 1. Schematic representation of different types of TRs. (a) Different conservation of repeat unit sequence. (b) Different sizes of repeat unit.
Space between repeat units has only been introduced to improve visual clarity of the figure.

ª 2013 Federation of European Microbiological Societies. FEMS Microbiol Rev 38 (2014) 119–141
Published by John Wiley & Sons Ltd. All rights reserved
Variable tandem repeats and bacterial adaptation 121

magnitude higher than that of other repeat types (i.e. Closer examination of the SSR distribution across the
minisatellite and macrosatellite) in the genomes of most, genome shows significant differences in coding and non-
if not all bacteria. Of course, this is not unexpected coding regions. Because bacterial genomes are more com-
because this SSR count included even mononucleotide pact than those of eukaryotes, they have comparatively
trimers (e.g. AAA), which account for about 70% of the more intragenic than intergenic SSRs. For example, in
total number of SSRs. While SSRs are generally believed Escherichia coli K-12, 79.5% of SSRs locate in coding
to contribute to genome polymorphism and adaptation regions (Gur-Arie et al., 2000), whereas in the genome
potential of bacteria (Kassai-Jager et al., 2008), the contri- of the Japanese pufferfish (Fugu rubripes), only 11.6% of
bution of these very small SSRs like mononucleotide SSRs are intragenic (Edwards et al., 1998). Generally, long
trimers is probably limited. In fact, a rough threshold of mono- and dinucleotide SSRs are excluded from coding
minimum TR unit number (4–9) has been noted, below regions, probably because they have a higher probability
which a SSR is not likely to mutate or be variable (Lai & to rearrange and cause frameshift mutations in genes (Co-
Sun, 2003; Dettman & Taylor, 2004; Kelkar et al., 2010). enye & Vandamme, 2005; Ackermann & Chao, 2006; Orsi
Intriguingly, heptameric repeats were found to be over- et al., 2010; Lin & Kussell, 2012). In contrast, SSRs whose
represented among these SSRs in most prokaryotes, and unit size is a multiple of three nucleotides (3, 6, 9 …) are
it was hypothesized that the seemingly preferred 7 bp overrepresented in open reading frames (ORFs) because
length of a repeat unit might relate to the DNA segment their expansion or contraction does not disrupt the read-
size that interacts with the active site of the DNA poly- ing frame (Mrazek et al., 2007). However, exceptions have
merase, thus facilitating the occurrence of polymerase been reported. For example, tetranucleotide SSRs of H. in-
slippage (Mrazek et al., 2007). fluenzae are exclusively found in ORFs, which is consistent
A remarkable feature of SSRs is their widely diverse dis- with their role in phase variation (Power et al., 2009). An
tribution across species, even closely related ones, and this interesting situation exists in the mycoplasmas, where long
may indicate that they are subject to rapid evolutionary trinucleotide repeats are overrepresented in Mycoplasma
change (Yang et al., 2003; Mrazek, 2006; Kassai-Jager et al., genitalium, Mycoplasma gallisepticum, and Mycoplasma
2008). Analysis of more than 300 prokaryotic genomes hyopneumoniae, but occur mainly in intergenic regions in
showed that the distribution of SSRs varied with the bacte- the former two species, but in coding regions in the latter
rial species, genome size, and G + C content (Mrazek one (Mrazek, 2006). This difference in distribution is also
et al., 2007). More specifically, SSRs with small motif reflected in different functional roles. In M. gallisepticum,
(1–4 bp) are more abundant in small genomes and particu- the most prominent trinucleotide TRs are the GAA
larly in host-adapted pathogens with reduced genomes repeats in the 5′ untranslated region of the 42 up to 70
(< 2 Mb) and low G + C content (< 40%), such as Myco- vlpA adhesin gene paralogs that exist in each strain, which
plasma and Haemophilus spp. (Moxon et al., 2006; Trean- regulate vlpA gene expression (Glew et al., 1998, 2000;
gen et al., 2009). In contrast, SSRs with a larger motif Liu et al., 2000; Papazisi et al., 2003). In contrast,
(5–11 bp) are more frequent in nonpathogens and oppor- M. hyopneumoniae trinucleotide repeats are found mostly
tunistic pathogens with large genomes (> 4 Mb) and high within hypothetical ORFs, but also in some adhesins, and
G + C content (> 60%), such as Burkholderia and Anabae- their contraction or expansion results in variability of
na spp. Based on this observation, it was hypothesized that amino acid repeats that are believed to play a role in pro-
the differential representations of SSRs in bacteria may cor- tein–protein interaction or adhesion (Mrazek, 2006).
relate with pathogenicity, but more work is needed to cor- A more detailed study on the occurrence of intragenic
roborate this. Another interesting observation is that some TRs in 44 bacteria and archaea revealed additional
relatively large bacterial genomes (e.g. Pseudomonas aeru- features (Lin & Kussell, 2012). Intragenic SSRs were
ginosa, c. 5 Mb) have fewer SSRs than would be predicted found more frequently near the termini (5′ and 3′ ends)
based on their genome properties, but harbor compara- of the ORF rather than in the middle, which most likely
tively more two-component sensor transducers. In stems from biophysical constraints of protein structure.
contrast, some host-adapted pathogens with small genome In addition, SSR-induced frameshifts at the 3′ end are less
size (i.e. Haemophilus influenzae, Neisseria meningitidis, harmful than at other parts, because most of the
and Helicobacter pylori) have comparatively more SSRs, but upstream coding region will not be affected. Nevertheless,
less two-component sensor transducers (Moxon et al., an overrepresentation of SSRs was found in the 5′ end in
2006). Thus, it seems that environmental adaptability in ORFs of pathogens, probably because this allows SSR-
host-adapted pathogens depends primarily on SSR varia- induced frameshifts to function as an ON/OFF switch for
tions, while in opportunistic pathogens with a more versa- these ORFs, which can be advantageous for pathogens
tile lifestyle it depends primarily on two-component sensor because it facilitates rapid adaptation of populations.
transducers. Similar observations had already been made earlier for

FEMS Microbiol Rev 38 (2014) 119–141 ª 2013 Federation of European Microbiological Societies.
Published by John Wiley & Sons Ltd. All rights reserved
122 K. Zhou et al.

some intragenic mononucleotide repeats at the 5′ end of replication slippage and recombination, are currently
genes (van Passel & Ochman, 2007; Janulczyk et al., 2010; widely accepted to explain TR variation (Pearson et al.,
Orsi et al., 2010). However, it remains unclear and often 2005; Bichara et al., 2006; Gemayel et al., 2010). With
difficult to prove whether this type of distribution bias of regard to the slippage mechanism, several models have
intragenic SSRs is linked to selection pressures in bacteria. been proposed, and the most common one is strand-
An argument in favor of such a link is that intragenic slippage mispairing (SSM), also called DNA slippage or
SSRs show a preference for certain categories of genes. In polymerase slippage (Kornberg et al., 1964; Streisinger
both Gram-negative (e.g. Haemophilus and Helicobacter) et al., 1966). This model proposes that TR rearrange-
and Gram-positive (e.g. Streptococcus) pathogens, SSR- ments result from strand slippage during DNA replica-
associated genes frequently encode virulence factors, cell tion. The process is initiated by the formation of a bulge
surface components, and restriction–modification structure of unpaired repeats either on the template or
enzymes (van Belkum, 1999; Moxon et al., 2006; Guo & on the nascent strand. If the bulge is present on the tem-
Mrazek, 2008; Power et al., 2009; Janulczyk et al., 2010). plate strand, it will result in a TR contraction (deletion)
On the other hand, several intragenic SSRs with numer- in the newly synthesized DNA. In contrast, TR expansion
ous repeat copies and a unit size that is not a multiple of (insertion) will result when a bulge forms on the nascent
three are also found in housekeeping genes whose prod- strand (Fig. 2). Currently, substantial evidence supports
ucts are essential for important cellular processes, such as the involvement of replication in TR instability in bacte-
cell division, energy production, and DNA replication ria. In E. coli, for example, triplet nucleotide tracts in a
and repair (Guo & Mrazek, 2008). Obviously, corre- plasmid or in the chromosome were dramatically destabi-
sponding TR rearrangements leading to reading frame lized in a dnaQ49 mutant. The dnaQ gene encodes the
disruption are anticipated to be detrimental or even lethal 3′–5′ exonucleolytic e-subunit of DNA polymerase III,
for the cell, and it remains unclear why such TRs have which is involved in proofreading, and it was therefore
been maintained during evolution. suggested that this mutant failed to correctly remove
Intergenic SSRs also show a nonrandom distribution, slipped structures in the TR tracts during replication (Iyer
being found more frequently in the immediate vicinity of et al., 2000; Zahra et al., 2007). Moreover, mutations in
genes than at distant positions. For example, intergenic the a subunit (encoded by dnaE) of DNA polymerase III
SSRs of E. coli K-12 concentrate in a region up to 200 bp holoenzyme, c and s subunits of the clamp-loading com-
from the start codon, which contains proximal regulators plex (encoded by dnaX), and b clamp (encoded by dnaN)
of gene expression (Gur-Arie et al., 2000). Another study have also been shown to increase instability of microsatel-
showed that in most cases, the intergenic SSRs with lites and tandemly repeated DNA sequences (reviewed in
numerous copies are located upstream of the first gene in Bichara et al., 2006). In general, the effect of DNA repli-
prokaryotic operons (Guo & Mrazek, 2008). Together, cation on TR rearrangements provides evidence for the
both studies reflect the potential role of intergenic SSRs replication slippage mechanism, because a prerequisite of
in the regulation of gene expression. this mechanism is that DNA replication is stalled by the
secondary loop structures formed by TRs. Notably, the
SSM mechanism is not only widely accepted to account
The variability of TRs
for the majority of SSR variations, it is also invoked to
The variability of TRs is thought to be one of the drivers explain the genesis of TRs from unrepeated DNA (Levin-
of genomic plasticity. The regions containing TRs are son & Gutman, 1987; Waite et al., 2003; Lindb€ack et al.,
potentially hypermutable by contraction (deletion) or 2011).
expansion (insertion) of TR units, and mutation frequen- Besides SSM, recombination is considered as another
cies up to 10 1 have been reported in bacteria (Rando & mechanism of TR instability that could explain phenom-
Verstrepen, 2007). Not surprisingly, the polymorphisms ena that cannot be explained by the slippage mechanism.
found in TR loci provide a foundation for DNA genotyp- Generally, recombination is more important for the rear-
ing approaches such as variable number TR-based typing rangement of TRs with large unit size, whereas SSM is
or multilocus variable repeat analysis, which are com- the dominant mechanism underlying variation of TRs
monly applied for pathogen typing (Lindstedt, 2005, with small unit size (Bi & Liu, 1996; Richard & P^aques,
2011; reviewed in Chiou, 2010). 2000; Bzymek & Lovett, 2001; Rocha, 2003; Gemayel
et al., 2010). Both homologous (RecA-dependent) and
illegitimate (RecA-independent) recombination can be
The molecular mechanisms of TR variation
involved. Several models have been proposed for the
Based on extensive studies in both plasmid-based and recombination mechanism, such as unequal crossover and
chromosomal systems, two nonexclusive mechanisms, intramolecular recombination (Fig. 3), and evidence for

ª 2013 Federation of European Microbiological Societies. FEMS Microbiol Rev 38 (2014) 119–141
Published by John Wiley & Sons Ltd. All rights reserved
Variable tandem repeats and bacterial adaptation 123

5’ 3’ ReplicaƟon
3’ 5’

3’
Strand
5’ dissociaƟon
3’ 5’

Expansion ContracƟon

5’ 3’ 5’ 3’
3’ 5’ 3’ 5’

Fig. 2. Diagram illustrating the replication slippage mechanism of TR rearrangement. Repeat units are shown as blocks on nascent (light green)
and template (dark green) DNA strands. Shown is a partially replicated TR region undergoing transient dissociation and mispairing, resulting in a
bulge on the template or the nascent strand and leading to insertion or deletion of TRs, respectively. Space between repeat units has only been
introduced to improve visual clarity of the figure.

the involvement of recombination in TR variations is can vary widely depending on both intrinsic (structural)
accumulating. For example, it has been suggested that and extrinsic (environmental) factors. Regarding intrinsic
double-strand DNA break (DSB) repair can induce TR TR features, a positive correlation has been established
instability via homologous recombination in prokaryotes between TR copy number and rearrangement frequency
and eukaryotes (Hebert & Wells, 2005; reviewed in Rich- in several studies. Through in silico genome analysis of
ard et al., 2008; Malkova & Haber, 2012), and mechanis- multiple strains of 42 fully sequenced prokaryotic species,
tic models explaining this process have been elaborated. Lin & Kussell (2012) observed that the variability of three
One such model is the DSB repair slippage model, which types of SSRs (monomeric, dimeric, and trimeric repeats)
combines the double Holliday junction intermediate increased dramatically with the number of repeat units. In
pathway with the strand-slippage model (Richard et al., another study, an exponential relation between the num-
2008). In an alternative explanation, the synthesis-depen- ber of repeat units and rearrangement frequency was
dent strand annealing pathway also contributes to the TR observed in a comparison of 30 artificially constructed
rearrangements mediated by DSB repair (P^aques et al., TRs (unit lengths of 2, 10, and 20 nucleotides; number of
1998, 2001; Richard et al., 1999; Richard & P^aques, units between 2 and 50; sequence conservation between
2000). This model is proposed for explaining TR rear- 62.5% and 100%; Legendre et al., 2007). This also
rangements that are rarely associated with crossover accounts for the fact that long TRs are relatively uncom-
events (P^aques et al., 1998, 2001; Richard et al., 1999). mon (Lai & Sun, 2003). Similar findings have been
reported in several other studies (Goldstein & Clark,
1995; Brinkmann et al., 1998; Lai & Sun, 2003; Vogler
Factors influencing TR rearrangement
et al., 2006). Likewise, a positive relationship is generally
frequencies
found between TR mutation frequency and the size of the
While TRs generally are intrinsically prone to incur con- repeat unit (Sia et al., 1997; Schug et al., 1998; Eckert &
traction or expansion, the actual frequency of these events Hile, 2009; Bayliss et al., 2012) as well as the degree of

FEMS Microbiol Rev 38 (2014) 119–141 ª 2013 Federation of European Microbiological Societies.
Published by John Wiley & Sons Ltd. All rights reserved
124 K. Zhou et al.

5’ 3’ (a) (b)
3’ 5’

5’ 3’ 3’
3’ 5’
5’
5’

Unequal 3’
crossover
Intramolecular
5’ 3’ recombinaƟon
3’ 5’
Expansion
5’ 3’
3’ 3’ 3’ 5’
5’ 5’ ContracƟon
ContracƟon

Fig. 3. Diagram illustrating the recombination mechanism of TR rearrangement. DNA molecules with repeat units represented as blocks are
shown in different colors. (a) Model of unequal crossover. When repeat units of two different DNA molecules misalign, unequal crossover can
occur, resulting in repeat expansion in one crossover product and repeat contraction in the other. (b) Model of intramolecular recombination.
When recombination occurs between repeat units within a DNA molecule, two products are generated that have undergone a repeat
contraction.

conservation between repeats (Legendre et al., 2007). In variation at increased growth temperature and upon starva-
addition, the GC content of the TR is also an important tion in E. coli O157:H7, but not upon irradiation (Cooley
factor determining stability, because repeated sequences et al., 2010). Likewise, in sclB of Streptococcus pyogenes,
are prone to form diverse non-B DNA structures (i.e. encoding a collagen-like surface protein, TR variations
hairpins and triplexes), which may cause pausing of the occurred during growth in fresh human blood, but not in
DNA polymerase and replication fork collapse, and in medium (Rasmussen & Bj€ ork, 2001). However, the
turn necessitate intervention of the repair and recombina- underlying mechanisms by which environmental stresses
tion machinery to reinitiate replication (Wells et al., 2005; affect the TR mutation frequency are poorly understood.
Choudhary & Trivedi, 2010). Besides, the orientation of
the repeats with respect to the direction of replication can
The phenotypical impact of intergenic
affect the mutation ratio as well, because TRs are more
TR variations in bacteria
prone to form secondary structures in one orientation
than in the other (Hebert et al., 2004), and replication Accumulating evidence indicates that rearrangements of in-
fidelity is not equal in the leading strand compared with tergenic TRs can confer transcriptional evolvability (Jansen
the lagging strand (Gawel et al., 2002). Intriguingly, based et al., 2012). More specifically, SSRs positioned as cis-regu-
on a theoretical model, Lai & Sun (2003) suggested that latory elements around the promoter region can induce
expansion occurs more frequently for short microsatel- phase variation (i.e. stochastic, high frequency, reversible
lites, while contraction occurs more frequently for long switching of genotype, and/or phenotype) by modulating
ones, suggesting the rearrangement pattern might be the transcription of the corresponding genes. Most studies
dependent on the repeat type. However, the lack of exper- indicate that intergenic SSRs, except monomeric SSRs,
imental evidence so far cannot validate this observation. involved in phase variation tend to be A/T rich, which
Aside from intrinsic factors, extrinsic environmental makes them prone to melting and SSM. In this section, we
conditions may also affect TR rearrangement frequencies. review examples of this mechanism of phase variation
For instance, it was shown that several TRs used in a according to the different positions of intergenic TRs rela-
multilocus TR typing scheme of E. coli showed enhanced tive to the transcriptional start site (Fig. 4; Table 1).

ª 2013 Federation of European Microbiological Societies. FEMS Microbiol Rev 38 (2014) 119–141
Published by John Wiley & Sons Ltd. All rights reserved
Variable tandem repeats and bacterial adaptation 125

ATG regulator protein to the nadA promoter (Martin et al.,


UAS RNA pol
SD 2003, 2005). More recently, Metruccio et al. (2009) dis-
ORF
–35 –10 +1 covered that depending on the number of TAAA repeats,
A B C a novel repressor (NadR) prevents transcription of nadA
through binding of two operators flanking the variable
Fig. 4. Scheme showing possible positions of intergenic TRs in a tetrameric repeat tract on both sides. As a result, it was
standard promotor region that can cause phase variation. Indicated proposed that alteration of the spacing between these two
are an ORF, a promoter with 10 and 35 signatures that are
operators by variation of the number of TAAA repeats
recognized by RNA polymerase (RNA pol), an upstream regulatory
sequence to which activators or repressors can bind (UAS), the
may affect the ability of NadR to repress nadA expression
transcription initiation site (+1), a Shine–Dalgarno sequence for (Metruccio et al., 2009).
ribosome binding (SD), and a translation start codon (ATG). Repeats
in regions A and B (double-headed arrows) can modulate gene
expression by affecting transcription initiation (see text TRs upstream
TRs between 35 and 10 sites of promotor
of 35 site of promotor and TRs between 35 and 10 sites of The spacing between the 35 and 10 sites of a standard
promotor), whereas repeats in region C operate via as yet
promoter is critical for the binding efficiency of RNA
unidentified means (see text TRs between transcriptional start site and
ORF; adapted from van der Woude & Baumler, 2004).
polymerase (Fig. 4), and the optimal distance is around
17 bp in bacteria. Consequently, when TRs locate in that
region, copy number changes are expected to modulate
TRs upstream of 35 site of promotor the transcription level of the genes in the transcriptional
unit. An example of this is found in the host-adapted
When TRs are present upstream of the 35 site, the pathogen H. influenzae, which adheres to human epithe-
repeat copy number is expected to affect the binding of lial cells with the help of LKP fimbriae (also called long,
transcription factors and thus modulate gene expression. thick pili). These fimbriae are encoded by the hif gene
This has been reported in the pathogen N. meningitidis, cluster and important for H. influenzae infection at differ-
where expression of the nadA gene (encoding a protein ent stages (van Ham et al., 1993). Phase variation in the
that functions as invasin and adhesin) is regulated by the expression of these fimbriae is mediated by a string of
number of tetrameric repeats (TAAA) within the dinucleotide repeats between the 35 and 10 sites
upstream region of the RNA polymerase binding site, within the overlapping, but divergent promoter regions of
resulting in three significantly distinct transcriptional lev- hifA and hifB genes, with TR copy numbers of 9, 10, or
els (high, intermediate, and low). The frequency of phase 11, respectively, resulting in no, high, and low expression
variation between these levels has been estimated at of both genes.
c. 4.4 9 10 4. Remarkably, the levels of nadA transcrip- Another intriguing example is the FetA protein of Nei-
tion show a periodic rather than a monotonous relation sseria gonorrhoeae, an iron-repressible protein functioning
to the number of repeat units. As such, the transcription as ferric enterobactin receptor. The expression of FetA
level varies in the order low–high–intermediate–low–high exhibits extremely rapid phase variation (switching
for repeat copy numbers of 4–5–6–7–8 and again for frequency up to 1.3% per generation) correlating with
9–10–11–12–13 repeats. Further mechanistic studies indi- polymorphism of a poly-C tract between the 10 and
cated that variation of the tetrameric repeats affects the 35 regions of the fetA promoter. The various lengths of
binding of the integration host factor transcriptional the poly-C tract result in either high or low expression.

Table 1. Overview of mechanisms by which intergenic TRs can modulate gene expression

Location of TRs Mechanisms References


Upstream of 35 site Affects transcription initiation by modifying binding Miller et al. (1987), Martin et al. (2003, 2005),
affinity of regulatory proteins Metruccio et al. (2009)
Between 35 and 10 sites Affects transcription initiation by altering the Willems et al. (1990), Yogev et al. (1991), van
distance of promoter elements Ham et al. (1993), Sarkari et al. (1994),
Carson et al. (2000), van der Ende et al. (2000)
Between transcriptional start and ORF May modify binding affinity of regulatory Lafontaine et al. (2001), Attia & Hansen (2006)
proteins or mRNA stability
Between two separate transcription Unknown Dawid et al. (1999)
start sites
ORF, open reading frame; TR, tandem repeat.

FEMS Microbiol Rev 38 (2014) 119–141 ª 2013 Federation of European Microbiological Societies.
Published by John Wiley & Sons Ltd. All rights reserved
126 K. Zhou et al.

It was suggested that phase variation of FetA reflects a were increased as the repeat copy number increased, pos-
balance between the advantages of iron scavenging, on sibly because the variable number of AGAT repeats may
the one hand, and evasion of the host immune response affect the secondary structure of the UspA2 mRNA
(based on FetA immunogenicity), on the other (Carson transcript.
et al., 2000).
A more complex situation is found in the PorA outer
TRs between two separated transcription start
membrane protein in N. meningitidis. Not only is expres-
sites
sion of the porA gene stochastically modulated by two
variable homopolymeric tracts (poly-G and poly-T) This special example of phase switching induced by
between the 35 and 10 sites of the promoter region, variable intergenic TRs has so far only been described in
there is also a variable poly-A tract within the porA H. influenzae (Dawid et al., 1999). HMW1 and HMW2
coding region. Both sites are believed to serve evasion of are two adhesins of H. influenzae that exhibit different
the host immune response to this protein and explain the cellular binding specificities and are encoded by two sepa-
poor efficacy observed for PorA-based vaccines (van der rate chromosomal loci hmw1AC and hmw2AC, respec-
Ende et al., 2000). tively. Both genes show 80% similarity, suggesting that
Further examples include the genes encoding the lipo- they can be considered as alleles (Barenkamp & Leininger,
protein Vlps of Mycoplasma hyorhinis (Yogev et al., 1992; Ecevit et al., 2004). Promoter analysis revealed two
1991), the fimbrial subunits of Bordetella pertussis (Wil- transcription start sites (P1 and P2) within the upstream
lems et al., 1990), and the outer membrane protein Opc region of each gene and a heptameric (ATCTTTC) TR
of N. meningitidis (Sarkari et al., 1994), and it can be array between P1 and P2 of both genes. The occurrence of
concluded that variable SSRs within the 35 and 10 variations in the number of TR copies has been confirmed
region represent a widespread strategy of phase variation in H. influenzae isolates from patients with chronic
by modulated gene expression in pathogens. obstructive pulmonary disease (Dawid et al., 1999; Cholon
et al., 2008). These variations can affect mRNA synthesis
and thereby influence the expression of corresponding pro-
TRs between transcriptional start site and ORF
teins. For example, an increasing number of TR units
While intergenic TR-dependent phase variations in bacte- resulted in a gradual decrease in specific mRNA synthesis
ria mostly belong to the two classes described above, and protein expression and vice versa. However, the
some cases are mediated by TRs located between the tran- underlying molecular mechanism remains obscure.
scriptional start site and the ORF. In the Gram-negative
pathogen Moraxella catarrhalis, the UspA1 protein func-
The phenotypical impact of intragenic
tions as an adhesin to mediate binding to human epithe-
TRs in bacteria
lial cells (Lafontaine et al., 2000). Its expression was
shown to be phase variable and correlated with adherence There is abundant evidence that besides intergenic TRs,
capacity. Sequence analysis revealed a variable homopoly- also intragenic TRs can trigger phase variation (Table 2).
meric poly-G tract between the transcriptional start site However, the underlying mechanisms are dependent on
and the start codon to be responsible for the variability the nature of the TR. More specifically, if the TR unit size
in uspA1 expression. Stable mRNA and strong expression is not a multiple of three, rearrangements are able to
of UspA1 was detected with a 10-bp G repeat tract, while induce frameshift mutations as the cause of ON–OFF
truncated mRNA and weak expression occurred with a 9- phase variation. In comparison, phase variation induced
bp G tract. Based on these observations, it was proposed by TRs whose unit size is a multiple of three is more
that alterations in the poly-G tract affect the binding effi- complex and is probably related to specific structural and
ciency of transcriptional regulators and/or the stability of functional alterations of the corresponding proteins
the uspA1 mRNA (Lafontaine et al., 2001). (Gemayel et al., 2010). In this section, we review studies
Interestingly, a similar case was recently uncovered in on the phenotypical impact of intragenic TRs, grouped
another outer membrane protein of M. catarrhalis, according to their location in different functional classes
UspA2, which is involved in serum resistance and vitro- of genes.
nectin binding (Attia & Hansen, 2006). It was observed
that a tetrameric (AGAT) repeat tract, located between
Cell surface structural genes with TRs
the uspA2 transcription start site and the start codon, is
highly variable in M. catarrhalis isolates. Moreover, the It has been noted that TRs are most abundant in genes
expression level of UspA2 and, as a result, the serum whose products are either exposed on the cell surface or
resistance and vitronectin binding capacity of the cells involved in the biogenesis of cellular surface structures,

ª 2013 Federation of European Microbiological Societies. FEMS Microbiol Rev 38 (2014) 119–141
Published by John Wiley & Sons Ltd. All rights reserved
Variable tandem repeats and bacterial adaptation 127

Table 2. Examples of intragenic TRs causing phase switching in bacteria

Affected moiety Bacterial species Gene(s) or operon Repeat motif* (5′–3′) References
Adhesin Haemophilus influenzae cha 56 bp Sheets & St Geme (2011)
Helicobacter pylori sabA CT Goodwin et al. (2008)
Neisseria gonorrhoeae opa CTCTT Murphy et al. (1989)
Mycoplasma hominis vaa A Zhang & Wise (1997)
Legionella pneumophila lcl 45 bp† Vandersmissen et al. (2010)
Capsule Streptococcus pneumoniae cps15bM TA van Selm et al. (2003)
cap8E 223 bp Waite et al. (2003)
tts 22 bp Waite et al. (2003)
Neisseria meningitidis siaD C Hammerschmidt et al. (1996b)
Escherichia coli neuO AAGACTC Deszo et al. (2005)
Effector Xanthomonas campestris avrBs3 102 bp Herbers et al. (1992), Kay et al. (2007)
Fe binding Haemophilus influenzae hgp CCAA Jin et al. (1999), Ren et al. (1999)
Neisseria meningitidis hpuA, hmbR G Lewis et al. (1999), Richardson &
Stojiljkovic (1999)
Flagellin Campylobacter coli flhA T Park et al. (2000)
Lipoprotein Mycoplasma hyorhinis vlp 24/26 bp† Rosengarten & Wise (1991)
Mycoplasma bovis vspA 18/24 bp† Lysnyansky et al. (1996)
Mycoplasma pulmonis vsa 34 bp Bhugra et al. (1995)
Lipopolysaccharides Haemophilus influenzae losA CGAGCATA Erwin et al. (2006)
Haemophilus influenzae lic1, lic2 A, lic3 A CAAT Hosking et al. (1999), Dixon et al. (2007)
Haemophilus influenzae lex2, oafA GCAA Griffin et al. (2003), Fox et al. (2005)
Neisseria meningitidis lgtA, C, D G Yang & Gotschlich (1996), Jennings
et al. (1999)
Helicobacter pylori futA, futB C & 21 bp‡ Appelmelk et al. (1999), Nilsson
et al. (2008)
Metabolism Escherichia coli xylB G Funchain et al. (2000)
Mismatch repair Escherichia coli mutL CTGGCG Shaver & Sniegowski (2003)
Salmonella Typhimurium mutL GCTGGC Chen et al. (2010)
Outer membrane protein Neisseria meningitidis porA G van der Ende et al. (2000)
Group B streptococci bca 246 bp Gravekamp et al. (1996, 1997)
Mycoplasma fermentans p78 A Theiss & Wise (1997)
Inner membrane protein Escherichia coli tolA 15-18 bp† Zhou et al. (2012a, b)
Peroxiredoxin Escherichia coli ahpC TCT Ritz et al. (2001)
Pilus Neisseria gonorrhoeae pilC G Jonsson et al. (1991, 1992)
Legionella pneumophila fimV 18 bp† Coil & Anne (2010)
Regulator Listeria monocytogenes ctsR GGT Karatzas et al. (2003, 2005)
prfA CAGGAGT Lindb€ ack et al. (2011)
R-M§ system I Haemophilus influenzae hsdM GACGA Zaleski et al. (2005)
Neisseria gonorrhoeae hsdS G Adamczyk-Poplawska et al. (2011)
R-M§ system III Haemophilus influenzae modA AGTC Srikhanta et al. (2005)
Neisseria spp. modA AGCC Srikhanta et al. (2009)
Neisseria spp. modB CCCAA Srikhanta et al. (2009)
Neisseria meningitidis modD ACCGA Seib et al. (2011)
Helicobacter pylori res C de Vries et al. (2002)
Helicobacter pylori modH G Srikhanta et al. (2011)
Pasteurella haemolytica mod CACAG Ryan & Lo (1999)
R-M§ system IV Escherichia coli mrr G Tesfazgi Mebrhatu et al. (2011)
TR, tandem repeat.
*The motif sequences of microsatellites (≤ 9 bp) are listed; for longer TRs, only the length is given.

Different repeat unit lengths or sequences have been reported for this TR locus.

Two TR loci exist at different positions in the same gene.
§
Restriction–modification system.

such as lipopolysaccharides (LPS), adhesins, pili, fimbriae, et al., 2010; Jerome et al., 2011). Extensive studies in dif-
and capsules (Moxon et al., 1994; Jordan et al., 2003; ferent organisms and with different cell surface genes
Verstrepen et al., 2004; Gibbons & Rokas, 2009; Janulczyk support the notion that stochastic TR-based switching

FEMS Microbiol Rev 38 (2014) 119–141 ª 2013 Federation of European Microbiological Societies.
Published by John Wiley & Sons Ltd. All rights reserved
128 K. Zhou et al.

contributes to the rapid generation of diversity in surface Interestingly, searching for genes harboring tetrameric
structures, which in pathogens can serve as a mechanism TRs has proven to be a successful strategy to identify
for escaping the immune response and/or for determining novel LPS biosynthesis genes in H. influenzae genomes
tissue tropism. Some examples are reviewed more elabo- (Hood et al., 1996; Fox et al., 2005). Similar mechanisms
rately below. generating LPS diversity have also been described in
N. meningitidis, where the ON/OFF variation in expres-
sion of genes of the lgt gene family (encoding LPS bio-
TRs within LPS biosynthesis genes
synthesis functions) is mediated by stochastic expansions
LPS is a complex macromolecule in Gram-negative bacte- or contractions in their intragenic homopolymeric tracts
ria that is composed of three distinct parts (i.e. lipid A, and where the corresponding LPS structures have so far
core sugar, and O-antigen side chains), and a large num- been classified into 12 immunotypes (Jennings et al.,
ber of genes are involved in the synthesis and export of 1999; Berrington et al., 2002; Bayliss et al., 2008).
LPS. As one of the major cell surface antigens, LPS is The O-chain of H. pylori LPS can be fucosylated, thereby
often implicated in cell adhesion and virulence. Further- generating Lewis antigens that mimic human blood group
more, because LPS molecules are an essential structural antigens and mediate immune evasion (reviewed in Kusters
component of the outer membrane that forms the outer et al., 2006). Lewis antigens of H. pylori are subject to
shell of the Gram-negative cell, it also determines bacte- reversible, high-frequency phase variation, and one of the
rial resistance to a variety of toxic chemicals, including mechanisms is the slipped-strand mispairing of poly-C
some antibiotics and xenobiotics. A notable feature of tracts within three fucosyltransferase genes (futA, futB, and
LPS of some pathogens is the extensive intra- and inter- futC; Appelmelk et al., 1999; Wang et al., 1999). In addi-
strain heterogeneity of the glycoform structure (i.e. the tion, a unique TR region with a 21-bp unit size was recently
moieties comprising the core and O side chain sugars), uncovered in the 3′ end of futA and futB, but not in futC.
which is mainly due to incomplete biosynthesis during Strikingly, although copy number variations of the 21-bp
the stepwise assembly of the sugar residues resulting from TR tract in both futA and futB did not alter the reading
the phase-variable expression of the corresponding frame, they could affect the fucosyltransferase activity. In
biosynthesis genes (Schweda et al., 2007). Typically, the fact, a correlation was observed between the copy number
phase variability of these genes derives from the fact that of the 21-bp TRs and the number of O-antigen units being
they contain nontrimeric TR tracts that exhibit stochastic fucosylated, and the addition of one repeat unit led to the
variation. In some pathogens, this type of stochastic vari- addition of an N-acetyl-b-lactosamine (LacNAc) unit in
ation occurs in more than one gene for LPS synthesis, the O-antigen polysaccharide (Nilsson et al., 2006, 2008).
turning phase switching into a combinatorial process. These studies show that the variability of TRs in futA, futB,
Unlike that of most Gram-negative bacteria, the core and futC increases the antigen diversity and population
LPS of H. influenzae lacks the homopolymeric sugar units heterogeneity and thereby supports adaptability of
comprising the O-antigen side chains. Therefore, phase H. pylori to fluctuating conditions in the gastric mucosa.
variation in LPS biosynthesis of H. influenzae occurs
mainly through reversible expression switching of a subset
TRs within capsule biosynthesis genes
of TR-containing genes, involved in the addition of core
sugars (i.e. glucose and sialic acid) to the conserved As one of the most external structures on the bacterial
tri-heptose backbone and in the addition of phospho- surface, capsules may completely conceal other antigenic
rylcholine or acyl groups to these core sugars (Moxon surface molecules or may be co-exposed with other anti-
et al., 2006; Fig. 5a). These genes include lic1A, lic1B, gens, which are thought to be important for pathogenicity
lic1C, and lic1D (collectively designated as lic1 locus), and of bacteria. Production of a capsule provides some patho-
lic2A, lic3A, lgtC, lex2, and oafA, each of which contains a genic bacteria with resistance to phagocytic and comple-
variable tetrameric repeat tract (reviewed in Schweda ment-mediated killing and, at the same time, affects
et al., 2007). Rearrangements in the tetrameric repeat bacterial attachment to host cells. Consequently, the ability
tracts of these genes can individually turn their expression to regulate capsule expression might confer a selective
on or off, resulting in a modified LPS molecule at the advantage for pathogens to cope with host immune
single cell level, and a repertoire of different LPS epitopes responses during different stages of the infection process.
throughout the population (Fig. 5b; Moxon et al., 1994; For example, it was found that acapsulate variants of
Bayliss et al., 2001; Moxon et al., 2006). Remarkably, all N. meningitidis serogroup B show much higher adherence
the phase-variable genes involved in LPS biosynthesis of and invasion of epithelial cells than their capsulated pro-
H. influenzae account for structural elements directly rele- genitors. Such variants can be generated at high frequency
vant to virulence as well (Schweda et al., 2007). due to SSM of a poly-C tract in the polysialyltransferase

ª 2013 Federation of European Microbiological Societies. FEMS Microbiol Rev 38 (2014) 119–141
Published by John Wiley & Sons Ltd. All rights reserved
Variable tandem repeats and bacterial adaptation 129

(a) PEtn

lic1A 4
PC 6Glc Hep Kdo Lipid A
β-1,4 α-1,5

α-1,3

Hep6 PEtn

α-1,2
lic3A lic2A
NeuAc Gal Glc Hep
α-2,3 β-1,4 β-1,2

(b)
NeuAc
lic3A (ON) (OFF) (ON) (ON)
Gal
lic2A (ON) (OFF) (ON) (OFF)
PC
lic1 (ON) (ON) (OFF) (OFF)

Type I Type II Type III Type IV

Fig. 5. Structural variations of LPS molecules induced by TR rearrangements in LPS biosynthesis genes. (a) Representation of one possible
structure of the LPS from Haemophilus influenzae strain Rd. The conserved tri-heptose backbone is highlighted in bold. Three phase-variable lic
genes involved in the addition of specific components to the backbone are shown. Kdo, 2-keto-3-deoxyoctulosonic acid; Hep, L-glycero-D-manno-
heptose; Glc, D-glucose; Gal, D-galactose; NeuAc, N-acetylneuraminic acid; PEtn, phosphoethanolamine; PC, phosphorylcholine. (b) Scheme
showing some of the possible LPS types generated on the cell surface of H. influenzae Rd by the combinatorial effect of three LPS synthesis
genes with phase-variable expression. lic2A is responsible for adding a galactose (Gal); lic3A, for adding sialic acid (NeuAc); and lic1, for adding
phosphorylcholine (PC). The repeat tracts are shown by hatched boxes in the gray arrow representation of each gene. The TRs in each of these
genes are subject to variation, which can cause a frameshift and thus block expression (indicated by ON or OFF). Types I–IV represent microbial
cells with different LPS antigens depending on lic1, lic3A, and lic2A gene expression (adapted from Moxon et al., 2006).

gene siaD (Hammerschmidt et al., 1996a, b; Spinosa et al., E. coli K1, which can be modified through phase-variable
2007). On the other hand, meningococcus isolates from expression of the sialyl O-acetylating activity, resulting in
the blood of meningitis patients are almost always capsu- an altered immunogenicity and susceptibility to glycosid-
lated, while both capsulated and noncapsulated strains ases. The phase-variable acetylation is driven by a hep-
typically coexist in the nose or throat of healthy carriers. tanucleotide TR tract (AAGACTC; copy number typically
Because phase variation of siaD is reversible, it was there- 14–39) within the O-acetyltransferase gene neuO (Deszo
fore proposed that infection is initiated by acapsulate et al., 2005). Loss or gain of a number of repeat units
strains, but that capsule biosynthesis is reactivated at a that is not a multiple of three results in a disruption of
later stage during infection, allowing N. meningitidis to the neuO reading frame and subsequent NeuO expression
resist the host immune system and to cause disease (i.e. (Deszo et al., 2005). Functional analysis furthermore
sepsis and meningitis). However, other findings indicate revealed enhanced desiccation resistance, but reduced bio-
that the presence of a capsule does not necessarily preclude film formation in E. coli K1 with active NeuO, suggesting
invasiveness, and the role of the capsule and capsule phase not only a role in host interaction, but also a more subtle
switching in different stages of meningococcal infection ecological impact of phase-variable neuO expression
may therefore be more subtle and strain dependent (Mordhorst et al., 2009). Interestingly, each set of three
(Spinosa et al., 2007; Bartley et al., 2013). repeat units encodes a protein structure designated the
Besides binary ON/OFF switching, some bacteria can poly(w) motif, and NeuO enzymatic activity was found
also modulate the composition of their capsule by a to increase with the number of poly(w) motifs, support-
mechanism of TR-dependent phase switching. This is ing maintenance of high repeat copy numbers in the
exemplified by the polysialic acid capsule (K1 antigen) of population (Bergfeld et al., 2007).

FEMS Microbiol Rev 38 (2014) 119–141 ª 2013 Federation of European Microbiological Societies.
Published by John Wiley & Sons Ltd. All rights reserved
130 K. Zhou et al.

Another intriguing aspect of this capsular acetylation is argued that the occurrence of different arrays of Opa
that the neuO gene resides in a lambdoid prophage variants in clinical (disease) and carriage (nondisease) iso-
termed ‘CUS-3’, and mitomycin treatment of E. coli K1 lates of N. meningitidis may be the result of immune
can induce release of this phage, which specifically infects selection pressure (Callaghan et al., 2006, 2008; Sadaran-
K1 antigen-expressing bacteria (Deszo et al., 2005; King gani et al., 2011).
et al., 2007). This suggests that variant neuO alleles can In the same vein, adhesins of H. pylori are categorized
be redistributed among K1-encapsulated bacteria via hori- into two main subgroups, Hop (Helicobacter outer mem-
zontal transfer. Indeed, CUS-3/neuO has been found in brane porins) and Hor (Hop-related). For some, but not
several serotypes, such as O18 and O45 (King et al., all of these adhesins, the corresponding host cell receptors
2007). On the other hand, although the receptor for have been identified (reviewed in Backert et al., 2011).
CUS-3 is polysialic acid, superinfection was not prevented For example, BabA binds to the Lewis B (Leb) antigen
by NeuO-mediated acetylation (Vimr & Steenbergen, expressed on the gastric mucosa (Ilver et al., 1998), and
2006; King et al., 2007). Notably, sialic acid is also known SabA binds to glycosphingolipids of host cells that display
as a modification of LPS in numerous pathogens, under- a sialyl-dimeric Lewis X (sialyl-Lex) antigen (Mahdavi
scoring the vital role of O-acetylation in causing structure et al., 2002). Interestingly, expression of some of the
variation of polysaccharide epitopes (also see TRs within adhesins (i.e. BabB, HopZ, SabA, OipA) is subject to ON/
LPS biosynthesis genes). OFF phase variation due to the presence of variable dinu-
Also the mini- and macrosatellites within capsule cleotide (CT) tracts within the corresponding genes. As a
biosynthesis genes of Gram-positive pathogens can medi- consequence, the expression patterns of adhesins gener-
ate capsule diversity. As such, noncapsulated serotype 3 ated by stochastic phase variation can not only affect the
Streptococcus pneumoniae strains were shown to carry an adhesion efficiency, but also alter the tissue tropism
out-of-frame perfect tandem duplication in one of the (Yamaoka et al., 2002; Backert et al., 2011). Additionally,
capsule biosynthesis genes (i.e. cap3A). Interestingly, the resulting repertoire of phase-variable adhesins can be
based on the sequence and length of the duplication, at advantageous for H. pylori to escape the host immune
least seven different cap3A alleles were found in different system.
nonencapsulated strains. Analysis of the phase reversion A final example in this category is the Eap protein of
frequency (OFF to ON) induced by TR contractions the Gram-positive pathogen Staphylococcus aureus. Eap is
revealed a positive correlation between the frequency of a multifunctional adhesin with a variable TR tract, in
reversion and the length of the duplication (Waite et al., which the size of each repeat unit is not identical (93–
2001). A similar mechanism of capsular phase variation 110 aa). A minimum of two TRs in the eap gene are
correlating with a 223-bp and a 22-bp perfect tandem required for Eap to cause agglutination, adherence and
duplication in cap8E and tts was also demonstrated in cellular invasion by S. aureus. Furthermore, as the repeat
serotype 8 and serotype 37, respectively, of S. pneumoniae copy number increases from 2 to 5, those capacities are
(Waite et al., 2003). significantly enhanced, suggesting that TR copy number
expansion in eap supports host adaptation of S. aureus
(Hussain et al., 2008).
TRs within adhesin-associated genes
Adhesins mediate bacterial attachment to and further
TRs within iron (heme) acquisition genes
colonization of host tissues, but they also act as surface
antigens (Bayliss et al., 2001). The opacity proteins Opa Free iron is typically limited in the host and is usually
of Neisseria spp. constitute a family of closely related, but sequestered by iron-binding proteins (e.g. hemoglobin,
size-variable outer membrane proteins, which enhance transferrin, and lactoferrin). Iron acquisition mechanisms
the adherence to epithelial, leukocyte, and phagocytic cells are therefore considered indispensable for bacterial patho-
(reviewed in Sadarangani et al., 2011). They do not only genicity, and pathogens have evolved many different
determine host and tissue specificity, but also facilitate strategies for adaptation to fluctuating iron concentra-
efficient cellular invasion (Carbonnelle et al., 2009). tions. As such, pathogens are able to extract the iron
Neisseria spp. strains have 3–11 opa genes whose phase- from heme groups of iron-binding proteins via surface
variable expression is modulated by a pentanucleotide TR receptors. In H. influenzae, a family of hemoglobin-bind-
tract (CTCTT) in their coding regions or by intergenic ing and hemoglobin–haptoglobin-binding proteins (Hgp)
recombination. As a result, a vast array of Opa variants is known to mediate heme scavenging (Ren et al., 1998;
can be generated to confer differential molecular specifici- Jin et al., 1999; Morton et al., 1999). Individual strains of
ties, allowing Neisseria both to alter its tissue tropism and H. influenzae have 1–4 hgp genes, of which knockout
to escape the host immune system. In fact, it has been analysis has confirmed that they are indispensable

ª 2013 Federation of European Microbiological Societies. FEMS Microbiol Rev 38 (2014) 119–141
Published by John Wiley & Sons Ltd. All rights reserved
Variable tandem repeats and bacterial adaptation 131

virulence determinants in invasive disease (Seale et al., product of the hsdSNgoAV1 is responsible for the specific
2006). Interestingly, a CCAA tetranucleotide repeat tract recognition of the target site, whereas hsdSNgoAV2 is
exists in each hgp gene, which is reminiscent of the LPS nonfunctional. It was postulated that hsdSNgoAV1 and
biogenesis genes of H. influenzae harboring a CAAT hsdSNgoAV2 are actually truncated proteins derived from
repeat tract (Moxon et al., 2006; see TRs within LPS an integral hsdS locus that has become interrupted by a
biosynthesis genes). Phase variation induced by repeat frameshift mutation resulting in the formation of a stop
rearrangements within the hgp genes has been observed; codon between hsdSNgoAV1 and hsdSNgoAV2 (Piekarowicz
however, its biological significance is still obscure. et al., 2001). Interestingly, a recent study uncovered a
A similar case exists in N. meningitidis where iron variable poly-G tract within the 3′ end of hsdSNgoAV1 in
acquisition is modulated by the variable poly-G tract in N. gonorrhoeae, and loss of a guanine in that tract
the hpuA and hmbR genes that are involved in the bio- restores the fusion of the hsdSNgoAV1 and hsdSNgoAV2
synthesis of two hemoglobin receptors (Lewis et al., 1999; genes, resulting in the generation of a new HsdSNgoAVD
Richardson & Stojiljkovic, 1999). More recently, it was protein, responsible for a novel NgoAV R-M system,
revealed that 91% of N. meningitidis pathogenic isolates, termed ‘NgoAVD’. The NgoAVD system has a modified
but only 71% of commensal isolates have at least one DNA recognition specificity, thereby conferring an altered
receptor in an ON state, suggesting expression of hemo- susceptibility to various phages (Adamczyk-Poplawska
globin receptor(s) to be important for the systemic spread et al., 2011).
of meningococci (Tauseef et al., 2011). Type III R-M systems only consist of two subunits, the
methyltransferase (encoded by a mod gene) and
restriction endonuclease (encoded by a res gene). In
TRs within genes involved in restriction–
host-adapted pathogens, ON/OFF phase variation of this
modification systems
system has been reported due to variable TRs (with
In addition to their role as drivers of genetic variation as repeat unit not being a multiple of three bases) within
reviewed above, TRs can also drive epigenetic variation either the mod or the res gene (Ryan & Lo, 1999; De Bolle
when they affect restriction–modification (R-M) systems. et al., 2000; de Vries et al., 2002; Srikhanta et al., 2005,
Currently, TR-dependent phase variations have been doc- 2009, 2011; also see review Srikhanta et al., 2010). Inter-
umented only in type I and III R-M systems. Type I R-M estingly, TR-induced phase variation in the mod gene has
systems are generally comprised of three subunits (S, M, been shown to affect the expression of a number of genes,
and R), which together form a holoenzyme that has both referred to as a phase-variable regulon or phasevarion
methylation and restriction activity. In H. influenzae, the (Srikhanta et al., 2005, 2010). In H. influenzae strain Rd,
type I R-M system HindI functions as the main defense when mod expression was switched off by a TR rearrange-
system against the entry of foreign DNA (Glover & Pie- ment in a tetrameric repeat tract, nine other genes were
karowicz, 1972; Piekarowicz et al., 1974). Phase variation down-regulated, and seven, up-regulated (Srikhanta et al.,
of HindI is driven by a GACGA pentanucleotide repeat 2005). In the same vein, N. gonorrhoeae formed signifi-
tract in the methyltransferase subunit gene (hsdM), as cantly thicker biofilms and thus may indirectly benefit
supported by the finding that an hsdM allele with four from an increased resistance to external stresses, when its
repeat units encoding an HsdM protein of normal length mod was in the OFF state. Additionally, a mod-ON
was associated with resistance to phage HP1c1 infection, phenotype resulted in an increased ability to associate
while gain or loss of one repeat unit resulted in phage with human cervical epithelial (pex) cells, whereas the
sensitivity (Zaleski et al., 2005). Interestingly, phase mod-OFF configuration enhanced the ability to invade
switching of phage susceptibility could also independently and survive within pex cells following invasion (Srikhanta
be conferred by LPS alterations induced by the variable et al., 2009). Further studies will be needed to clarify
tetranucleotide repeat tract of the lic2A gene, which con- whether these phenotypes directly relate to mod deficiency
firmed the role of Lic2A-modified LPS as the receptor of itself or to altered expression of one or more members of
HP1c1 phage (Zaleski et al., 2005; see also TRs within the mod phasevarion.
LPS biosynthesis genes). Moreover, the frequencies of the Interestingly, most N. meningitidis and N. gonorrhoeae
phase variations of phage susceptibility described above strains have a second phase-variable methyltransferase
are affected by Dam (deoxyadenosine methyltransferase) gene (Srikhanta et al., 2009), and some, even a third
activity, although the mechanism is not clear yet (Zaleski (Seib et al., 2011), and it has been anticipated that the
et al., 2005). combinatorial use of different phasevarions may contrib-
Another example was found in the NgoAV type I R-M ute to further phenotypic variability (Seib et al., 2011).
system of N. gonorrhoeae, which is encoded by four genes: Furthermore, Fox et al. (2007) suggested that the
hsdMNgoAV, hsdRNgoAV, hsdSNgoAV1, and hsdSNgoAV2. The apparent evolution of this type III R-M system into an

FEMS Microbiol Rev 38 (2014) 119–141 ª 2013 Federation of European Microbiological Societies.
Published by John Wiley & Sons Ltd. All rights reserved
132 K. Zhou et al.

epigenetic mechanism for controlling gene expression has III effectors are currently classified into nearly 40 groups
resulted in loss of the DNA restriction function in some based on sequence similarity and biochemical activity,
strains. In the latter context, it is also interesting to note and the largest group is the AvrBs3/PthA family, also
the existence of solitary type IV restriction endonucleases known as transcription activator-like effector (TALE)
that have no cognate methyltransferase. Because these family (reviewed in White et al., 2009). The most con-
peculiar enzymes display a specificity for methylated spicuous feature of TALE genes is the variation of their
DNA, it has been anticipated that they might function to central domain, mostly consisting of 15.5–19.5 nearly
ward off the lateral acquisition of methyltransferases that identical TRs with a 102-bp unit (G€ urlebeck et al., 2006;
might affect the cell’s epigenetic regulation (Fukuda et al., Mak et al., 2013; Fig. 6).
2008; Tesfazgi Mebrhatu et al., 2011). As such, the phase A recent study revealed that the AvrBs3 can determine
variability of methyltransferase functions might serve to bacterial fitness in plants during infection (Kay et al.,
prevent detection of their activity during their establish- 2007). A set of upa (up-regulated by AvrBs3) genes among
ment in a new host (Tesfazgi Mebrhatu et al., 2011). which upa20, the key regulator of the plant cell hypertro-
phy phenotype, has been identified as AvrBs3 targets in
pepper plants. AvrBs3 acts as a transcription factor by
TRs within transcription activator-like effectors
binding to a conserved promoter element (UPA box) of
Gram-negative plant-pathogenic bacteria of the genus upa20, resulting in up-regulation. Binding was shown to be
Xanthomonas can infect a broad spectrum of plants. A mediated by the TR region of AvrBs3, suggesting the
common feature of their infection process is the injection repeats to act as a DNA-binding motif (Kay et al., 2007).
of virulence proteins (termed effectors) into host cells, More specifically, it was found that the number of base
mostly by means of a type III secretion system. The type pairs of the UPA box closely matches the TR copy number

(a)

TTSS DNA-binding TR domain TranscripƟonal


secreƟon acƟvaƟon
signal 0 1 17.5 domain

N HD NG NS NG NI NI NI HD HD NG NS NS HD HD HD NG HD NG C
Nuclear
localizaƟon
signal
LT P E Q V VA I A S H D G G KQ A L E T V Q R L L P V L C Q A H G
1 12/13 34

(b)

UPA box T A T A T AA A C C T NN C CC T C T
UPA gene

Fig. 6. Domain organization and molecular function of Xanthomonas TALE AvrBs3. (a) The AvrBs3 functional domains shown include a type
three secretion system (TTSS) signal sequence (dark blue), a central DNA-binding TR domain, a nuclear localization signal (purple), and a
transcriptional activation domain (olive green). The TR domain in this case comprises 17.5 imperfect repeat units, each of which consists of 34
amino acids (aa). The unit numbered zero (red, dashed rectangle) is not a true repeat because it has a different aa sequence, but it also
contributes to DNA binding. Each repeat binds to a base in the target sequence, and the binding specificity of the repeats is determined by aa
12 and 13 (known as RVD) and displayed with repeat-specific colors. The complete aa sequence of one repeat (no. 9) is shown with its RVD
highlighted in green. (b) After injection into the plant cell by the bacterial type three secretion system, AvrBs3 is targeted to the nucleus and will
bind with its TR domain to a specific DNA sequence known as UPA box. The consensus UPA box matches closely the binding specificity of the TR
region of AvrBs3 as determined by the different TR units and their RVD (aa 12 and 13). AvrBs3 binding activates transcription of several UPA
genes (adapted from Mak et al., 2013).

ª 2013 Federation of European Microbiological Societies. FEMS Microbiol Rev 38 (2014) 119–141
Published by John Wiley & Sons Ltd. All rights reserved
Variable tandem repeats and bacterial adaptation 133

in AvrBs3. An elegant study demonstrated that each repeat expression of the clp genes (Karatzas et al., 2003, 2005).
unit of AvrBs3 binds to one base pair of the UPA box and This alteration confers increased resistance to high hydro-
that the base recognition specificity of each repeat is deter- static pressure, heat, acid, and H2O2, but attenuates viru-
mined by two hypervariable amino acids (the 12th and lence in L. monocytogenes (Karatzas & Bennik, 2002).
13th amino acids of each repeat unit), known as repeat var- Another example is mutL, which is involved in mis-
iable di-residues (RVDs; Boch et al., 2009). Moreover, a match repair in Salmonella Typhimurium and E. coli. The
minimum of 6.5 TRs in AvrBs3 are necessary for activating functional allele of mutL carries a trimeric hexanucleotide
upa gene expression (Boch et al., 2009; Moscou & Bogda- repeat in the region encoding the ATP-binding pocket of
nove, 2009; Scholze & Boch, 2011; Fig. 6). Most recently, the protein. Spontaneous loss of one repeat unit resulting
the structural basis for this kind of sequence-specific recog- in a mutator phenotype due to MutL deficiency has been
nition has been uncovered by crystallographic analysis of observed in long-term cultures of both E. coli and
an artificially engineered TAL effector hybridized with its S.Typhimurium (Shaver & Sniegowski, 2003; Chen et al.,
target DNA (Deng et al., 2012). AvrBs3-dependent modu- 2010). Expansion of this TR region from 3 to 4 units and
lation of plant signaling pathways causes enlargement of from 2 to 3 units has also been observed and, in the
mesophyll cells and hypertrophy of the infected tissue, latter case, caused reversion of the mutator phenotype
which might help the bacteria to proliferate and escape (Shaver & Sniegowski, 2003; Chen et al., 2010; Le Bars
from infection sites to facilitate bacterial spreading (Kay et al., 2013). This genetic switching of MutL may serve as
et al., 2007; Boch & Bonas, 2010). Consequently, TR varia- a strategy to control the balance between genetic stability
tions in the avrBs3 gene preclude activation of the UPA and mutability and thus serve as an element controlling
box causing a failure in inducing the hypersensitive bacterial evolution. Interestingly, not only mutL, but also
response in plants (Kay et al., 2007). many other genes (i.e. mutT, mutY, mutS, dinJ, and ruvC)
While the above studies highlight the biological impor- involved in DNA repair of E. coli harbor SSRs, further
tance of TALEs as major virulence determinants, TALEs underscoring the putative regulatory role of TRs in stress
also provide interesting avenues for biotechnological appli- response (Rocha et al., 2002).
cations. As such, it will be possible to engineer broader Some membrane proteins are essential for membrane
pathogen resistance in crops by combining several UPA integrity and as such for tolerance to a variety of toxic
boxes into the promoter of plant resistance genes like Bs3, chemicals and other stresses. One such protein, that is pres-
which will render these transgenic plants resistant to infec- ent in many Gram-negative bacteria, is TolA, which har-
tion by bacteria delivering matching TAL effectors (Boch bors a variable TR tract composed of 8–16 imperfect copies
& Bonas, 2010; Scholze & Boch, 2011). Alternatively, the of a 15- to 18-bp repeat unit in E. coli (Levengood et al.,
TR region of TAL effectors can be tailored as to recognize 1991; Zhou et al., 2012b). TR variations in TolA occurred
and bind to a predefined DNA sequence, resulting in acti- at a frequency of at least 6.9 9 10 5 in a clonal wild-type
vating of the expression of downstream target genes (Mor- population of E. coli MG1655 and were shown to modulate
bitzer et al., 2010). Moreover, engineered TAL effectors stress tolerance, with the most outspoken TR-dependent
can be fused with endonuclease domains to generate TAL phenotype being deoxycholic acid tolerance (Zhou et al.,
effector nucleases that can introduce cuts or double-strand 2012a). However, the precise molecular mechanism under-
breaks in or near specific sequences on the chromosome, lying this phenotypic variation remains unclear.
targeting these loci for mutagenesis or recombinational A peculiar case is the triplet repeat (TCT) in E. coli
repair and gene therapy (Bogdanove & Voytas, 2011; ahpC, where expansion of the TR tract with 1 unit con-
Mu~ noz Bodnar et al., 2013). verts the AhpC protein from a peroxidase into a disulfide
reductase, as demonstrated by the ability of this newly
acquired enzyme activity to restore normal growth of a
TRs within stress response genes
mutant lacking thioredoxin and glutathione reductase
Regulation of stress response genes is one of the most (Ritz et al., 2001). To our knowledge, this is the only
common strategies employed by bacteria to cope with example of an intragenic TR variation generating a truly
stresses. TRs have been identified in a number of stress novel function in bacteria.
response genes (Rocha et al., 2002), and some studies
have addressed the role of these repeats in the modula-
Conclusions and outlook: TRs and
tion of a stress response. For example, the gene encoding
bacterial evolution
the CtsR regulator (class III stress gene repressor) of
Listeria monocytogenes carries a triplet repeat (GGT) tract The generation of mutations is the basis of evolution in
with three copies. Stochastic deletion of one triplet in the bacteria and all other living organisms. Under changing
ctsR gene results in an inactive CtsR repressor, leading to environmental conditions, the evolution of better adapted

FEMS Microbiol Rev 38 (2014) 119–141 ª 2013 Federation of European Microbiological Societies.
Published by John Wiley & Sons Ltd. All rights reserved
134 K. Zhou et al.

descendants may be essential for survival of the species, Although rapid TR-dependent phase switching facili-
and an increased mutation rate obviously could confer tates bacterial adaption on a short time scale, it is proba-
higher survival chances. On the other hand, the genera- bly not necessary for bacteria to maintain hypermutable
tion of random mutations all over the genome also causes regions in the absence of selection pressures, because of
an important burden because most mutations are delete- the associated cost of DNA replication fidelity and meta-
rious. Therefore, to be successful, bacteria will have to bolic energy. An interesting, but mostly unanswered ques-
maintain a balance between genome plasticity and stabil- tion is how bacteria optimize the mutation rate of TRs in
ity. One way to do this is to control the spatial distribu- phase-variable genes in a fluctuating environment. Some
tion of mutations over the genome by evolving highly intrinsic features of the TR sequence (i.e. conservation
mutable sequences that are associated with loci (also and copy number of repeat unit) and cellular function
known as contingency loci) that are needed for a flexible (i.e. DNA replication, recombination, and repair) are
response to environmental conditions or stresses, espe- known to affect the frequency of TR-dependent phase
cially those that cannot be detected by conventional variation and may be used to this purpose (Bayliss,
bacterial sensors (e.g. phages or host receptors; Fonville 2009). On the other hand, some studies have shown a
et al., 2011). TRs and their variability provide a paradigm modulation of the TR rearrangement frequency by envi-
for this regulatory strategy of adaptability. ronmental stress (Kanbashi et al., 1997; Jackson & Loeb,
The formation of TRs is suggested to be a random pro- 2000; Rasmussen & Bj€ ork, 2001; Srikhanta et al., 2009;
cess based on replication slippage (Levinson & Gutman, Cooley et al., 2010). However, in general, it remains
1987); however, only some of the formed TRs are unclear how environmental signals are transduced to
believed to be maintained during evolution. Generally, modulate the switching rate of TR-containing genes.
variable TRs tend to localize in flexible genes involved in In conclusion, TRs confer local sequence flexibility in
the biosynthesis of surface structures and with a function bacterial genomes, thereby allowing targeted mutation
in adhesion and (for pathogens) invasion, although they and evolution. The genotypic and phenotypic variations
are occasionally also found in genes encoding critical cel- modulated by TRs are rapid and coordinative and sup-
lular functions like DNA replication (Moxon et al., 2006; port the generation of substantial biological diversity on a
Guo & Mrazek, 2008). The association between TRs and short time scale. In spite of a certain metabolic cost, ‘pre-
cell surface structures is suggested to allow populations to pared genomes’ (i.e. with TR-based contingency loci;
anticipate changes in the environment in order to Caporale, 1999) have a higher adaptability and thus a fit-
enhance their survival rate (Moxon et al., 1994). This is ness advantage in frequently fluctuating environments.
particularly common and critical for pathogens such as
H. influenzae and H. pylori with limited genetic informa-
tion (i.e. reduced genome) to cope with complex environ- Acknowledgements
ments (Razin et al., 1998). This work was supported by the Research Foundation –
As such, TR-dependent phase variation can be regarded Flanders (FWO – Vlaanderen; Research project G.0289.06)
as a strategy for bacterial adaptation that is complemen- and by the KU Leuven Research Fund (project METH/07/
tary to conventional mutations (i.e. single nucleotide 03).
polymorphism), but has some distinct features. First, the
hypermutability (typical mutation rates of 10 2–10 5 per
generation) can be advantageous for adaption on a short References
time scale, and it has been demonstrated using a theoreti-
cal model that TRs mediating stochastic switching can Ackermann M & Chao L (2006) DNA sequences shaped by
evolve and be maintained under a wide range of alternat- selection for stability. PLoS Genet 2: e22.
Adamczyk-Poplawska M, Lower M & Piekarowicz A (2011)
ing selection regimens (Palmer et al., 2013). Second, TR
Deletion of one nucleotide within the homonucleotide tract
rearrangements, both local and combinatorial, can effec-
present in the hsdS gene alters the DNA sequence specificity
tuate alterations at both the transcriptional and the of type I restriction-modification system NgoAV. J Bacteriol
translational levels, resulting in either binary switching 193: 6750–9675.
(‘ON’ and ‘OFF’) or gradual control, and this facilitates Aertsen A & Michiels CW (2004) Stress and how bacteria cope
subtle adaptation of bacterial fitness under stress. Further, with death and survival. Crit Rev Microbiol 30: 263–273.
TR-dependent mechanisms operate not only in classic Aertsen A & Michiels CW (2005) Diversify or die: generation
genetic, but also in epigenetic pathways and from the sin- of diversity in response to stress. Crit Rev Microbiol 31:
gle locus to the more global level of phasevarions, sug- 69–78.
gesting the power of TR intermediate regulation in Appelmelk BJ, Martin SL, Monteiro MA et al. (1999) Phase
bacterial adaptation. variation in Helicobacter pylori lipopolysaccharide due to

ª 2013 Federation of European Microbiological Societies. FEMS Microbiol Rev 38 (2014) 119–141
Published by John Wiley & Sons Ltd. All rights reserved
Variable tandem repeats and bacterial adaptation 135

changes in the lengths of poly(C) tracts in Boch J & Bonas U (2010) Xanthomonas AvrBs3 family-type III
alpha3-fucosyltransferase genes. Infect Immun 67: effectors: discovery and function. Annu Rev Phytopathol 48:
5361–5366. 419–436.
Attia AS & Hansen EJ (2006) A conserved tetranucleotide Boch J, Scholze H, Schornack S, Landgraf A, Hahn S, Kay S,
repeat is necessary for wild-type expression of the Moraxella Lahaye T, Nickstadt A & Bonas U (2009) Breaking the code
catarrhalis UspA2 protein. J Bacteriol 188: 7840–7852. of DNA binding specificity of TAL-type III effectors. Science
Backert S, Clyne M & Tegtmeyer N (2011) Molecular 326: 1509–1512.
mechanisms of gastric epithelial cell adhesion and injection Bogdanove AJ & Voytas DF (2011) TAL effectors:
of CagA by Helicobacter pylori. Cell Commun Signal 9: 28. customizable proteins for DNA targeting. Science 333:
Barenkamp SJ & Leininger E (1992) Cloning, expression, 1843–1846.
and DNA sequence analysis of genes encoding Brinkmann B, Klintschar M, Neuhuber F, H€ uhne J & Rolf B
nontypeable Haemophilus influenzae high-molecular-weight (1998) Mutation rate in human microsatellites: influence of
surface-exposed proteins related to filamentous the structure and length of the tandem repeat. Am J Hum
hemagglutinin of Bordetella pertussis. Infect Immun 60: Genet 62: 1408–1415.
1302–1313. Bzymek M & Lovett ST (2001) Instability of repetitive DNA
Bartley SN, Tzeng YL, Heel K et al. (2013) Attachment and sequences: the role of replication in multiple mechanisms. P
invasion of Neisseria meningitidis to host cells is related to Natl Acad Sci USA 98: 8319–8325.
surface hydrophobicity, bacterial cell size and capsule. PLoS Callaghan MJ, Jolley KA & Maiden MC (2006)
ONE 8: e55798. Opacity-associated adhesin repertoire in hyperinvasive
Bayliss CD (2009) Determinants of phase variation rate and Neisseria meningitidis. Infect Immun 74: 5085–5094.
the fitness implications of differing rates for bacterial Callaghan MJ, Buckee CO, Jolley KA, Kriz P, Maiden MC &
pathogens and commensals. FEMS Microbiol Rev 33: Gupta S (2008) The effect of immune selection on the
504–520. structure of the meningococcal Opa protein repertoire. PLoS
Bayliss CD, Field D & Moxon ER (2001) The simple sequence Pathog 4: e1000020.
contingency loci of Haemophilus influenzae and Neisseria Caporale LH (1999) Chance favors the prepared genome. Ann
meningitidis. J Clin Invest 107: 657–662. NY Acad Sci 870: 1–21.
Bayliss CD, Hoe JC, Makepeace K, Martin P, Hood DW & Carbonnelle E, Hill DJ, Morand P, Griffiths NJ, Bourdoulous
Moxon ER (2008) Neisseria meningitidis escape from the S, Murillo I, Nassif X & Virji M (2009) Meningococcal
bactericidal activity of a monoclonal antibody is mediated interactions with the host. Vaccine 27(suppl 2): B78–B89.
by phase variation of lgtG and enhanced by a mutator Carson SD, Stone B, Beucher M, Fu J & Sparling PF (2000)
phenotype. Infect Immun 76: 5038–5048. Phase variation of the gonococcal siderophore receptor
Bayliss CD, Bidmos FA, Anjum A et al. (2012) Phase variable FetA. Mol Microbiol 36: 585–593.
genes of Campylobacter jejuni exhibit high mutation rates Chen F, Liu WQ, Eisenstark A, Johnston RN, Liu GR & Liu
and specific mutational patterns but mutability is not the SL (2010) Multiple genetic switches spontaneously
major determinant of population structure during host modulating bacterial mutability. BMC Evol Biol 10: 277.
colonization. Nucleic Acids Res 40: 5876–5889. Chiou CS (2010) Multilocus variable-number tandem repeat
Benson G (1999) Tandem repeats finder: a program to analyze analysis as a molecular tool for subtyping and phylogenetic
DNA sequences. Nucleic Acids Res 27: 573–580. analysis of bacterial pathogens. Expert Rev Mol Diagn 10:
̈
Bergfeld AK, Claus H, Vogel U & Muhlenhoff M (2007) 5–7.
Biochemical characterization of the polysialic acid specific Cholon DM, Cutter D, Richardson SK, Sethi S, Murphy TF,
O-acetyltransferase NeuO of Escherichia coli K1. J Biol Chem Look DC & St Geme III JW (2008) Serial isolates of
282: 22217–22227. persistent Haemophilus influenzae in patients with chronic
Berrington AW, Tan YC, Srikhanta Y, Kuipers B, van der Ley obstructive pulmonary disease express diminishing
P, Peak IR & Jennings MP (2002) Phase variation in quantities of the HMW1 and HMW2 adhesins. Infect
meningococcal lipooligosaccharide biosynthesis genes. FEMS Immun 76: 4463–4468.
Immunol Med Microbiol 34: 267–275. Choudhary OP & Trivedi S (2010) Microsatellite or simple
Bhugra B, Voelker LL, Zou N, Yu H & Dybvig K (1995) sequence repeat (SSR) instability depends on repeat
Mechanism of antigenic variation in Mycoplasma pulmonis: characteristics during replication and repair. J Cell Mol Biol
interwoven, site-specific DNA inversions. Mol Microbiol 18: 8: 21–34.
703–714. Coenye T & Vandamme P (2005) Characterization of
Bi X & Liu LF (1996) A replicational model for DNA mononucleotide repeats in sequenced prokaryotic genomes.
recombination between direct repeats. J Mol Biol 256: DNA Res 12: 221–233.
849–858. Coil DA & Anne J (2010) The role of fimV and the
Bichara M, Wagner J & Lambert IB (2006) Mechanisms of importance of its tandem repeat copy number in twitching
tandem repeat instability in bacteria. Mutat Res 598: motility, pigment production, and morphology in Legionella
144–163. pneumophila. Arch Microbiol 192: 625–631.

FEMS Microbiol Rev 38 (2014) 119–141 ª 2013 Federation of European Microbiological Societies.
Published by John Wiley & Sons Ltd. All rights reserved
136 K. Zhou et al.

Cooley MB, Carychao D, Nguyen K, Whitehand L & Mandrell Foster PL (2005) Stress responses and genetic variation in
R (2010) Effects of environmental stress on stability of bacteria. Mutat Res 569: 3–11.
tandem repeats in Escherichia coli O157:H7. Appl Environ Foster PL (2007) Stress-induced mutagenesis in bacteria. Crit
Microbiol 76: 3398–3400. Rev Biochem Mol Biol 42: 373–397.
Dawid S, Barenkamp SJ & St Geme III JW (1999) Variation in Fox KL, Yildirim HH, Deadman ME, Schweda EK, Moxon ER
expression of the Haemophilus influenzae HMW adhesins: a & Hood DW (2005) Novel lipopolysaccharide biosynthetic
prokaryotic system reminiscent of eukaryotes. P Natl Acad genes containing tetranucleotide repeats in Haemophilus
Sci USA 96: 1077–1082. influenzae, identification of a gene for adding O-acetyl
De Bolle X, Bayliss CD, Field D, van de Ven T, Saunders NJ, groups. Mol Microbiol 58: 207–216.
Hood DW & Moxon ER (2000) The length of a Fox KL, Srikhanta YN & Jennings MP (2007) Phase variable
tetranucleotide repeat tract in Haemophilus influenzae type III restriction-modification systems of host-adapted
determines the phase variation rate of a gene with bacterial pathogens. Mol Microbiol 65: 1375–1379.
homology to type III DNA methyltransferases. Mol Fukuda E, Kaminska KH, Bujnicki JM & Kobayashi I (2008)
Microbiol 35: 211–222. Cell death upon epigenetic genome methylation: a novel
de Vries N, Duinsbergen D, Kuipers EJ, Pot RG, Wiesenekker function of methyl-specific deoxyribonucleases. Genome Biol
P, Penn CW, van Vliet AH, Vandenbroucke-Grauls CM & 9: R163.
Kusters JG (2002) Transcriptional phase variation of a type Funchain P, Yeung A, Stewart JL, Lin R, Slupska MM & Miller
III restriction-modification system in Helicobacter pylori. JH (2000) The consequences of growth of a mutator strain of
J Bacteriol 184: 6615–6623. Escherichia coli as measured by loss of function among
Deng D, Yan C, Pan X, Mahfouz M, Wang J, Zhu JK, Shi Y multiple gene targets and loss of fitness. Genetics 154: 959–970.
& Yan N (2012) Structural basis for sequence-specific Gawel D, Jonczyk P, Bialoskorska M, Schaaper RM &
recognition of DNA by TAL effectors. Science 335: Fijalkowska IJ (2002) Asymmetry of frameshift mutagenesis
720–723. during leading and lagging-strand replication in Escherichia
Deszo EL, Steenbergen SM, Freedberg DI & Vimr ER (2005) coli. Mutat Res 501: 129–136.
Escherichia coli K1 polysialic acid O-acetyltransferase gene, Gemayel R, Vinces MD, Legendre M & Verstrepen KJ (2010)
neuO, and the mechanism of capsule form variation Variable tandem repeats accelerate evolution of coding and
involving a mobile contingency locus. P Natl Acad Sci USA regulatory sequences. Annu Rev Genet 44: 445–477.
102: 5564–5569. Gibbons JG & Rokas A (2009) Comparative and functional
Dettman JR & Taylor JW (2004) Mutation and evolution of characterization of intragenic tandem repeats in 10
microsatellite loci in Neurospora. Genetics 168: 1231–1248. Aspergillus genomes. Mol Biol Evol 26: 591–602.
Dixon K, Bayliss CD, Makepeace K, Moxon ER & Hood DW Glew MD, Baseggio N, Markham PF, Browning GF & Walker
(2007) Identification of the functional initiation codons of a ID (1998) Expression of the pMGA genes of Mycoplasma
phase-variable gene of Haemophilus influenzae, lic2A, with gallisepticum is controlled by variation in the GAA
the potential for differential expression. J Bacteriol 189: trinucleotide repeat lengths within the 5′ noncoding regions.
511–521. Infect Immun 66: 5833–5841.
Ecevit IZ, McCrea KW, Pettigrew MM, Sen A, Marrs CF & Glew MD, Browning GF, Markham PF & Walker ID (2000)
Gilsdorf JR (2004) Prevalence of the hifBC, hmw1A, hmw2A, pMGA phenotypic variation in Mycoplasma gallisepticum
hmwC, and hia genes in Haemophilus influenzae isolates. occurs in vivo and is mediated by trinucleotide repeat length
J Clin Microbiol 42: 3065–3072. variation. Infect Immun 68: 6027–6033.
Eckert KA & Hile SE (2009) Every microsatellite is different: Glover SW & Piekarowicz A (1972) Host specificity of DNA in
intrinsic DNA features dictate mutagenesis of common Haemophilus influenzae: restriction and modification in
microsatellites present in the human genome. Mol Carcinog strain Rd. Biochem Biophys Res Commun 46: 1610–1617.
48: 379–388. Goldstein DB & Clark AG (1995) Microsatellite variation in
Edwards YJ, Elgar G, Clark MS & Bishop MJ (1998) The North American populations of Drosophila melanogaster.
identification and characterization of microsatellites in the Nucleic Acids Res 23: 3882–3886.
compact genome of the Japanese pufferfish, Fugu rubripes: Goodwin AC, Weinberger DM, Ford CB, Nelson JC, Snider
perspectives in functional and comparative genomic JD, Hall JD, Paules CI, Peek Jr RM & Forsyth MH (2008)
analyses. J Mol Biol 278: 843–854. Expression of the Helicobacter pylori adhesin SabA is
Erwin AL, Bonthuis PJ, Geelhood JL, Nelson KL, McCrea KW, controlled via phase variation and the ArsRS signal
Gilsdorf JR & Smith AL (2006) Heterogeneity in tandem transduction system. Microbiology 154: 2231–2240.
octanucleotides within Haemophilus influenzae Gravekamp C, Horensky DS, Michel JL & Madoff LC (1996)
lipopolysaccharide biosynthetic gene losA affects serum Variation in repeat number within the alpha C protein of
resistance. Infect Immun 74: 3408–3414. group B streptococci alters antigenicity and protective
Fonville NC, Ward RM & Mittelman D (2011) Stress-induced epitopes. Infect Immun 64: 3576–3583.
modulators of repeat instability and genome evolution. Gravekamp C, Kasper DL, Michel JL, Kling DE, Carey V &
J Mol Microbiol Biotechnol 21: 36–44. Madoff LC (1997) Immunogenicity and protective efficacy

ª 2013 Federation of European Microbiological Societies. FEMS Microbiol Rev 38 (2014) 119–141
Published by John Wiley & Sons Ltd. All rights reserved
Variable tandem repeats and bacterial adaptation 137

of the alpha C protein of group B streptococci are inversely domain of the extracellular adherence protein of
related to the number of repeats. Infect Immun 65: 5216– Staphylococcus aureus is required for aggregation, adherence,
5221. and host cell invasion but not for leukocyte activation.
Griffin R, Cox AD, Makepeace K, Richards JC, Moxon ER & Infect Immun 76: 5615–5623.
Hood DW (2003) The role of lex2 in lipopolysaccharide Ilver D, Arnqvist A, Ogren J et al. (1998) Helicobacter pylori
biosynthesis in Haemophilus influenzae strains RM7004 and adhesin binding fucosylated histo-blood group antigens
RM153. Microbiology 149: 3165–3175. revealed by retagging. Science 279: 373–377.
Guo X & Mrazek J (2008) Long simple sequence repeats in Iyer RR, Pluciennik A, Rosche WA, Sinden RR & Wells RD
host-adapted pathogens localize near genes encoding (2000) DNA polymerase III proofreading mutants enhance
antigens, housekeeping genes, and pseudogenes. J Mol Evol the expansion and deletion of triplet repeat sequences in
67: 497–509. Escherichia coli. J Biol Chem 275: 2174–2184.
Gur-Arie R, Cohen CJ, Eitan Y, Shelef L, Hallerman EM & Jackson AL & Loeb LA (2000) Microsatellite instability
Kashi Y (2000) Simple sequence repeats in Escherichia coli: induced by hydrogen peroxide in Escherichia coli. Mutat Res
abundance, distribution, composition, and polymorphism. 447: 187–198.
Genome Res 10: 62–71. Jansen A, Gemayel R & Verstrepen KJ (2012) Unstable
G€
urlebeck D, Thieme F & Bonas U (2006) Type III effector microsatellite repeats facilitate rapid evolution of coding
proteins from the plant pathogen Xanthomonas and their and regulatory sequences. Genome Dyn 7: 108–125.
role in the interaction with the host plant. J Plant Physiol Janulczyk R, Masignani V, Maione D, Tettelin H, Grandi G &
163: 233–255. Telford JL (2010) Simple sequence repeats and genome
Hammerschmidt S, Hilse R, van Putten JP, Gerardy-Schahn R, plasticity in Streptococcus agalactiae. J Bacteriol 192:
Unkmeir A & Frosch M (1996a) Modulation of cell surface 3990–4000.
sialic acid expression in Neisseria meningitidis via a Jennings MP, Srikhanta YN, Moxon ER, Kramer M, Poolman
transposable genetic element. EMBO J 15: 192–198. JT, Kuipers B & van der Ley P (1999) The genetic basis of
Hammerschmidt S, M€ uller A, Sillmann H, M€ uhlenhoff M, the phase variation repertoire of lipopolysaccharide
Borrow R, Fox A, van Putten J, Zollinger WD, immunotypes in Neisseria meningitidis. Microbiology 145:
Gerardy-Schahn R & Frosch M (1996b) Capsule phase 3013–3021.
variation in Neisseria meningitidis serogroup B by Jerome JP, Bell JA, Plovanich-Jones AE, Barrick JE, Brown CT
slipped-strand mispairing in the polysialyltransferase gene & Mansfield LS (2011) Standing genetic variation in
(siaD): correlation with bacterial invasion and the contingency loci drives the rapid adaptation of
outbreak of meningococcal disease. Mol Microbiol 20: Campylobacter jejuni to a novel host. PLoS ONE 6: e16399.
1211–1220. Jin H, Ren Z, Whitby PW, Morton DJ & Stull TL (1999)
Hannan A (2010) TRPing up the genome: tandem repeat Characterization of hgpA, a gene encoding a haemoglobin/
polymorphisms as dynamic sources of genetic variability in haemoglobin-haptoglobin-binding protein of Haemophilus
health and disease. Discov Med 10: 314–321. influenzae. Microbiology 145: 905–914.
Hebert ML & Wells RD (2005) Roles of double-strand breaks, Jolivet-Gougeon A, Kovacs B, Le Gall-David S, Le Bars H,
nicks, and gaps in stimulating deletions of CTG.CAG Bousarghin L, Bonnaure-Mallet M, Lobel B, Guille F, Soussy
repeats by intramolecular DNA repair. J Mol Biol 353: CJ & Tenke P (2011) Bacterial hypermutation: clinical
961–979. implications. J Med Microbiol 60: 563–573.
Hebert ML, Spitz LA & Wells RD (2004) DNA double-strand Jonsson AB, Nyberg G & Normark S (1991) Phase variation of
breaks induce deletion of CTG.CAG repeats in an gonococcal pili by frameshift mutation in pilC, a novel gene
orientation-dependent manner in Escherichia coli. J Mol Biol for pilus assembly. EMBO J 10: 477–488.
336: 655–672. Jonsson AB, Pfeifer J & Normark S (1992) Neisseria
Herbers K, Conrads-Strauch J & Bonas U (1992) gonorrhoeae PilC expression provides a selective mechanism
Race-specificity of plant resistance to bacterial spot disease for structural diversity of pili. P Natl Acad Sci USA 89:
determined by repetitive motifs in a bacterial avirulence 3204–3208.
protein. Nature 356: 172–174. Jordan P, Snyder LA & Saunders NJ (2003) Diversity in
Hood DW, Deadman ME, Jennings MP, Bisercic M, coding tandem repeats in related Neisseria spp. BMC
Fleischmann RD, Venter JC & Moxon ER (1996) DNA Microbiol 3: 23.
repeats identify novel virulence genes in Haemophilus Kajava AV (2012) Tandem repeats in proteins: from sequence
influenzae. Proc Natl Acad Sci USA 93: 11121–11125. to structure. J Struct Biol 179: 279–288.
Hosking SL, Craig JE & High NJ (1999) Phase variation of Kanbashi K, Wang X, Komura J, Ono T & Yamamoto K
lic1A, lic2A and lic3A in colonization of the nasopharynx, (1997) Frameshifts, base substitutions and minute deletions
bloodstream and cerebrospinal fluid by Haemophilus constitute X ray-induced mutations in the endogenous tonB
influenzae type b. Microbiology 145: 3005–3011. gene of Escherichia coli K12. Mutat Res 385: 259–267.
Hussain M, Haggar A, Peters G, Chhatwal GS, Herrmann M, Karatzas KA & Bennik MH (2002) Characterization of a
Flock JI & Sinha B (2008) More than one tandem repeat Listeria monocytogenes Scott A isolate with high tolerance

FEMS Microbiol Rev 38 (2014) 119–141 ª 2013 Federation of European Microbiological Societies.
Published by John Wiley & Sons Ltd. All rights reserved
138 K. Zhou et al.

towards high hydrostatic pressure. Appl Environ Microbiol Levengood SK, Beyer WF Jr & Webster RE (1991) TolA: a
68: 3183–3189. membrane protein involved in colicin uptake contains an
Karatzas KA, Wouters JA, Gahan CG, Hill C, Abee T & extended helical region. P Natl Acad Sci USA 88: 5939–5943.
Bennik MH (2003) The CtsR regulator of Listeria Levinson G & Gutman GA (1987) Slipped-strand mispairing: a
monocytogenes contains a variant glycine repeat region that major mechanism for DNA sequence evolution. Mol Biol
affects piezotolerance, stress resistance, motility and Evol 4: 203–221.
virulence. Mol Microbiol 49: 1227–1238. Lewis LA, Gipson M, Hartman K, Ownbey T, Vaughn J &
Karatzas KA, Valdramidis VP & Wells-Bennik MH (2005) Dyer DW (1999) Phase variation of HpuAB and HmbR,
Contingency locus in ctsR of Listeria monocytogenes Scott A: two distinct haemoglobin receptors of Neisseria meningitidis
a strategy for occurrence of abundant piezotolerant isolates DNM2. Mol Microbiol 32: 977–989.
within clonal populations. Appl Environ Microbiol 71: Lim KG, Kwoh CK, Hsu LY & Wirawan A (2013) Review of
8390–8396. tandem repeat search tools: a systematic approach to
Kassai-Jager E, Ortutay C, Toth G, Vellai T & Gaspari Z evaluating algorithmic performance. Brief Bioinform 14:
(2008) Distribution and evolution of short tandem repeats 67–81.
in closely related bacterial genomes. Gene 410: 18–25. Lin WH & Kussell E (2012) Evolutionary pressures on simple
Kay S, Hahn S, Marois E, Hause G & Bonas U (2007) A sequence repeats in prokaryotic coding regions. Nucleic
bacterial effector acts as a plant transcription factor and Acids Res 40: 2399–2413.
induces a cell size regulator. Science 318: 648–651. Lindb€ack T, Secic I & Rørvik LM (2011) A contingency locus
Kelkar YD, Strubczewski N, Hile SE, Chiaromonte F, Eckert in prfA in a Listeria monocytogenes subgroup allows
KA & Makova KD (2010) What is a microsatellite: a reactivation of the PrfA virulence regulator during infection
computational and experimental definition based upon in mice. Appl Environ Microbiol 77: 3478–3483.
repeat mutational behavior at A/T and GT/AC repeats. Lindstedt BA (2005) Multiple-locus variable number tandem
Genome Biol Evol 2: 620–635. repeats analysis for genetic fingerprinting of pathogenic
King MR, Vimr RP, Steenbergen SM, Spanjaard L, Plunkett G bacteria. Electrophoresis 26: 2567–2582.
III, Blattner FR & Vimr ER (2007) Escherichia coli Lindstedt BA (2011) Genotyping of selected bacterial
K1-specific bacteriophage CUS-3 distribution and function enteropathogens in Norway. Int J Med Microbiol 301: 648–653.
in phase-variable capsular polysialic acid O acetylation. J Liu L, Dybvig K, Panangala VS, van Santen VL & French CT
Bacteriol 189: 6447–6456. (2000) GAA trinucleotide repeat region regulates M9/pMGA
Kornberg A, Bertsch LL, Jackson JF & Khorana HG (1964) gene expression in Mycoplasma gallisepticum. Infect Immun
Enzymatic synthesis of deoxyribonucleic acid, XVI. 68: 871–876.
oligonucleotides as templates and the mechanism of their Lopes J, Ribeyre C & Nicolas A (2006) Complex
replication. Proc Natl Acad Sci USA 51: 315–323. minisatellite rearrangements generated in the total or
Kusters JG, van Vliet AHM & Kuipers EJ (2006) Pathogenesis of partial absence of Rad27/hFEN1 activity occur in a single
Helicobacter pylori infection. Clin Microbiol Rev 19: 449–490. generation and are Rad51 and Rad52 dependent. Mol Cell
Lafontaine ER, Cope LD, Aebi C, Latimer JL, McCracken GH & Biol 26: 6675–6689.
Hansen EJ (2000) The UspA1 protein and a second type of Lysnyansky I, Rosengarten R & Yogev D (1996) Phenotypic
UspA2 protein mediate adherence of Moraxella catarrhalis to switching of variable surface lipoproteins in Mycoplasma
human epithelial cells in vitro. J Bacteriol 182: 1364–1373. bovis involves high-frequency chromosomal rearrangements.
Lafontaine ER, Wagner NJ & Hansen EJ (2001) Expression of J Bacteriol 178: 5395–5401.
the Moraxella catarrhalis UspA1 protein undergoes phase Mahdavi J, Sonden B, Hurtig M et al. (2002) Helicobacter
variation and is regulated at the transcriptional level. J pylori SabA adhesin in persistent infection and chronic
Bacteriol 183: 1540–1551. inflammation. Science 297: 573–578.
Lai Y & Sun F (2003) The relationship between microsatellite Mak ANS, Bradley P, Bogdanove AJ & Stoddard BL (2013) Tal
slippage mutation rate and the number of repeat units. Mol effectors: function, structure, engineering and applications.
Biol Evol 20: 2123–2131. Curr Opin Struct Biol 23: 93–99.
Le Bars H, Bousarghin L, Bonnaure-Mallet M & Malkova A & Haber JE (2012) Mutations arising during repair
Jolivet-Gougeon A (2013) Role of a short tandem leucine/ of chromosome breaks. Annu Rev Genet 46: 455–473.
arginine repeat in strong mutator phenotype acquisition in Martin P, van de Ven T, Mouchel N, Jeffries AC, Hood DW &
a clinical isolate of Salmonella Typhimurium. FEMS Moxon ER (2003) Experimentally revised repertoire of
Microbiol Lett 338: 101–106. putative contingency loci in Neisseria meningitidis strain
Leclercq S, Rivals E & Jarne P (2007) Detecting microsatellites MC58: evidence for a novel mechanism of phase variation.
within genomes: significant variation among algorithms. Mol Microbiol 50: 245–257.
BMC Bioinformatics 8: 125. Martin P, Makepeace K, Hill SA, Hood DW & Moxon ER
Legendre M, Pochet N, Pak T & Verstrepen KJ (2007) (2005) Microsatellite instability regulates transcription factor
Sequence-based estimation of minisatellite and microsatellite binding and gene expression. P Natl Acad Sci USA 102:
repeat variability. Genome Res 17: 1787–1796. 3800–3804.

ª 2013 Federation of European Microbiological Societies. FEMS Microbiol Rev 38 (2014) 119–141
Published by John Wiley & Sons Ltd. All rights reserved
Variable tandem repeats and bacterial adaptation 139

Massey RC & Buckling A (2002) Environmental regulation of Nilsson C, Skoglund A, Moran AP, Annuk H, Engstrand L &
mutation rates at specific sites. Trends Microbiol 10: 580– Normark S (2008) Lipopolysaccharide diversity evolving in
584. Helicobacter pylori communities through genetic
Merkel A & Gemmell N (2008) Detecting short tandem modifications in fucosyltransferases. PLoS ONE 3: e3811.
repeats from genome data: opening the software black box. Orsi RH, Bowen BM & Wiedmann M (2010) Homopolymeric
Brief Bioinform 9: 355–366. tracts represent a general regulatory mechanism in
Metruccio MM, Pigozzi E, Roncarati D, Berlanda Scorza F, prokaryotes. Genomics 11: 102.
Norais N, Hill SA, Scarlato V & Delany I (2009) A novel Palmer ME, Lipsitch M, Moxon ER & Bayliss CD (2013)
phase variation mechanism in the meningococcus driven by Broad conditions favor the evolution of phase-variable loci.
a ligand-responsive repressor and differential spacing of mBio 4: e00430–12.
distal promoter elements. PLoS Pathog 5: e1000710. Papazisi L, Gorton TS, Kutish G, Markham PF, Browning GF,
Miller VL, Taylor RK & Mekalanos JJ (1987) Cholera toxin Nguyen DK, Swartzell S, Madan A, Mahairas G & Geary SJ
transcriptional activator toxR is a transmembrane DNA (2003) The complete genome sequence of the avian
binding protein. Cell 48: 271–279. pathogen Mycoplasma gallisepticum strain R(low).
Morbitzer R, R€ omer P, Boch J & Lahaye T (2010) Regulation Microbiology 149: 2307–2316.
of selected genome loci using de novo-engineered P^aques F, Leung WY & Haber JE (1998) Expansions and
transcription activator-like effector (TALE)-type contractions in a tandem repeat induced by double-strand
transcription factors. P Natl Acad Sci USA 107: break repair. Mol Cell Biol 18: 2045–2054.
21617–21622. P^aques F, Richard GF & Haber JE (2001) Expansions and
Mordhorst IL, Claus H, Ewers C et al. (2009) contractions in 36-bp minisatellites by gene conversion in
O-acetyltransferase gene neuO is segregated according to yeast. Genetics 158: 155–166.
phylogenetic background and contributes to environmental Park SF, Purdy D & Leach S (2000) Localized reversible
desiccation resistance in Escherichia coli K1. Environ frameshift mutation in the flhA gene confers phase
Microbiol 11: 3154–3165. variability to flagellin gene expression in Campylobacter coli.
Morton DJ, Whitby PW, Jin H, Ren Z & Stull TL (1999) J Bacteriol 182: 207–210.
Effect of multiple mutations in the hemoglobin- and Pearson CE, Nichol Edamura K & Cleary JD (2005) Repeat
hemoglobin-haptoglobin-binding proteins, HgpA, HgpB, instability: mechanisms of dynamic mutations. Nat Rev
and HgpC, of Haemophilus influenzae type b. Infect Immun Genet 6: 729–742.
67: 2729–2739. Piekarowicz A, Brzezi nski R & Kauc L (1974) Host specificity
Moscou MJ & Bogdanove AJ (2009) A simple cipher governs of DNA in Haemophilus influenzae: the in vivo action of the
DNA recognition by TAL effectors. Science 326: 1501. restriction endonucleases on phage and bacterial DNA. Acta
Moxon ER, Rainey PB, Nowak MA & Lenski RE (1994) Microbiol Pol A 27: 51–65.
Adaptive evolution of highly mutable loci in pathogenic Piekarowicz A, Klyz A, Kwiatek A & Stein DC (2001) Analysis
bacteria. Curr Biol 4: 24–33. of type I restriction modification systems in the
Moxon R, Bayliss C & Hood D (2006) Bacterial contingency Neisseriaceae: genetic organization and properties of the
loci: the role of simple sequence DNA repeats in bacterial gene products. Mol Microbiol 41: 1199–1210.
adaptation. Annu Rev Genet 40: 307–333. Power PM, Sweetman WA, Gallacher NJ, Woodhall MR,
Mrazek J (2006) Analysis of distribution indicates diverse Kumar GA, Moxon ER & Hood DW (2009) Simple
functions of simple sequence repeats in Mycoplasma sequence repeats in Haemophilus influenzae. Infect Genet
genomes. Mol Biol Evol 23: 1370–1385. Evol 9: 216–228.
Mrazek J, Guo X & Shah A (2007) Simple sequence repeats in Rando OJ & Verstrepen KJ (2007) Timescales of genetic and
prokaryotic genomes. Proc Natl Acad Sci USA 104: 8472– epigenetic inheritance. Cell 128: 655–668.
8477. Rasmussen M & Bj€ ork L (2001) Unique regulation of SclB – a
Mudunuri SB & Nagarajaram HA (2007) IMEx: imperfect novel collagen-like surface protein of Streptococcus pyogenes.
microsatellite extractor. Bioinformatics 23: 1181–1187. Mol Microbiol 40: 1427–1438.
Mu~noz Bodnar A, Bernal A, Szurek B & L opez CE (2013) Tell Razin S, Yogev D & Naot Y (1998) Molecular biology and
me a tale of TALEs. Mol Biotechnol 53: 228–235. pathogenicity of mycoplasmas. Microbiol Mol Biol Rev 62:
Murphy GL, Connell TD, Barritt DS, Koomey M & Cannon 1094–1156.
JG (1989) Phase variation of gonococcal protein II: Ren Z, Jin H, Morton DJ & Stull TL (1998) hgpB, a gene
regulation of gene expression by slipped-strand mispairing encoding a second Haemophilus influenzae hemoglobin- and
of a repetitive DNA sequence. Cell 56: 539–547. hemoglobin-haptoglobin-binding protein. Infect Immun 66:
Nilsson C, Skoglund A, Moran AP, Annuk H, Engstrand L & 4733–4741.
Normark S (2006) An enzymatic ruler modulates Lewis Ren Z, Jin H, Whitby PW, Morton DJ & Stull TL (1999)
antigen glycosylation of Helicobacter pylori LPS during Role of CCAA nucleotide repeats in regulation of
persistent infection. P Natl Acad Sci USA 103: 2863–2868. hemoglobin and hemoglobin-haptoglobin binding protein

FEMS Microbiol Rev 38 (2014) 119–141 ª 2013 Federation of European Microbiological Societies.
Published by John Wiley & Sons Ltd. All rights reserved
140 K. Zhou et al.

genes of Haemophilus influenzae. J Bacteriol 181: di-, tri- and tetranucleotide repeats in Drosophila
5865–5870. melanogaster. Mol Biol Evol 15: 1751–1760.
Richard GF & P^aques F (2000) Mini- and microsatellite Schweda EKH, Richards JC, Hood DW & Moxon ER (2007)
expansions: the recombination connection. EMBO Rep 1: Expression and structural diversity of the lipopolysaccharide
122–126. of Haemophilus influenzae: implication in virulence. Int J
Richard GF, Dujon B & Haber JE (1999) Double-strand break Med Microbiol 297: 297–306.
repair can lead to high frequencies of deletions within short Seale TW, Morton DJ, Whitby PW, Wolf R, Kosanke SD,
CAG/CTG trinucleotide repeats. Mol Gen Genet 261: 871–882. VanWagoner TM & Stull TL (2006) Complex role of
Richard GF, Kerrest A & Dujon B (2008) Comparative hemoglobin and hemoglobin-haptoglobin binding proteins
genomics and molecular dynamics of DNA repeats in in Haemophilus influenzae virulence in the infant rat model
eukaryotes. Microbiol Mol Biol Rev 72: 686–727. of invasive infection. Infect Immun 74: 6213–6225.
Richardson AR & Stojiljkovic I (1999) HmbR, a Seib KL, Pigozzi E, Muzzi A, Gawthorne JA, Delany I,
hemoglobin-binding outer membrane protein of Neisseria Jennings MP & Rappuoli R (2011) A novel epigenetic
meningitidis, undergoes phase variation. J Bacteriol 181: regulator associated with the hypervirulent Neisseria
2067–2074. meningitidis clonal complex 41/44. FASEB J 25: 3622–3633.
Ritz D, Lim J, Reynolds CM, Poole LB & Beckwith J (2001) Shaver AC & Sniegowski PD (2003) Spontaneously arising
Conversion of a peroxiredoxin into a disulfide reductase by mutL mutators in evolving Escherichia coli populations are
a triplet repeat expansion. Science 294: 158–160. the result of changes in repeat length. J Bacteriol 185:
Rocha EP (2003) An appraisal of the potential for illegitimate 6076–6082.
recombination in bacterial genomes and its consequences: Sheets AJ & St Geme III JW (2011) Adhesive activity of the
from duplications to genome reduction. Genome Res 13: Haemophilus cryptic genospecies Cha autotransporter is
1123–1132. modulated by variation in tandem peptide repeats. J
Rocha EP, Matic I & Taddei F (2002) Over-representation of Bacteriol 193: 329–339.
repeats in stress response genes: a strategy to increase Sia EA, Kokoska RJ, Dominska M, Greenwell P & Petes TD
versatility under stressful conditions? Nucleic Acids Res 30: (1997) Microsatellite instability in yeast: dependence on
1886–1894. repeat unit size and DNA mismatch repair genes. Mol Cell
Rosengarten R & Wise KS (1991) The Vlp system of Biol 17: 2851–2858.
Mycoplasma hyorhinis: combinatorial expression of distinct Spinosa MR, Progida C, Tala A, Cogli L, Alifano P & Bucci C
size variant lipoproteins generating high-frequency surface (2007) The Neisseria meningitidis capsule is important for
antigenic variation. J Bacteriol 173: 4782–4793. intracellular survival in human cells. Infect Immun 75:
Ryan KA & Lo RY (1999) Characterization of a CACAG 3594–3603.
pentanucleotide repeat in Pasteurella haemolytica and its Srikhanta YN, Maguire TL, Stacey KJ, Grimmond SM &
possible role in modulation of a novel type III Jennings MP (2005) The phasevarion: a genetic system
restriction-modification system. Nucleic Acids Res 27: controlling coordinated, random switching of expression of
1505–1511. multiple genes. P Natl Acad Sci USA 102: 5547–5551.
Sadarangani M, Pollard AJ & Gray-Owen SD (2011) Opa Srikhanta YN, Dowideit SJ, Edwards JL et al. (2009)
proteins and CEACAMs: pathways of immune engagement Phasevarions mediate random switching of gene expression
for pathogenic Neisseria. FEMS Microbiol Rev 35: 498–514. in pathogenic Neisseria. PLoS Pathog 5: e1000400.
Saint-Ruf C & Matic I (2006) Environmental tuning of Srikhanta YN, Fox KL & Jennings MP (2010) The phasevarion:
mutation rates. Environ Microbiol 8: 193–199. phase variation of type III DNA methyltransferases controls
Sarkari J, Pandit N, Moxon ER & Achtman M (1994) Variable coordinated switching in multiple genes. Nat Rev Microbiol
expression of the Opc outer membrane protein in Neisseria 8: 196–206.
meningitidis is caused by size variation of a promoter Srikhanta YN, Gorrell RJ, Steen JA, Gawthorne JA, Kwok T,
containing poly-cytidine. Mol Microbiol 13: 207–217. Grimmond SM, Robins-Browne RM & Jennings MP (2011)
Schaper E, Kajava AV, Hauser A & Anisimova M (2012) Phasevarion mediated epigenetic gene regulation in
Repeat or not repeat? Statistical validation of tandem repeat Helicobacter pylori. PLoS ONE 6: e27569.
prediction in genomic sequences. Nucleic Acids Res 40: Streisinger G, Okada Y, Emrich J, Newton J, Tsugita A,
10005–10017. Terzaghi E & Inouye M (1966) Frameshift mutations and
Schlotterer C, Imhof M, Wang H, Nolte V & Harr B (2006) the genetic code. Cold Spring Harb Symp Quant Biol 31:
Low abundance of Escherichia coli microsatellites is 77–84.
associated with an extremely low mutation rate. J Evol Biol Tauseef I, Harrison OB, Wooldridge KG et al. (2011)
19: 1671–1676. Influence of the combination and phase variation status of
Scholze H & Boch J (2011) TAL effectors are remote controls the haemoglobin receptors HmbR and HpuAB on
for gene activation. Curr Opin Microbiol 14: 47–53. meningococcal virulence. Microbiology 157: 1446–1456.
Schug MD, Hutter CM, Wetterstrand KA, Gaudette MS, Tesfazgi Mebrhatu M, Wywial E, Ghosh A, Michiels CW,
Mackay TF & Aquadro CF (1998) The mutation rates of Lindner AB, Taddei F, Bujnicki JM, Van Melderen L &

ª 2013 Federation of European Microbiological Societies. FEMS Microbiol Rev 38 (2014) 119–141
Published by John Wiley & Sons Ltd. All rights reserved
Variable tandem repeats and bacterial adaptation 141

Aertsen A (2011) Evidence for an evolutionary antagonism Wang G, Rasko DA, Sherburne R & Taylor DE (1999)
between Mrr and Type III modification systems. Nucleic Molecular genetic basis for the variable expression of
Acids Res 39: 5991–6001. Lewis Y antigen in Helicobacter pylori: analysis of the
Theiss P & Wise KS (1997) Localized frameshift mutation alpha (1,2) fucosyltransferase gene. Mol Microbiol 31:
generates selective, high-frequency phase variation of a 1265–1274.
surface lipoprotein encoded by a mycoplasma ABC Wells RD, Dere R, Hebert ML, Napierala M & Son LS (2005)
transporter operon. J Bacteriol 179: 4013–4022. Advances in mechanisms of genetic instability related to
Treangen TJ, Abraham AL, Touchon M & Rocha EP (2009) hereditary neurological diseases. Nucleic Acids Res 33:
Genesis, effects and fates of repeats in prokaryotic genomes. 3785–3798.
FEMS Microbiol Rev 33: 539–571. White FF, Potnis N, Jones JB & Koebnik R (2009) The type III
van Belkum A (1999) Short sequence repeats in microbial effectors of Xanthomonas. Mol Plant Pathol 10: 749–766.
pathogenesis and evolution. Cell Mol Life Sci 56: 729–734. Willems R, Paul A, van der Heide HG, ter Avest AR & Mooi
van der Ende A, Hopman CT & Dankert J (2000) Multiple FR (1990) Fimbrial phase variation in Bordetella pertussis: a
mechanisms of phase variation of PorA in Neisseria novel mechanism for transcriptional regulation. EMBO J 9:
meningitidis. Infect Immun 68: 6685–6690. 2803–2809.
van der Woude MW & Baumler AJ (2004) Phase and antigenic Yamaoka Y, Kita M, Kodama T, Imamura S, Ohno T, Sawai
variation in bacteria. Clin Microbiol Rev 17: 581–611. N, Ishimaru A, Imanishi J & Graham DY (2002)
van Ham SM, van Alphen L, Mooi FR & van Putten JP (1993) Helicobacter pylori infection in mice: role of outer
Phase variation of H. influenzae fimbriae: transcriptional membrane proteins in colonization and inflammation.
control of two divergent genes through a variable combined Gastroenterology 123: 1992–2004.
promoter region. Cell 73: 1187–1196. Yang QL & Gotschlich EC (1996) Variation of gonococcal
van Passel MW & Ochman H (2007) Selection on the genic lipooligosaccharide structure is due to alterations in poly-G
location of disruptive elements. Trends Genet 23: 601–604. tracts in lgt genes encoding glycosyl transferases. J Exp Med
van Selm S, van Cann LM, Kolkman MA, van der Zeijst BA & 183: 323–327.
van Putten JP (2003) Genetic basis for the structural Yang J, Wang J, Chen L, Yu J, Dong J, Yao ZJ, Shen Y, Jin Q
difference between Streptococcus pneumoniae serotype 15B & Chen R (2003) Identification and characterization of
and 15C capsular polysaccharides. Infect Immun 71: 6192– simple sequence repeats in the genomes of Shigella species.
6198. Gene 322: 85–92.
Vandersmissen L, De Buck E, Saels V, Coil DA & Anne J Yogev D, Rosengarten R, Watson-McKown R & Wise KS
(2010) A Legionella pneumophila collagen-like protein (1991) Molecular basis of Mycoplasma surface antigenic
encoded by a gene with a variable number of tandem variation: a novel set of divergent genes undergo
repeats is involved in the adherence and invasion of host spontaneous mutation of periodic coding regions and 5′
cells. FEMS Microbiol Lett 306: 168–176. regulatory sequences. EMBO J 10: 4069–4079.
Verstrepen KJ, Reynolds TB & Fink GR (2004) Origins of Zahra R, Blackwood JK, Sales J & Leach DR (2007)
variation in the fungal cell surface. Nat Rev Microbiol 2: Proofreading and secondary structure processing determine
533–540. the orientation dependence of CAG 9 CTG trinucleotide
Vimr E & Steenbergen SM (2006) Mobile contingency locus repeat instability in Escherichia coli. Genetics 176: 27–41.
controlling Escherichia coli K1 polysialic acid capsule Zaleski P, Wojciechowski M & Piekarowicz A (2005) The role
acetylation. Mol Microbiol 60: 828–837. of Dam methylation in phase variation of Haemophilus
Vogler AJ, Keys C, Nemoto Y, Colman RE, Jay Z & Keim P influenzae genes involved in defence against phage infection.
(2006) Effect of repeat copy number on variable-number Microbiology 151: 3361–3369.
tandem repeat mutations in Escherichia coli O157:H7. J Zhang Q & Wise KS (1997) Localized reversible frameshift
Bacteriol 188: 4253–4263. mutation in an adhesin gene confers a phase-variable
Waite RD, Struthers JK & Dowson CG (2001) Spontaneous adherence phenotype in mycoplasma. Mol Microbiol 25:
sequence duplication within an open reading frame of the 859–869.
pneumococcal type 3 capsule locus causes high-frequency Zhou K, Michiels CW & Aertsen A (2012a) Variation of
phase variation. Mol Microbiol 42: 1223–1232. intragenic tandem repeat tract of tolA modulates Escherichia
Waite RD, Penfold DW, Struthers JK & Dowson CG (2003) coli stress tolerance. PLoS ONE 7: e47766.
Spontaneous sequence duplications within capsule genes Zhou K, Vanoirbeek K, Aertsen A & Michiels CW (2012b)
cap8E and tts control phase variation in Streptococcus Variability of the tandem repeat region of the Escherichia
pneumoniae serotypes 8 and 37. Microbiology 149: 497–504. coli tolA gene. Res Microbiol 163: 316–322.

FEMS Microbiol Rev 38 (2014) 119–141 ª 2013 Federation of European Microbiological Societies.
Published by John Wiley & Sons Ltd. All rights reserved
Europe PMC Funders Group
Author Manuscript
J Mol Evol. Author manuscript; available in PMC 2014 April 14.
Published in final edited form as:
J Mol Evol. 2012 June ; 74(0): 273–280. doi:10.1007/s00239-012-9505-4.
Europe PMC Funders Author Manuscripts

Estimating the Relative Roles of Recombination and Point


Mutation in the Generation of Single Locus Variants in
Campylobacter jejuni and Campylobacter coli
Shoukai Yu,
Infectious Disease Research Centre, Institute of Veterinary, Animal, and Biomedical Sciences,
Massey University, Palmerston North, New Zealand; S.Yu1@massey.ac.nz
Paul Fearnhead,
Department of Mathematics and Statistics, Lancaster University, Lancaster, UK
Barbara R. Holland,
School of Mathematics and Physics, University of Tasmania, Hobart, Australia
Patrick Biggs,
Infectious Disease Research Centre, Institute of Veterinary, Animal, and Biomedical Sciences,
Massey University, Palmerston North, New Zealand
Martin Maiden, and
Department of Zoology, University of Oxford, Oxford, UK
Nigel French
Infectious Disease Research Centre, Institute of Veterinary, Animal, and Biomedical Sciences,
Massey University, Palmerston North, New Zealand; Allan Wilson Centre for Molecular Ecology
and Evolution, Massey University, Palmerston North, New Zealand; n.p.french@massey.ac.nz
Europe PMC Funders Author Manuscripts

Abstract
Single locus variants (SLVs) are bacterial sequence types that differ at only one of the seven
canonical multilocus sequence typing (MLST) loci. Estimating the relative roles of recombination
and point mutation in the generation of new alleles that lead to SLVs is helpful in understanding
how organisms evolve. The relative rates of recombination and mutation for Campylobacter jejuni
and Campylobacter coli were estimated at seven different housekeeping loci from publically
available MLST data. The probability of recombination generating a new allele that leads to an
SLV is estimated to be roughly seven times more than that of mutation for C. jejuni, but for C. coli
recombination and mutation were estimated to have a similar contribution to the generation of
SLVs. The majority of nucleotide differences (98 % for C. jejuni and 85 % for C. coli) between
strains that make up an SLV are attributable to recombination. These estimates are much larger
than estimates of the relative rate of recombination to mutation calculated from more distantly
related isolates using MLST data. One explanation for this is that purifying selection plays an
important role in the evolution of Campylobacter. A simulation study was performed to test the
performance of our method under a range of biologically realistic parameters. We found that our
method performed well when the recombination tract length was longer than 3 kb. For situations
in which recombination may occur with shorter tract lengths, our estimates are likely to be an
underestimate of the ratio of recombination to mutation, and of the importance of recombination
for creating diversity in closely related isolates. A parametric bootstrap method was applied to
calculate the uncertainty of these estimates.

© Springer Science+Business Media, LLC 2012


Correspondence to: Nigel French.
Yu et al. Page 2

Keywords
Single locus variant; MLST; Purifying; selection; Evolution of Campylobacter; Mutation;
Recombination

Introduction
Europe PMC Funders Author Manuscripts

The genus Campylobacter is the major cause of gastroenteritis in many industrialized


countries (Tauxe et al. 1992), with approximately 1 % of the population throughout the
western world being affected by campylobacteriosis every year (The World Health
Organization, cited in Humphrey et al. 2007). The species Campylobacter jejuni (C. jejuni)
and Campylobacter coli (C. coli) are the main causes of bacterial food-borne disease in
developed countries, compared to other members of the family Campylobacteriaceae
(Konkel et al. 1999).

Substantial evidence for the presence of recombination at specific genes has been found in
several studies (Suerbaum et al. 2001; Fearnhead et al. 2005). The relative contributions of
recombination and point mutation to genetic diversity have also been investigated (Feil et al.
1999, 2000,2001; Sarkar and Guttman 2004). Although most research indicates that
recombination contributes more to genetic diversity than mutation, there is considerable
uncertainty about the relative number of events and the number of nucleotide differences
that may be attributable to these two processes (Schouls et al. 2003; Richman et al. 2003;
Fearnhead et al. 2005). This paper is focused on estimating the relative contributions of
recombination and point mutation to the generation of new alleles that lead to single locus
variants (SLVs), based on C. jejuni and C. coli from the seven gene multilocus sequence
typing (MLST) scheme.

An SLV is a pair of sequence types (STs) that differ at exactly one of the seven alleles that
make up the MLST profile (Feil et al. 2004). SLVs are pairs of STs that most likely share a
very recent common ancestor and the analysis of SLVs can be helpful in understanding the
Europe PMC Funders Author Manuscripts

evolution and molecular epidemiology of pathogens. The large collections of isolates that
have been characterized by MLST provide a good opportunity to study SLVs in detail.

This research is based on distinct STs of C. jejuni and C. coli in the PubMLST database
(http://pubmlst.org/campylobacter). In order to understand whether there are differences in
the mechanisms that produce SLVs across the genome, SLVs were divided into groups
depending on the locus at which the STs differ. The distribution of nucleotide differences
within SLVs was explored. The nucleotide differences between two STs that form an SLV
can be generated by two different kinds of events: recombination or mutation. Intuitively,
SLVs that comprise two STs which differ at many nucleotide positions are more likely to be
due to recombination, whereas those that differ at only a few nucleotide positions may be
the result of point mutations. In this study, an EM algorithm was applied to allocate SLVs
into either a point mutation only model or a recombination model. Two key parameters were
estimated: the probability that an SLV arose due to point mutation(s) only, and the relative
rate of recombination to mutation. In order to test the performance of our method, a
simulation study was performed under a range of biologically realistic parameters. When the
recombination tract length was longer than 3 kb our method performed well. Three kilobase
pairs is the average of the estimated mean tract lengths suggested by previous research on
Campylobacter (Schouls et al. 2003; Fearnhead et al. 2005; Wilson et al.2009; Biggs et al.
2011). When recombination occurs with shorter tract lengths, our estimates may
underestimate the ratio of recombination to mutation.

J Mol Evol. Author manuscript; available in PMC 2014 April 14.


Yu et al. Page 3

Materials and Methods


Campylobacter Data
The data were taken from the PubMLST database (September 27, 2010); at that time the
PubMLST database contained 4676 distinct C. jejuni and C. coli STs. MLST is a way of
typing strains that is based on nucleotide sequences (Maiden et al. 1998). Using the MLST
technique (Dingle et al. 2001), these isolates are sequenced at seven housekeeping loci
Europe PMC Funders Author Manuscripts

(aspA, glnA, gltA, glyA, pgm, tkt, and uncA). These seven loci are widely dispersed around
the genome, which means there is a very low chance for one recombination to change two or
more loci.

We separated ST datasets for C. jejuni and C. coli, and excluded the 22 STs found in both
species. Furthermore, we separated C. coli by clades according to previous research
(Sheppard et al. 2008, 2011), and we chose C. coli clade 1 to investigate in detail because C.
coli clade 1 contains more STs, and is more diverse, compared to the other two clades
(Sheppard et al. 2008). We selected clade 1 from C. coli by extracting all STs that are
members of ST-828 clonal complex and ST-1150 clonal complex (Sheppard et al. 2010).
There are 3654 STs for C. jejuni, and 606 distinct STs for C. coli clade1.

Methods Overview
Either mutation(s) or recombination(s) can generate SLVs. In this paper, mutation is defined
as a single nucleotide change (a point mutation), whereas recombination represents the
transfer of several adjacent nucleotides from one DNA source to another. An event is either
a mutation or a recombination. An SLV can be generated by one or more events; however,
recombination will tend to mask mutation. We model separately the mutation and
recombination process to derive a probability model for the number of nucleotide
differences between STs, under both the assumption that the SLV has been created solely by
mutation, and that it has not. This then enables us to estimate the proportion of SLVs that
have been caused solely by mutation, and also estimate the relative rate of recombination to
mutation. More details of the analysis are given in the Supplementary Material.
Europe PMC Funders Author Manuscripts

Modeling SLV Evolution


The data consists of, for each SLV, the locus at which the pair of STs differ, and the number
of nucleotide differences at that locus. From this, we aim to infer how likely it is that the
differences observed at this locus arise from point mutation only, as opposed to being
produced by recombination.

To do this, we first model the distribution of nucleotide differences we would expect at an


SLV at a given locus if these differences are solely due to mutation. This can be done by
first calculating the probability of an SLV given the number of point of mutations that have
occurred in one locus as the likelihood function, introducing a prior distribution for the
number of mutations to occur between two STs in that locus. The former probability is
based on the need for all mutation events to occur at the same locus. Under the coalescent
theory, a geometric distribution is chosen to use as the prior distribution (Hein et al. 2005).
Under Bayesian theory, we can obtain the required conditional distribution (Equation 1 in
Supplementary Material). The resulting conditional distribution of the number of nucleotide
differences is concentrated on small numbers of nucleotide differences, and is robust to the
choice of prior.

Second, the probability of observing h (h = 1, 2, 3…) nucleotide differences introduced by


recombination was estimated using Bayesian methods. It was calculated by sampling the
alleles based on their frequencies in the current database. Two (simplifying) assumptions for

J Mol Evol. Author manuscript; available in PMC 2014 April 14.


Yu et al. Page 4

the recombination model were made: (1) if recombination occurs between two alleles it
affects an entire locus rather than just part of a locus; and (2) we ignore the effect of any
additional mutation events. Under these assumptions, our model suggests that in most cases
recombination will introduce many more nucleotide differences than expected under the
mutation only model. Note that our results are robust to the assumption in (1) unless
recombination affects only small fragments of a locus, in these cases our assumption will
tend to lead to overestimates of the proportion of SLVs due to mutation only. Hence, it will
Europe PMC Funders Author Manuscripts

tend to underestimate the ratio of recombination to mutation.

Given these two models, we can then estimate the proportion of our SLVs at each locus that
are due to mutation only. In practice, we use an expectation-maximization (EM) algorithm
(Dempster et al. 1977) to infer this proportion. Finally, based on the estimated proportion of
SLVs at a given locus that is due to mutation only we estimate the probability that the single
event that led to the generation of a new allele was a mutation. The above analysis was
carried out by an R script (available by request from the first author).

To test the accuracy of our method for estimating the ratio of recombination to mutation,
MLST data were simulated under different known ratios of recombination to mutation with
different recombination tract lengths using SimMLST software (Didelot et al. 2009).

We used a parametric bootstrap to assess uncertainty in estimates. We simulated 100


datasets for both C. coli and C. jejuni. These datasets matched the true data in terms of
number of STs, relative rate of mutation to recombination, and overall mutation rate across
the seven gene loci. Within the simulations we assumed that the mutation rate and
recombination rate were the same across loci. For our simulated data we estimated the
probability of an event being a mutation, and calculated the variability of estimates of this
quantity across the simulations: both for estimates for a single locus, and for the estimate
obtained by averaging across loci. We consider estimates of this quantity as the variance of
the estimates changed little when we varied the true value of the relative rate of
recombination to mutation. Confidence intervals were then calculated using a normal
approximation, and transformed to confidence intervals for the relative rate of
recombination to mutation.
Europe PMC Funders Author Manuscripts

Results
SLV Analysis on the Campylobacter MLST Databases
From our downloaded dataset, there were 7417 SLVs (aspA: 992; glnA: 1045; gltA: 1250;
glyA: 773; pgm: 1580; tkt: 1060; and uncA 717) for C. jejuni, and 1842 SLVs (aspA: 110;
glnA: 179; gltA: 128; glyA: 292; pgm: 325; tkt: 647; and uncA: 161) for C. coli clade1. The
difference in the number of SLVs at each locus suggests that it is worthwhile estimating the
relative mutation and recombination rates separately for each locus.

The Distribution of Nucleotide Differences Between Each SLV for Each Locus
Each SLV relates to one pair of STs, and the plots (Figs. 1, 2) show the nucleotide
differences that occurred within those pairs of STs at each MLST locus for C. jejuni and C.
coli clade 1. These plots show that SLVs with a large number of nucleotide differences
(>45) occurred in every locus. The pairs of STs with a large number of nucleotide
differences (50–80) are almost certainly due to recombination, as it is highly unlikely that
more than 50 independent point mutations would occur at a single locus while the other six
loci remained the same. These large differences are likely to be due to recombination
between C. jejuni and C. coli (Sheppard et al. 2008; Wilson et al.2009). Species were
designated according to the PubMLST data, and only those SLVs that comprised STs that

J Mol Evol. Author manuscript; available in PMC 2014 April 14.


Yu et al. Page 5

were assigned 100 % C. jejuni or C. coli were plotted. Even with this strict species
designation, there were still large nucleotide differences visible between SLVs within
species. There were second peaks in the range of 15-20 differences at the loci glyA, pgm,
and tkt for both C. jejuni and C. coli clade 1. These peaks are likely to be due to
recombination as well. The first peak of most loci (except for pgm for C. jejuni and tkt for C.
coli clade 1) represented approximately 100-200 SLVs for C. jejuni and around 100 SLVs
for C. coli clade 1 with only one nucleotide difference; most of these are more likely to be
Europe PMC Funders Author Manuscripts

due to mutation.

Relative Contributions of Recombination and Mutation Separately for C. jejuni and C. coli
Clade 1
Tables 1 and 2 demonstrate that recombination contributed more to the generation of SLVs
than did mutation for both the groups (C. jejuni and C. coli clade 1), but the range of
estimates vary for the two groups. The average ratio of recombination events to mutation
events from the seven loci is 6.96 (95 % CI 6.08, 8.09) for C. jejuni (Table 1), and 1.01 (95
% CI 0.78, 1.30) for C. coli clade 1 (Table 2).

For each locus, we also estimated the proportion of nucleotide differences introduced by
recombination as opposed to mutation, and this ranged from 97 % (gltA and glyA) to 99 %
(aspA, tkt, and uncA) for C. jejuni, and from 60 % (glnA) to 98 % (aspA) for C. coli clade 1.

We also investigated the robustness of the mutation model to different prior distributions of
the probability of events caused by mutations only. These suggest that the results in
Supplementary Table 1 and 2 are conservative regarding the importance of recombination in
producing new variation for C. jejuni and C. coli clade 1.

We see evidence for differences in the relative role of recombination to mutation across the
genes (Tables 1, 2). In particular, the parametric bootstrap results show that there is evidence
for a lower rate of recombination in glnA for C. coli and for glyA in C. jejuni, and for a
higher rate in aspA in C. coli. To assess the strength of this evidence, we looked at the
lowest (and the highest) estimated value of the relative rate of recombination to mutation
Europe PMC Funders Author Manuscripts

across the seven genes in our simulated data divided by the average of estimated rate across
the seven genes. For both C. coli and C. jejuni, we never observed an estimate as low as that
for glnA and glyA, respectively, across the 100 simulations in each case (the lowest
estimates were 0.36 and 0.62 for C. coli and C. jejuni, respectively, compared to observed
values of 0.23 and 0.43) or as high as aspA for C. coli (highest estimate was 2.04, compared
to an observed value of 2.21).

Discussion
We have analyzed SLVs to infer the relative importance of recombination and mutation to
generate differences between closely related C. jejuni and C. coli clade 1 isolates. The
higher average estimates for C. jejuni compared to C. coli demonstrates higher
recombination in C. jejuni, compared to C. coli. This is consistent with the existing
population structure (three clades) of C. coli, but not with apparent subclade structure in C.
jejuni (Sheppard et al.2008). We estimate that recombination contributes between 2.97 and
8.91 times more than mutation to events that generate new alleles for C. jejuni, depending
on the MLST locus, and between 0.23 and 2.23 for C. coli clade 1. The variations between
housekeeping genes within species also show the different evolution pressure on different
genes. For C. jejuni, glyA has less recombination contribution, compared to the other six
genes. For C. coli, glnA has less recombination contribution, compared to the other six
genes.

J Mol Evol. Author manuscript; available in PMC 2014 April 14.


Yu et al. Page 6

Our analysis has similarities to that of Schouls et al. (2003), who used the approach
described by Feil et al. (2000) to estimate the relative rate of recombination and mutation for
C. jejuni. The original idea of Feil et al.’s method (2000) is put forward by Guttman and
Dykhuizen (1994). However, their method overestimates the ratio of recombination to
mutation, compared to ours. They also analyzed SLVs, though restricted to pairs of SLVs
within the same clonal complex. Furthermore, rather than the model-based approach we
consider, they used a simple rule to classify which SLVs had been caused by mutation as
Europe PMC Funders Author Manuscripts

opposed to recombination. The rule was that if a pair of SLVs varies by a single nucleotide
difference and one of the MLST alleles at the locus was unique, it is due to a mutation,
whereas all other pairs of SLVs are caused by recombination. This means that, under this
algorithm, SLVs that differ by two nucleotide differences could not have arisen by two
independent mutation events, and recombination events that mask mutation events are not
considered. Both assumptions may lead to an underestimate of mutation. The analysis of
Schouls et al. (2003) estimates that recombination is approximately eight times more likely
to change an allele than mutation. This is larger than our estimate, which is likely to be due
to these biases in the method used by Schouls et al. (2003). According to Feil et al.’s method
(2000), Schouls et al. (2003) estimated a recombination size of about 3.3 kb. We
implemented a simplified version of Feil et al.’s method (2000) (details in the
Supplementary Material), and the results show that under the 3 kb recombination size, the
ratio is overestimated.

Our estimates suggest a more important role for recombination in producing new diversity
into C. jejuni than more recent studies which have analyzed samples of C. jejuni isolates
from different source populations. Fearnhead et al. (2005) estimate that recombination rates
are, if anything, less than the mutation rates. While Vos and Didelot (2008), and Wilson et
al. (2009) give estimates of the proportion of nucleotide differences introduced by
recombination as opposed to mutation which are much smaller than the ones we obtained.
Both the studies concluded that the number of nucleotide differences introduced by
recombination are only approximately twice as many as those introduced by point mutation:
2.2 for Vos and Didelot (2008) and 2.67 (95 % CI 1.39, 4.95) for Wilson et al. (2009).
Europe PMC Funders Author Manuscripts

The difference between our study and these is that we analyze only SLVs, which means we
are looking at closely related STs for which there has been less time for selection to act.
Intuitively, selection is likely to be the strongest against recombination events that introduce
large differences, although it is possible that some recombination events may introduce a
section of DNA from an organism that is highly adapted and “successful” in the given
environment. Therefore, although we estimate that recombination is introducing more
differences than previously thought in our closely related and recently evolved STs, many of
these differences may be subsequently purged from the population due to weak purifying
selection. This is consistent with the effects of purifying selection described in Wilson et
al.’s paper (2009).

Whole genome analysis may provide a greater insight into the genome-wide evolution of
Campylobacter and provide further explanations for the apparent differences between
previous estimates of recombination and mutation. Recently, Biggs et al. (2011) analyzed
the genomes of two closely related Campylobacter ST-474 isolates that also had identical
flaA SVR regions and compared them to available C. jejuni reference strains. They
estimated that around 97 % of the nucleotide differences between these two closely related
isolates were caused by recombination. This estimate is similar to ours, and suggests that the
importance of recombination for driving changes in C. jejuni is not just confined to the
MLST housekeeping genes we have studied.

J Mol Evol. Author manuscript; available in PMC 2014 April 14.


Yu et al. Page 7

The aim of this study was to increase our understanding of the evolution of C. jejuni and C.
coli by investigating the generation of SLVs. The availability of the large database of C.
jejuni and C. coli isolates provides a good opportunity to investigate the evolution of C.
jejuni and C. coli using SLVs. Using seven independent housekeeping loci we used the
method proposed in this paper to estimate that recombination contributes roughly seven
times as much as mutation to the generation of SLVs for C. jejuni, and equally for C. coli,
which provides further evidence that recombination plays a more important role in the
Europe PMC Funders Author Manuscripts

evolution of C. jejuni and C. coli than mutation.

Our results also point to important differences in terms of the forces driving evolution for C.
jejuni and C. coli; and suggest that the relative role of recombination to mutation may differ
between genes, and these differences themselves may be different for C. jejuni and C. coli.
Understanding what is causing these differences will be important for fully understanding
how these bacteria may evolve in the future. However, the fact that we observed differences
in recombination between C. jejuni and C. coli is consistent with the introgression
hypothesis of Sheppard et al.’s paper (2011), which implies that patterns of genetic
exchange have changed over time. The research on SLVs described in this paper could be
extended either by considering more genes, such as flagellin genes (flaA and flaB)
(Meinersmann and Hiett 2000), and porA, the gene encoding the major outer membrane
proteins (MOMPs) (Zhang et al. 2000; Clark et al. 2005), or by considering other species of
Campylobacter.

Supplementary Material
Refer to Web version on PubMed Central for supplementary material.

Acknowledgments
The authors acknowledge the Marsden Fund project 08-MAU-099 (Cows, starlings and Campylobacter in New
Zealand: unifying phylogeny, genealogy, and epidemiology to gain insight into pathogen evolution) for funding this
project. This publication made use of the Campylobacter Multi Locus Sequence Typing website (http://
pubmlst.org/campylobacter/) developed by Keith Jolley and sited at the University of Oxford (Jolley and Maiden
Europe PMC Funders Author Manuscripts

2010, BMC Bioinformatics, 11:595). The development of this site has been funded by the Wellcome Trust. BRH
acknowledges the Australian Research Council (Grant FT100100031).

References
Biggs PJ, Fearnhead P, Hotter G, Mohan V, Collins-Emerson J, Kwan E, Besser TE, Cookson A,
Carter PE, French NP. Whole-genome comparison of two Campylobacter jejuni isolates of the same
sequence type reveals multiple loci of different ancestral lineage. PLoS One. 2011; 6(11):e27121.
[PubMed: 22096527]
Clark CG, Bryden L, Cuff WR, Johnson PL, Jamieson F, Ciebin B, Wang G. Use ofthe Oxford
multilocus sequence typing protocol and sequencing of the flagellin short variable region to
characterize isolates from a large outbreak of waterborne Campylobacter sp. strains in Walkerton,
Ontario, Canada. J Clin Microbiol. 2005; 43:2080. [PubMed: 15872226]
Dempster AP, Laird NM, Rubin DB. Maximum likelihood from incomplete data via the EM
algorithm. J Roy Statist Soc Ser B. 1977; 39(1):1–38.
Didelot X, Lawson D, Falush D. Simmlst: simulation of multilocus sequence typing data under a
neutral model. Bioinformatics. 2009; 25:1442. [PubMed: 19286834]
Dingle KE, Colles FM, Wareing DRA, Ure R, Fox AJ, Bolton FE, Bootsma HJ, Willems RJL, Urwin
R, Maiden MCJ. Multilocus sequence typing system for Campylobacter jejuni. J Clin Microbiol.
2001; 39:14. [PubMed: 11136741]
Fearnhead P, Smith NGC, Barrigas M, Fox A, French N. Analysis of recombination in Campylobacter
jejuni from MLST population data. J Mol Evol. 2005; 61:333–340. [PubMed: 16044246]

J Mol Evol. Author manuscript; available in PMC 2014 April 14.


Yu et al. Page 8

Feil EJ, Maiden MC, Achtman M, Spratt BG. The relative contributions of recombination and
mutation to the divergence of clones of Neisseria meningitidis. Mol Biol Evol. 1999; 16:1496.
[PubMed: 10555280]
Feil EJ, Smith JM, Enright MC, Spratt BG. Estimating recombinational parameters in Streptococcus
pneumoniae from multilocus sequence typing data. Genetics. 2000; 154:1439. [PubMed: 10747043]
Feil EJ, Holmes EC, Bessen DE, Chan MS, Day NPJ, Enright MC, Goldstein R, Hood DW, Kalia A,
Moore CE, et al. Recombination within natural populations of pathogenic bacteria: short-term
Europe PMC Funders Author Manuscripts

empirical estimates and long-term phylogenetic consequences. Proc Natl Acad Sci. 2001; 98:182.
[PubMed: 11136255]
Feil EJ, Li BC, Aanensen DM, Hanage WP, Spratt BG. eBURST: inferring patterns of evolutionary
descent among clusters of related bacterial genotypes from multilocus sequence typing data. J
Bacteriol. 2004; 186:1518. [PubMed: 14973027]
Guttman DS, Dykhuizen DE. Clonal divergence in Escherichia coli as a result of recombination, not
mutation. Science. 1994; 266:1380–1383. [PubMed: 7973728]
Hein, J.; Schierup, MH.; Wiuf, C. Gene genealogies, variation and evolution: a primer in coalescent
theory. Oxford University Press; Oxford: 2005.
Humphrey T, O’Brien S, Madsen M. Campylobacters as zoonotic pathogens: a food production
perspective. Int J Food Microbiol. 2007; 117:237–257. [PubMed: 17368847]
Jolley KA, Maiden MCJ. BIGSdb: scalable analysis of bacterial genome variationat thepopulation
level. BMC Bioinformatics. 2010; 11:595. [PubMed: 21143983]
Konkel ME, Gray SA, Kim BJ, Garvis SG, Yoon J. Identification of the enteropathogens
Campylobacter jejuni and Campylobacter coli based on the cadF virulence gene and its product. J
Clin Microbiol. 1999; 37:510. [PubMed: 9986804]
Maiden MCJ, Bygraves JA, Feil EJ, Morelli G, Russell JE, Urwin R, Zhang Q, Zhou J, Zurth K,
Caugant DA. Multilocus sequence typing: a portable approach to the identification of clones
within populations of pathogenic microorganisms. Proc Nat Acad Sci USA. 1998; 95:3140.
[PubMed: 9501229]
Meinersmann RJ, Hiett KL. Concerted evolution of duplicate fla genes in Campylobacter.
Microbiology. 2000; 146:2283. [PubMed: 10974116]
Richman AD, Herrera LG, Nash D, Schierup MH. Relative roles of mutation and recombination in
generating allelic polymorphism at an MHC class II locus in Peromyscus maniculatus. Genet Res.
2003; 82:89–99. [PubMed: 14768893]
Europe PMC Funders Author Manuscripts

Sarkar SF, Guttman DS. Evolution of the core genome of Pseudomonas syringae, a highly clonal,
endemic plant pathogen. Appl Environ Microbiol. 2004; 70:1999. [PubMed: 15066790]
Schouls LM, Reulen S, Duim B, Wagenaar JA, Willems RJL, Dingle KE, Colles FM, Van Embden
JDA. Comparative genotyping of Campylobacter jejuni by amplified fragment length
polymorphism, multilocus sequence typing, and short repeat sequencing: strain diversity, host
range, and recombination. J Clin Microbiol. 2003; 41:15. [PubMed: 12517820]
Sheppard SK, McCarthy ND, Falush D, Maiden MCJ. Convergence of Campylobacter species:
implications for bacterial evolution. Science. 2008; 320:237–239. [PubMed: 18403712]
Sheppard SK, Dallas JF, Wilson DJ, Strachan NJC, McCarthy ND, et al. Evolution of an agriculture-
associated disease causing Campylobacter coli clade: evidence from National Surveillance Data in
Scotland. PLoS ONE. 2010; 5(12)
Sheppard SK, McCarthy ND, Jolley KA, Maiden MCJ. Introgression in the genus Campylobacter:
generation and spread of mosaic alleles. Microbiology. 2011; 157:1066–1074. [PubMed:
21212120]
Suerbaum S, Lohrengel M, Sonnevend A, Ruberg F, Kist M. Allelic diversity and recombination in
Campylobacter jejuni. J Bacteriol. 2001; 183:2553. [PubMed: 11274115]
Tauxe, RV.; Nachamkin, I.; Blaser, MJ.; Tompkins, LS. Epidemiology of Campylobacter jejuni
infections in the United States and other industrialized nations. MBio; Washington, DC: 1992.
Vos M, Didelot X. A comparison of homologous recombination rates in bacteria and archaea. ISME J.
2008; 3:199–208. [PubMed: 18830278]

J Mol Evol. Author manuscript; available in PMC 2014 April 14.


Yu et al. Page 9

Wilson DJ, Gabriel E, Leatherbarrow AJH, Cheesbrough J, Gee S, Bolton E, Fox A, Hart CA, Diggle
PJ, Fearnhead P. Rapid evolution and the importance of recombination to the gastroenteric
pathogen Campylobacter jejuni. Mol Biol Evol. 2009; 26:385. [PubMed: 19008526]
Zhang Q, Meitzler JC, Huang S, Morishita T. Sequence polymorphism, predicted secondary structures,
and surface-exposed conformational epitopes of Campylobacter major outer membrane protein.
Infect Immun. 2000; 68:5679. [PubMed: 10992471]
Europe PMC Funders Author Manuscripts
Europe PMC Funders Author Manuscripts

J Mol Evol. Author manuscript; available in PMC 2014 April 14.


Yu et al. Page 10
Europe PMC Funders Author Manuscripts
Europe PMC Funders Author Manuscripts

Fig. 1.
SLVs of PubMLST data. The x axes represent the number of nucleotide differences between
STs that make up an SLV; y axes represent the number of recorded events. A represents the
nucleotide differences for SLVs in the PubMLST database for C. jejuni; others are the
nucleotide differences for SLVs by loci

J Mol Evol. Author manuscript; available in PMC 2014 April 14.


Yu et al. Page 11
Europe PMC Funders Author Manuscripts
Europe PMC Funders Author Manuscripts

Fig. 2.
SLVs of PubMLST data. The x axes represent the number of nucleotide differences between
STs that make up an SLV; y axes represent the number of recorded events. A represents the
nucleotide differences for SLVs in the PubMLST database for C. coli clade 1; others are the
nucleotide differences for SLVs by loci

J Mol Evol. Author manuscript; available in PMC 2014 April 14.


Yu et al. Page 12

Table 1
Allele lengths for each locus; estimates for C. jejuni for each housekeeping locus of the probability of an SLV
being caused by mutation only (p); the expected number of mutations for an SLV; the relative rate of
recombination to mutation; 95 % CI for the estimated relative rate of recombination to mutation; and the % of
Europe PMC Funders Author Manuscripts

nucleotide differences of an SLV that were introduced by recombination

Locus Allele lengths (bp) p Expected number mut. Relative rate of rec 95 % CI % Diff due to rec
aspA 477 0.09 0.11 8.91 (5.98, 16.06) 99
glnA 477 0.09 0.11 8.86 (5.96, 15.91) 98
gltA 402 0.10 0.12 7.84 (5.43, 13.12) 97
glyA 507 0.21 0.32 2.97 (2.40, 3.77) 97
pgm 498 0.10 0.15 7.14 (5.06, 11.41) 98
tkt 459 0.11 0.15 6.81 (4.87, 10.66) 99
uncA 489 0.12 0.16 6.21 (4.53, 9.37) 99
Average 472.71 0.12 0.16 6.96 (6.08, 8.09) 98
Europe PMC Funders Author Manuscripts

J Mol Evol. Author manuscript; available in PMC 2014 April 14.


Yu et al. Page 13

Table 2
Allele lengths for each locus; estimates for C. coli clade 1 for each housekeeping locus of the probability of an
SLV being caused by mutation only (p); the expected number of mutations for an SLV; the relative rate of
recombination to mutation; 95 % CI for the estimated relative rate of recombination to mutation; and the % of
Europe PMC Funders Author Manuscripts

nucleotide differences of an SLV that were introduced by recombination

Locus Allele lengths (bp) p Expected number mut. Relative rate of rec 95 % CI % Diff due to rec
aspA 477 0.18 0.65 2.23 (1.13, 5.69) 98
glnA 477 0.80 0.84 0.23 (0.03, 0.53) 60
gltA 402 0.56 0.61 0.72 (0.35, 1.37) 80
glyA 507 0.63 0.72 0.54 (0.24, 1.04) 86
pgm 498 0.47 0.54 1.04 (0.54, 2.03) 90
tkt 459 0.51 0.62 0.85 (0.43, 1.63) 91
uncA 489 0.39 0.44 1.44 (0.75, 3.01) 89
Average 472.71 0.51 0.63 1.01 (0.78, 1.30) 85
Europe PMC Funders Author Manuscripts

J Mol Evol. Author manuscript; available in PMC 2014 April 14.

You might also like