You are on page 1of 11

REVIEWS

Bacterial nucleoid-associated
proteins, nucleoid structure and
gene expression
Shane C. Dillon and Charles J. Dorman
Abstract | Emerging models of the bacterial nucleoid show that nucleoid-associated
proteins (NAPs) and transcription contribute in combination to the dynamic nature of
nucleoid structure. NAPs and other DNA-binding proteins that display gene-silencing and
anti-silencing activities are emerging as key antagonistic regulators of nucleoid structure.
Furthermore, it is becoming clear that the boundary between NAPs and conventional
transcriptional regulators is quite blurred and that NAPs facilitate the evolution of novel
gene regulatory circuits. Here, NAP biology is considered from the standpoints of both gene
regulation and nucleoid structure.

Stable RNA genes


Despite differences in the cellular organization of a positive or negative manner. An appreciation of the
Genes that code for ribosomal Eukarya, Bacteria and Archaea, it is important to remem- higher-order superstructures that link gene expression
RNA (rRNA) and tRNA. ber that all three domains rely on DNA to store and rep- and the architecture of the bacterial genome requires
Compared with mRNA, rRNA licate genetic information. In each domain the genetic an understanding of the basic organization of the
and tRNA are chemically very
material has to be organized for storage in ways that are nucleoid (BOX 1).
stable.
compatible with DNA replication, chromosome segrega-
tion and gene expression. Proteins that alter the shape Transcription and the nucleoid structure
of the DNA to make it more compact and that have the Transcription and looped domain boundaries.
potential to influence transcription have been identified Experimental data show that there is a good correlation
in all three kingdoms of life. In eukaryotes these pro- between transcriptional activity and the number and sta-
teins are known as histones, and their ability to influence bility of looped DNA domains in a particular region3,4,5.
chromatin structure and transcription is understood in Transcriptional activity varies with the physiological
considerable detail. Initially, this depth of understand- state of the cell. Fast-growing bacteria have more activity,
ing influenced our appreciation of those proteins that especially at genes coding for ribosome components and
carry out analogous tasks in bacteria, leading them to other parts of the translation machinery, and have more
be referred to as ‘histone-like’ proteins1,2. However, this looped DNA domains. When bacterial growth slows, as
term is becoming less appropriate as the distinct nature happens during starvation or at the onset of the station-
of these bacterial proteins is revealed. They are now ary phase of growth in batch culture, there is less tran-
collectively referred to as nucleoid-associated proteins scriptional activity and a corresponding relaxation in the
(NAPs), which more accurately reflects their cellular looped domain structure of the nucleoid (FIG. 1).
location without implying that they have any structural Among the most active promoters in Escherichia coli
similarity to histones. Members of the NAP group are are those of the stable RNA genes, in particular genes
Department of Microbiology, numerous, diverse and unevenly understood, and the coding for ribosomal RNA, which are encoded by seven
School of Genetics and
current list is probably not exhaustive (TABLE 1). Archaeal rRNA operons6. Expression of rRNA genes is controlled
Microbiology, Moyne
Institute of Preventive NAPs are less widely studied, although several have been by the stringent response, which is a mechanism that
Medicine, Trinity College, investigated in detail (TABLE 1). stops the production of components of the translational
Dublin 2, Ireland. Most NAPs possess DNA-binding activity and an abil- machinery under restrictive growth conditions. During
Correspondence to C.J.D. ity to alter the trajectory of the DNA molecule (that is, the the stringent response, the alarmone guanosine tetra-
e‑mail: cjdorman@tcd.ie
doi:10.1038/nrmicro2261
direction taken by the DNA through three-dimensional phosphate (ppGpp) and the polymerase-interacting
Published online space) by bending, wrapping or bridging it. In addition, DnaK suppressor protein (DksA) facilitate a targeted
8 February 2010 many NAPs are able to influence transcription in either weakening of RNA polymerase activity at stringently

NATuRE REvIEWs | Microbiology voluME 8 | MARcH 2010 | 185

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Table 1 | Nucleoid-associated proteins of eukaryotes, archaea and bacteria


Protein or group DNA DNA DNA binding motif Molecular Native protomer refs
of proteins wrapping bridging bending mass
Eukaryotes
Core histones, Yes ND ND A ~10 bp periodic oscillation of AA/TT/TA 11–14 kDa Homodimer 115
H2A, H2B, H3 elements in-phase with each other and
and H4 out-of-phase with ~10 bp periodic GCs
Linker histones, ND Yes ND AT-rich DNA ~21 kDa Homodimer 116
H1 and H5
Smc ND Yes ND AT-rich DNA able to form secondary ~140 kDa Heterodimer (for example, 117
structures SMC1–SMC3)
Hmg ND ND Yes AT-tract sites 11–38 kDa Homodimer or heterodimer 118
(for example, HMG1–HMG2)
Euryarchaeota
Archaeal histones Yes ND ND (A/T)3NN(G/C)3NN ~7.5 kDa Homodimer or heterodimer 119
HMfA and HMfB
Lrp Yes Yes ND ND 15–22 kDa Homodimer 120
Alba ND Yes ND ND ~10 kDa Homotetramer 20
MC1 ND ND Yes AT-rich DNA ~10 kDa Homodimer 121
HU ND ND Yes ND ~10 kDa Homodimer 122
SMC ND Yes ND ND ~135 kDa Homodimer 123
Crenarchaeota
Lrp Yes Yes ND ND ~18 kDa Homodimer 124
Cren7 ND ND Yes ND ~7 kDa Monomer 125
Sul7d ND ND Yes ND ~7 kDa Monomer 126
Alba ND Yes ND ND ~10 kDa Homodimer or 127
homotetramer
SMC ND Yes ND ND ~100 kDa Homodimer 128
CC1 ND ND ND ND ~6 kDa Monomer 129
Gram-negative bacteria
HU Yes ND Yes A DNA structural motif in dsDNA joined ~9 kDa Heterodimer (for example, 42,
to either dsDNA or ssDNA, with a mild HUα–HUβ) 130
preference for AT-rich or curved DNA
Lrp Yes Yes ND (T/C)AG(A/T/C)A(A/T)ATT(A/T)T(A/T/G) ~18 kDa Homodimer 69
CT(A/G)
MukB ND Yes ND ND ~175 kDa Homodimer 57
Fis Yes Yes Yes A6 tracts and AT tracts ~11 kDa Homodimer 77,79
(helically
phased
sites)
H-NS ND Yes ND AT-rich DNA and TCGATAAATT ~15 kDa Homodimer or heterodimer 19
(H-NS–StpA)
IHF ND ND Yes (A/T)ATCAANNNNTT(A/G) ~11 kDa Heterodimer (IHFα– IHFβ) 44,45
Dps ND ND ND ND ~19 kDa Monomer or dodecamer 94
StpA ND Yes ND AT-rich DNA ~15 kDa Homodimer or heterodimer 29
(StpA–H-NS)
CbpA ND ND ND Curved DNA ~33 kDa Homodimer or heterodimer 92
(CbpA–CbpM)
CbpB ND ND ND Curved DNA ~33 kDa Monomer 102
EbfC ND Suggested ND GTNAC ~11 kDa Homodimer 93
MvaT ND Yes ND AT-rich DNA Homodimer 26
Gram-positive bacteria
MukB ND Yes ND Preference for ssDNA ~130 kDa Homodimer 131
Lrp Yes Yes ND ND ~17 kDa Homodimer 72
HU ND ND Yes ND ~10 kDa Homodimer 37
Lsr2 ND Yes ND AT-rich DNA ~12 kDa Homodimer 24
Hlp ND ND ND ND ~21 kDa Monomer 38
MrgA ND ND ND ND ~17 kDa Monomer or dodecamer 132
Alba, acetylation lowers binding affinity; CbpA, curved-DNA-binding protein A; CbpB, curved-DNA-binding protein B (also known as Rob); CbpM, chaperone
modulatory protein; Dps, DNA protection from starvation; dsDNA, double-stranded DNA; Fis, factor for inversion stimulation; Hlp, histone-like protein; Hmg,
high mobility group; H-NS, histone-like nucleoid-structuring; IHF, integration host factor; Lrp, leucine-responsive regulatory protein; MrgA, metalloregulation
DNA-binding stress protein; ND, not determined; Smc, structural maintenance of chromosome; ssDNA, single-stranded DNA.

186 | MARcH 2010 | voluME 8 www.nature.com/reviews/micro

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 1 | Organization of the bacterial nucleoid transcription factories (FIG. 1). However, bacteria depleted
of NAPs, including H-Ns and Fis, can maintain their
Escherichia coli and its close relatives continue to provide the paradigm for our nucleoid structure18, which contradicts the proposed
understanding of chromosome storage in the nucleoid. The chromosome exists structural role for NAPs. Although this discrepancy may
in a space that is subject to molecular crowding, where entropic forces can influence
reflect the limitations in the tools available for nucleoid
the path taken by the DNA. The DNA polymer adopts a self-avoiding random walk
analysis at the required scale or a divergence between the
characterized by a thermal persistence length (the distance traversed by the molecule
before it randomly takes a turn) of about 150 bp, equivalent to a distance of 50 nm. experimental and physiological conditions, it may also
This produces a cloud of randomly coiled DNA with a diameter of around 10 μm. suggest that additional factors could be important for
Subdivision of the chromosome into around 400 independent loops of ~10 kb provides the maintenance of nucleoid structure, perhaps acting in
further compaction, and negative supercoiling of the loops further reduces the concert with NAPs to form domain boundaries.
diameter of the folded chromosome110.
Negative supercoiling of DNA is mainly a product of the activity of topoisomerases, Bridge builders. When considering the potential for
but it is also influenced at a local level by the activities of DNA polymerase and RNA NAPs to engage in both nucleoid structuring and gene
polymerase. Nucleoid-associated proteins and other DNA-binding proteins can regulation, it is useful to examine their physical impact
constrain supercoils, altering the level of effective DNA supercoiling in the
on the DNA to which they bind. The importance of the
chromosome111. This decreases the superhelical density of the DNA in E. coli from a
DNA-bridging activity that is displayed by proteins such
value of –0.05 for naked DNA to –0.025 for the protein-bound molecule. This is of
considerable importance, because in many cases transcriptional promoter activity is as H-Ns in enteric bacteria and acetylation lowers bind-
sensitive to the local effective level of supercoiling and the activity of promoters can ing affinity protein (Alba) in archaea has been reported
be increased or decreased in response to superhelical variation112. in several studies19,20,21. In these studies, the authors used
single-molecule techniques such as optical tweezers
to study DNA–protein–DNA bridges, allowing these
controlled promoters 7,8. Experiments using RNA bridges to be assessed for their potential to influence
polymerase tagged with GFP show the presence of so- processes such as transcription initiation and nucleoid
called ‘transcription factories’ (or ‘transcription foci’) in architecture19. In vitro, bridge formation by H-Ns is sen-
bacteria undergoing rapid growth (FIG. 1). These struc- sitive to Mg 2+, such that the protein oligomerizes along
tures become dispersed in bacteria in which the stringent linear duplex DNA in the absence of Mg 2+ and forms
response is active but not in mutants deficient in RelA9, DNA–protein–DNA bridges on the same DNA molecules
a key regulator of this response7. Each cell possesses a in the presence of 10 mM Mgcl2 (REF. 133). Bridge forma-
small number of these factories (on average, three)9,10. tion lends itself to the control of transcription, because
However, given the observed correlation between high the bridges have the potential to trap RNA polymerase at
transcription levels and domain maintenance, most of or exclude it from promoters, thereby silencing the genes
the genes along the chromosome are expressed too infre- concerned. Furthermore, evidence from a range of stud-
quently or at too low a level to influence the domain ies has shown that H-Ns and RNA polymerase colocalize
structure of the nucleoid. Therefore, one must consider at bacterial promoters, consistent with a trapping mecha-
other elements with the potential to have an impact on nism13,22. It is thought that DNA–protein–DNA bridges
nucleoid architecture. are repressive for transcription and must be disrupted
for gene expression to commence. It is becoming clear
NAPs and looped domains. some NAPs have proper- that bridging is opposed by a wide range of mechanisms,
ties that could allow them to act as boundary elements each of which is based on a type of interaction with DNA
(that is, the structures that mark the beginning and that renders the bridges untenable21,23. Examples of such
end of chromosomal domain loops), and there is evi- anti-bridging activity include DNA bending and DNA
dence from genetics and bioinformatics studies that wrapping, which are activities that are associated with
Superhelical density some NAPs do indeed serve this function11,12. NAPs most DNA-binding proteins (FIG. 2).
The average number of with DNA-bridging activity are especially attractive
superhelical turns per helical
candidates. chromatin immunoprecipitation-on- Dual-purpose NAPs
turn of the DNA, usually
abbreviated as σ. chip (chIP-on-chip) studies have shown that the NAP The number of bacterial proteins in the NAP class is
histone-like nucleoid-structuring protein (H-Ns) gradually increasing, and their classification is discussed
Transcription factories binds AT-rich DNA throughout the genomes of below (TABLE 1). NAPs contribute to both nucleoid struc-
Foci in the nucleoid where E. coli 13,14 and Salmonella enterica15,16. The distribution ture and gene regulation, and they may perform both
highly transcribed promoters,
such as those of the ribosomal
of these binding sites is consistent with the likely loca- roles simultaneously.
RNA genes, coalesce during tions of domain loop boundaries12. Factor for inversion
periods of high transcriptional stimulation (Fis), another NAP, also binds throughout H-NS: genome guardian and universal repressor. Proteins
activity. the genome and, like H-Ns, is especially abundant at with an amino acid sequence related to that of H-Ns
intergenic sites 13. It has also been reported to form are widely found in bacteria and have been studied in
DNA bridging
Pertaining to a protein that DNA–protein–DNA bridges17. The expression pattern pathogens such as Mycobacterium tuberculosis 24, Vibrio
binds to two distinct DNA of Fis is in keeping with the dynamic nature of looped cholerae25 and Pseudomonas aeruginosa26. A protein capa-
molecules or different domain boundaries, in that Fis is most abundant in the ble of complementing an H-Ns deficiency has also been
portions of the same DNA early exponential phase of growth, when the looped isolated in mice27. Perhaps it is conservation of function
molecule simultaneously
through two DNA-binding
domains are most plentiful. Both Fis and H-Ns target rather than amino acid sequence that is crucial, and an
domains, producing a promoters of the rRNA genes, perhaps indicating that ability to form DNA–protein–DNA bridges may emerge
DNA–protein–DNA bridge. their bridging activities support superstructures such as as the common feature of these different proteins.

NATuRE REvIEWs | Microbiology voluME 8 | MARcH 2010 | 187

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

a Exponential phase of growth b Stationary phase of growth

RNA polymerase Transcription


H–NS Fis
at RNA promoters factories

Figure 1 | Nucleoid-associated proteins and DNA supercoiling influence nucleoid structure. a | The folded
chromosome is organized into looped domains that are negatively supercoiled during the exponential phase of growth.
In this phase, the abundant nucleoid-associated proteins histone-like nucleoid-structuring protein (H-NS) and factor for
Nature Reviews | Microbiology
inversion stimulation (Fis) bind throughout the nucleoid and are associated with the seven ribosomal RNA operons. As
shown here in two cases, these are organized into superstructures called transcription factories. b | In stationary phase the
rRNA operons are quiescent and Fis is almost undetectable. The chromosome has fewer looped domains, and those that
are visible consist of relaxed DNA.

Both H-Ns and its paralogue stpA can constrain HU: regulator of DNA flexibility. The Hu protein has
supercoils in DNA28,29, and data from atomic-force micro- two subunits, Huα and Huβ, that are 70% identical in
scopy indicate that H-Ns reinforces duplex interwinding amino acid sequence. In E. coli, Hu exists in homodimer
in plectonemically supercoiled DNA30. Negative supercoiling or heterodimer forms, depending on the stage of growth
of the DNA is likely to facilitate the formation of DNA– of the bacterium36. The equivalent protein in species
H-Ns–DNA bridges, accounting for the coincidence of such as Geobacillus stearothermophilus exists only as a
DNA-supercoiling sensitivity and H-Ns binding at many homodimer 37. Perhaps this reflects the differences in the
bacterial promoters31. Bridging allows H-Ns to influence ecological circumstances of the bacteria, as E. coli and its
both nucleoid structure and gene expression simultane- close relatives need to adapt in a wider environmental
ously. stpA has DNA-binding activity but is best charac- range. The equivalent protein in Mycobacterium spp.,
terized as an RNA-binding protein and RNA chaperone32. histone-like protein (Hlp), consists of two domains, with
Because these proteins have similar oligomerization the amino-terminal portion of the protein resembling
domains (FIG. 3), stpA can interact with H-Ns to form Hu from E. coli and the carboxy-terminal portion being
heteromers, as can other paralogues, such as the plasmid- similar to the eukaryotic histone H1 (REF. 38). This is a
encoded sfh33. It has been suggested that the homomers rare example of a bacterial NAP that is truly histone-like
and heteromers exert distinct effects on transcription, in its amino acid sequence.
although supporting data are limited. However, data Hu–DNA interactions seem to be nonspecific, but the
from transcriptomics studies show that changes in the protein has a preference for binding to distorted regions
populations of these multimeric forms of NAPs result in of the DNA, such as bends or four-way junctions39. This
new gene expression patterns34. The activity of H-Ns is is consistent with the roles of Hu in recombination
modulated through its interaction with members of the and DNA topology management 40. Hu influences the
haemolysin expression-modulating (Hha)/YdgT family expression of a wide range of genes in E. coli, including
of proteins. These molecules lack DNA-binding activ- those involved in central metabolism and respiration41.
Plectonemically ity of their own and are thought to bind to the oligomeri- It interacts with topoisomerase I, leading to alterations
supercoiled DNA zation region in the amino-terminal domain of H-Ns, in the superhelicity of DNA40, which has implications
DNA with a braided or influencing the genes that H-Ns regulates by concentrat- for both nucleoid structure and gene expression. X-ray
interwound appearance ing its effects on horizontally acquired genes, including crystallography of Hu has revealed that the protein
caused by the addition or
removal of helical turns in
pathogenicity genes with AT-rich DNA35 (FIG. 3). How can form multimers consisting of octameric units. In
topologically closed the Hha/YdgT proteins influence H-Ns interactions with one arrangement, these have the potential to form spi-
double-stranded DNA. different promoters is currently unknown. ral filaments onto which a negatively supercoiled DNA

188 | MARcH 2010 | voluME 8 www.nature.com/reviews/micro

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

transcription in both E. coli 47 and S. Typhimurium46. IHF


has some of the characteristics of a conventional trans-
cription factor, being capable of recruiting σ54-containing
Binding site for RNA polymerase to promoters48. Its DNA-bending activ-
protein antagonist
ity can influence transcription by facilitating contact
between regulatory proteins and RNA polymerase49.
H-NS
However, other mechanisms have also been described.
For example, the ilvPG promoter of the ilvGMEDA operon
of E. coli is activated by IHF through the release of free
DNA bridging DNA coating
energy that is normally stored in the IHF-binding site,
making this energy available to assist with the formation
DNA wrapping DNA bending of the open transcription complex at the promoter 50. IHF
quenches the tendency of the DNA at the IHF-binding
site to become single-stranded, transferring this tendency
to the next-nearest labile region, which in this case is the
target promoter. This is thought to occur through dis-
placement, by protein-mediated DNA bending, of the
torsional energy that arises from DNA twist in negatively
supercoiled DNA. When this displaced torsion is applied
H-NS displaced H-NS displaced to a neighbouring double-stranded DNA sequence that
is prone to base-pairing disruption, a single-stranded
Figure 2 | Antagonistic DNA-binding modes of nucleoid-associated
Nature Reviewsproteins.
| Microbiology ‘bubble’ may form51. Any DNA segment that is prone
Histone-like nucleoid-structuring protein (H-NS) binds to DNA and influences its
structure. This binding can result in DNA coating (top right), in which the protein binds to the formation of single-stranded DNA may be vul-
to two sites that are close together on the same strand, or DNA bridging (top left), in nerable to supercoiling-induced duplex destabilization
which it binds to two sites that are further apart and causes the DNA between them (sIDD)51. The ability to displace sIDD is not restricted to
to form a loop. The integrity of the resulting bridge is compromised by the intervention IHF; Fis performs a similar function at a stable RNA gene
of a DNA-wrapping protein (green; bottom left) or by a DNA-bending protein (turquoise; promoter 52. However, the promoter concerned, that of
bottom right). The alternative interactions that result from DNA-binding of these the leuV tRNA operon, is unusual in that it possesses
proteins make the DNA–protein–DNA bridge untenable, and H-NS becomes displaced. one binding site for Fis rather than three sites arranged
in-phase along the double helix52. other well-characterized
stable RNA gene promoters, such as that of the tyrT
can be wrapped in modelling experiments42, suggesting tRNA gene, use a torsional transmission mechanism in
a plausible mechanism by which Hu can influence the which DNA writhe is stabilized by Fis, bound at three
organization of the nucleoid by directly guiding the path consecutive sites, and converted into DNA untwisting
of the DNA. However, evidence that this form of Hu is at the promoter 30,53. Bioinformatic analysis has iden-
the functional unit in vivo is lacking. tified sIDD-prone regions at the promoters of E. coli
In addition to its influence on DNA superhelicity, Hu genes that respond to environmental stress, although
contributes to DNA flexibility by bending the duplex. evidence that this is exploited as a global regulatory
Hu reduces the effective stiffness of DNA over short mechanism is lacking.
distances at low protein concentrations, but it stiffens IHF also affects chromosome replication initiation
DNA at high concentrations21. Hu-induced flexibility at the chromosomal origin, oriC, as do Fis and Hu54,
facilitates DNA loop formation, which is important for and influences the timing of replication initiation at the
both gene regulation and the architecture of the chromo- datA locus55. IHF is a component of several site-specific
some43. Interestingly, H-Ns has the opposite effect, and recombination systems, and it affects transposition56.
DNA loops form much more easily in bacteria that lack IHF therefore has roles in nucleoid structuring, chro-
this NAP43. mosome replication and DNA rearrangements, in addi-
tion to its role in transcription. In all of these cases, the
IHF and DNA U-turns. Integration host factor (IHF) primary role of IHF is in remodelling DNA at a local
was described originally as a cofactor in the site-specific level through the introduction of u-turns.
recombination of bacteriophage-λ44. IHF is related to Hu
at the level of amino acid sequence (FIG. 3) but has a distinct MukB and chromosome structural maintenance.
mode of interaction with DNA. Although Hu engages E. coli depends on the chromosome partition protein
in DNA wrapping in a sequence-independent manner, MukB for efficient segregation of its daughter chro-
IHF binds to a well-conserved nucleotide sequence and mosomes at cell division57. MukB-deficient mutants
DNA writhe
This approximates to our introduces a u-turn into the DNA, centred at the bind- exhibit chromosomal decondensation and fail to seg-
intuitive sense of DNA ing site45. In E. coli and related bacteria, IHF is composed regate daughter chromosomes at cell division; sup-
supercoiling. When helical of an α-subunit and β-subunit. The αβ heterodimer is pressor mutations correcting these phenotypes map
turns are added to or the predominant active form, although transcriptomics to topA, which encodes topoisomerase I, and result in
subtracted from relaxed DNA,
the DNA adopts a minimum
data from Salmonella enterica subsp. enterica serovar. increased negative supercoiling 58. This implies that a
energy state by winding the Typhimurium show that the αα and ββ forms are also functional link exists between DNA supercoiling and
helical axis about itself. biologically active46. The IHF protein influences global MukB activity. consistent with this, binding of MukB

NATuRE REvIEWs | Microbiology voluME 8 | MARcH 2010 | 189

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

1 10 20 30 40 50 60 70 80 90 100 110 120 130


H-NS
Protein interaction Linker DNA binding
1 10 20 30 40 50 60 70 80 90 100 110 120 130
StpA
Protein interaction Linker DNA binding

1 10 20 30 40 50 60 70
Hha
Loop
α-helix Secondary
1 10 20 30 40 50 60 70
structure
YdgT β-sheet
Histone_HNS
1 10 20 30 40 50 60 70 80 90
HTH_8
HUα
Hha domain Pfam
domains
1 10 20 30 40 50 60 70 80 90 Bac_DNA binding
IHFα
AsnC_trans_reg
1 10 20 30 40 50 60 70 80 90
Fis

1 10 20 30 40 50 60 70 80 90 100 110 120 130 140 150 160


Lrp
Protein interaction Protein interaction

Figure 3 | Nucleoid-associated protein secondary structure and domain organization. The predicted secondary
structures113 and Pfam domain organizations114 of the best characterized nucleoid-associated proteins (NAPs) of Salmonella
enterica subsp. enterica serovar Typhimurium are shown. The secondary structures of the α-subunits of integration host
factor (IHF) and HU are identical to those of the β-subunits (not shown). The locations of α-helices, β-sheets and loop
regions show that structural modularity is a feature of related NAPs (for example, histone-like nucleoid-structuring
protein (H-NS) and StpA). Those proteins that have the same Pfam domain belong to a family of related proteins as
determined by Hidden Markov Model analysis of protein sequences114. Fis, factor for inversion stimulation; Hha,
haemolysin expression-modulating protein; Lrp, leucine-responsive regulatory protein.

to DNA results in the introduction of protein-stabilized at genes that have been examined in detail reveal either
negative supercoils59. Furthermore, mutations in the a series of approximately equally spaced lrp-binding
mukB gene alter DNA supercoiling in plasmids as well sites or a continuous region of around 100 bp that is
as in the chromosome, which is consistent with a direct altered by lrp in its sensitivity to DNase I digestion64.
role for this protein in modulating DNA topology 60. These data are consistent with a model in which lrp
MukB has also been implicated in the formation of inde- octamers (or hexadecamers) bind and wrap the DNA.
pendent topological domains in the E. coli chromosome, The observation of regularly spaced sites of hyper-
probably in association with DNA gyrase61. sensitivity to DNase I is certainly consistent with DNA
wrapping by lrp70. In addition to bending and wrap-
Lrp: a physiological barometer. leucine-responsive ping the DNA, lrpc from Bacillus subtilis has been
regulatory protein (lrp) influences the transcription of reported to bridge DNA71,72. All of these features have
~10% of the genes in E. coli and, depending on the target implications for the ability of lrp to influence nucleoid
gene, the activity of lrp can be potentiated, inhibited or structure and to regulate transcription.
unaffected by leucine62. The lrp regulon includes genes lrp influences stable RNA expression in E. coli by
involved in nutrient uptake and amino acid metabolism. cooperating with H-Ns to repress rRNA operon tran-
lrp also has a role in microbial virulence, regulating scription73. The lrp gene is positively controlled by
genes involved in the phase-variable expression of pili. ppGpp, probably in an indirect manner 74. This means
It binds to DNA at a degenerate consensus sequence63 that lrp levels are highest at the end of the exponen-
and has a profound influence on the trajectory of the tial phase of growth, when the demand for stable RNA
bound DNA64. and other components of the translational machinery
lrp exists in variable oligomeric states: it can be is declining. This would correlate with the negative
detected in dimeric, octameric and hexadecameric action of lrp on rRNA transcription.
forms, with leucine binding favouring the dissocia-
tion of the hexadecamer to the octamer 65. The DNA- The versatile Fis protein. Fis has been studied
binding domain of lrp is located in the amino-terminal intensively from the perspective of both genome
portion of the protein, and in the multimers these organization and gene regulation. Fis activity has
domains are on the surface66–69. lrp binding patterns a major impact on the transcriptomes of E. coli and

190 | MARcH 2010 | voluME 8 www.nature.com/reviews/micro

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

S. Typhimurium75,76. It binds as a homodimer to a con- the promoters of the genes encoding the A and B subu-
sensus sequence that is usually 17 bp in length and nits of DNA gyrase, which produces negative supercoil-
AT rich, except at positions 2 and 16, where G or c ing in DNA. Depending on growth conditions, Fis is
residues are commonly found77. Dimeric Fis bends the also capable of activating or repressing the transcription
DNA at its binding site78, allowing DNA to bind to of topA88. Regulating the expression of the two major
the helix–turn–helix motif in each monomer 79. like topoisomerases allows Fis to influence DNA supercoil-
some of the other NAPs reviewed here, Fis contrib- ing globally, which has the potential to affect sensitive
utes to many DNA-based cellular activities, including promoters at the stable RNA genes. As discussed above,
DNA transcription, replication and recombination80. it has also been suggested that Fis can preserve a nega-
Bioinformatic analysis has detected potential bind- tively supercoiled DNA micro-domain at the promoters
ing sites for Fis throughout the E. coli genome, and by binding to its sites there89.
chromatin immunoprecipitation data show that it does An appreciation of the expression pattern of Fis is key
indeed bind to many locations, especially those outside to understanding its physiological relevance as a tran-
the coding regions of genes13. This is consistent with scriptional regulator. The protein is maximally expressed
information from in silico work showing that Fis pre- at entry to the exponential phase of growth, correlating
fers to bind to sequences that are commonly located at with the rise in demand for components of the transla-
transcription promoters81. tion apparatus to support rapid growth. Fis is therefore
Fis can repress or activate promoters, depending well placed to stimulate and sustain the activity of the
on the position of its binding site relative to that of promoters of the relevant genes. Fis negatively regulates
RNA polymerase. Fis represses transcription initia- its own gene, and the concentration of the protein drops
tion either by imposing a blockade at the target pro- to almost undetectable levels by the time the bacterium
moter and excluding RNA polymerase or by a more enters the stationary phase. one of the consequences
subtle mechanism, in which it modulates the RNA of this drop in Fis levels is the removal of its inhibitory
polymerase-mediated isomerization of the closed control of the gene encoding RNA polymerase σ-factor
transcription complex to an open complex. This raises Rpos, the σ-factor that reprogrammes RNA polymerase
the activation barrier to values that make transcription to transcribe genes required for adaptation to the cessa-
initiation very unlikely 82,83. tion of growth90. Rpos also exerts a negative influence,
Fis can also be a conventional transcription activa- presumably indirectly, on the transcription of fis 91. In
tor, making physical contact with RNA polymerase84. In this way, Fis, like other NAPs, influences the global tran-
addition, it acts from binding sites located at a distance scription pattern in response to changes in growth phase
from the promoter, through several distinct mecha- and physiological state80.
nisms that involve local DNA topology. one mechanism
involves the displacement of DNA twisting from an CbpA. curved-DNA-binding protein (cbpA) is related
upstream site to the target promoter by Fis-mediated to the chaperone protein DnaJ but, unlike DnaJ, has
DNA bending 52. This allows exploitation of sIDD at DNA-binding activity and has been grouped with the
the binding site through a mechanism similar to that NAPs. It is found in E. coli and many other bacteria, in
described above for IHF40. Another mechanism concerns which it contributes to growth at low and high tem-
the preservation of DNA writhe through Fis binding at a peratures and is required for normal cell division. It
negatively supercoiled region of the duplex 85. This main- forms a complex with chaperone modulatory protein
tains a micro-domain of negatively supercoiled DNA at (cbpM), which inhibits its activity. cbpM is subject to
the target promoter, sustaining transcription when DNA proteolysis by lon and clpAP, but only when it is not
elsewhere in the genome is becoming relaxed. This same in a complex with cbpA. The transcription of cbpA is
type of interaction may serve to impart structure to the under the control of lrp and is modulated by leucine;
genome by impeding duplex slithering, an activity that is it is also controlled by Rpos92.
also within the scope of IHF and H-Ns81.
There is an interesting correlation between Fis and EbfC: a new type of NAP. Borrelia burgdorferi, the cause
genes belonging to the stringent response regulon, of of lyme disease, expresses Ebfc, which is a DNA bind-
which fis itself is a member 86. Fis activity is not restricted ing protein that binds as a homodimer to a specific DNA
to genes of the stringent response, however, and it can sequence and bends the DNA. The crystal structures of
affect transcription across the genome77,78. stringently Ebfc from B. burgdorferi and of the corresponding protein
regulated promoters contain a Gc-rich discriminator from Haemophilus influenzae exhibit an unusual tweezer-
sequence downstream of the –10 position, and they like motif that is required for DNA binding. Although
are sensitive to changes in DNA supercoiling. Many Ebfc is involved in gene regulation, the widespread distri-
of the stringently controlled promoters encode stable bution of the consensus binding site in the DNA suggests
Duplex slithering RNA, and the relationships between the different fac- that it contributes to genome structuring 93.
In the absence of constraints tors that affect these promoters are complex and, in
(for example, due to protein some cases, controversial87. The involvement of Fis is Dps: an unusual NAP. Although DNA protection during
binding), the DNA duplex is not straightforward, because the protein is implicated at starvation protein (Dps), a ferritin-like protein, is usu-
free to migrate in the
plectonemically interwound,
two levels, at the least. The first is direct, with Fis bind- ally grouped with the NAPs, evidence that it is directly
supercoiled structure by ing to sites upstream of the stable-RNA gene promoter. involved in transcriptional regulation is largely lack-
adopting a slithering motion. The other is indirect and arises because Fis represses ing. The purified protein from E. coli has DNA-binding

NATuRE REvIEWs | Microbiology voluME 8 | MARcH 2010 | 191

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

activity 94 and is usually expressed in the stationary H-Ns (FIG. 4). It is also subject to indirect control through
phase, when the protein is thought to provide the DNA Rpos, the expression of which is influenced by Fis; the
with protection from damage. Transcription of dps in mRNA encoding Rpos also has a sensitivity at the post-
the stationary phase requires Rpos and IHF94 and is transcriptional level, along with hns mRNA, to the small
influenced directly by at least two other NAPs, Fis and regulatory RNA dsrA and the RNA chaperone Hfq95.
The cross-regulation by IHF, Fis and H-Ns at the dps
promoter is not unique. For example, H-Ns represses
a the promoters of the genes encoding its paralogues, stpA
Lag phase–log phase Log phase–stationary
transition phase transition and sfh96, and hns is itself under the control of Fis, such
that Fis offsets the autorepressive action of H-Ns97. lrp
Dps
negatively autoregulates its own gene98 and is an activator

Protein concentration
of stpA, the transcription of which is strongly enhanced
Log cell number

in bacteria grown in minimal medium99.

H-NS Crp: the NAP–transcription factor interface. cyclic


AMP regulatory protein (crp; also known as catabolite
IHF gene-activator protein (cAP)) has been traditionally
Growth
curve regarded as a conventional transcription factor 100. It is
active as a dimer and binds to a well-conserved DNA
Fis
sequence, the location of which at target promoters
allows the protein to act as a repressor or activator of
Time transcription. It requires a cofactor, cAMP, for binding
b
to DNA, and this provides a link between crp activity
Log phase and the physiology of the cell. crp was one of the first
RpoS
gene-regulatory proteins to be studied in detail, and it
has been considered in the context of some paradig-
H-NS
matic examples of transcription control, including the
RpoD lac operon of E. coli. To some extent, this may account
for the view that crp is a standard regulator of promoter
–35 –10 dps
activity. Nevertheless, it has been appreciated for many
Fis years that crp controls a very large regulon of genes, and
recent chIP-on-chip studies have confirmed this view
Stationary phase and extended it by showing that crp binds to hundreds
H-NS IHF of sites on the E. coli chromosome101. This has led to
the suggestion that crp has many of the characteristics
RpoS associated with NAPs101. like NAPs, crp is effective at
remodelling DNA owing to the introduction of a bend
–35 –10 dps
of approximately 90˚ at its binding site. The behav-
Figure 4 | complex regulation of transcription by iour of crp and its wide distribution across the genome,
multiple nucleoid-associated proteins. a | The expression combined with its very precise interactions with RNA
Nature Reviews | Microbiology
patterns of the four nucleoid-associated proteins (NAPs), polymerase when it acts as a transcriptional activator,
DNA protection from starvation protein (Dps), factor for point to the existence of a spectrum of regulatory pro-
inversion stimulation (Fis), histone-like nucleoid-structur- teins rather than independent categories with clearly
ing protein (H-NS) and integration host factor (IHF). A distinct properties.
typical bacterial growth curve is shown, with the lag
phase–log phase and log phase–stationary phase
Miscellaneous NAPs and NAP-like proteins. some
transitions indicated. The expression curves of the
four NAPs102 are representational summaries. b | The chromosome replication proteins have been described
regulatory inputs at the promoter of dps. During the early as NAPs102, including DnaA, seqA and chromosome
log phase of growth, Fis is abundant and binds to the dps initiation inhibitor (IciA). seqA binds to the GATc
promoter, creating a repression complex that traps RNA sequences in DNA that are methylated or hemimethy-
polymerase containing the RNA polymerase σ-factor layted in association with the DNA replication cycle,
RpoD 82. H-NS has a negative influence at all stages of and it contributes negatively to the control of chromo-
growth. RNA polymerase containing the σ-factor RpoS some replication initiation103. Mutants deficient in seqA
fails to gain access to the dps promoter as long as the exhibit altered expression of genes with GATc motifs
Fis–RpoD–promoter repression complex is intact. This in their promoter regions104. IciA and DnaA act with
complex becomes less tenable as Fis concentrations fall
mutual antagonism at oriC, with IciA seeking to inhibit
throughout the log phase and reach almost undetectable
levels with the onset of stationary phase. RNA polymerase the initiation process105. IciA is a member of the lysR
containing RpoS can then overcome the repressive family of transcription factors, a group that is not usu-
influence of H-NS; IHF then makes a positive contribution ally considered to include NAPs. on the other hand, the
to dps promoter function. Levels of Dps therefore increase lysR-like DNA-binding protein leuo has widespread
at the end of the exponential phase of growth. effects on the transcriptome of Gram-negative bacteria

192 | MARcH 2010 | voluME 8 www.nature.com/reviews/micro

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

and is an effective antagonist of H-Ns23. These proteins, sophistication of ligand binding or covalent modification
which have a preference for binding to AT-rich DNA, lie in response to environmental signals, this growth phase
along the spectrum between clear-cut examples of NAPs dependency provides a basis for a crude but wide-ranging
and conventional transcriptional regulators. This spec- global regulatory system. control of crp activity by cAMP
trum may provide many opportunities for the evolution availability possibly represents a more advanced form of
and refinement of gene regulatory switches. regulation. This second messenger is a useful reporter
of cellular physiology, because its concentration varies as
Concluding remarks the reciprocal of glucose concentration108. The cAMP–crp
one of the features of NAPs that makes them excel- system regulates virulence-gene expression in a number
lent regulators of gene expression at the global level is of pathogens, highlighting the integrated nature of the
their promiscuity in their interactions with DNA. NAP- infectious and housekeeping functions of these cells and
binding sites, discussed above, tend to be AT-rich, which the pivotal part that is played by cAMP–crp in both109.
is a feature associated with promoters. Thus, NAPs are In addition, it is important to note that in many cases
DNA-binding proteins with requirements for binding cAMP–crp acts in concert with or antagonistically to
that tend to direct them to promoters. once they have other NAPs and transcription factors100. Furthermore,
bound to a target promoter, their subsequent interac- the simple requirements of NAPs for DNA binding and
tions with that DNA have the potential to determine the their mutually antagonistic activities may allow these pro-
expression status of the gene concerned. teins to contribute to the process by which horizontally
Bridging of DNA may be incompatible with DNA acquired genes become integrated into the pre-existing
wrapping or DNA bending, but all three activities are regulatory networks of the cell.
associated with proteins in this class (FIG. 2). This provides In the immediate future there is scope for more
the basis for a simple genetic switch, whereby mutually work using imaging techniques to investigate the role
antagonistic activities between NAPs set the expression of NAPs as agents of nucleoid structure in the context of
state of the target gene. The variables in this relationship DNA replication and transcription, with the intention
include the availability of the antagonists. H-Ns is present of obtaining a real-time picture of the nucleoid as it under-
at an almost constant level per chromosome copy 106, goes its major molecular transactions. current knowl-
whereas its paralogues, stpA and sfh, show variation in edge of NAP biology allows hypotheses to be formed
their levels in the cell during growth107. Fis is available about the ways in which these proteins can contribute to
at high concentrations for only a portion of the growth the evolution of gene regulation; these hypotheses need
cycle; IHF also has a growth phase component in its to be tested in ways that take into account the nucleoid-
expression pattern, as does Hu, with the added variable structuring roles of NAPs. By deepening our understand-
of its subunit composition. The expression pattern of Dps ing of these fundamental processes, we will enhance our
is the reciprocal of that of Fis. Even without the added ability to manipulate bacteria to our benefit.

1. Drlica, K. & Rouvière-Yaniv, J. Histone-like proteins 11. Hardy, C. D. & Cozzarelli, N. R. A genetic selection 19. Dame R. T., Noom, M. C. & Wuite, G. J. Bacterial
of bacteria. Microbiol. Rev. 51, 301–319 (1987). for supercoiling mutants of Escherichia coli reveals chromatin organization by H-NS protein unraveled
2. Dorman, C. J. & Deighan, P. Regulation of gene proteins implicated in chromosome structure. Mol. using dual DNA manipulation. Nature 444, 387–390
expression by histone-like proteins in bacteria. Microbiol. 57, 1636–1652 (2005). (2006).
Curr. Opin. Genet. 13, 179–184 (2003). 12. Noom, M. C., Navarre, W. W., Oshima, T., Wuite, G. J. A single‑molecule study of H‑NS‑mediated bridging
3. Deng, S., Stein, R. A. & Higgins, N. P. Organization of & Dame, R. T. H-NS promotes looped domain in DNA.
supercoil domains and their reorganization by formation in the bacterial chromosome. Curr. Biol. 20. Hada, K. et al. Crystal structure and functional analysis
transcription. Mol. Microbiol. 57, 1511–1521 (2005). 17, R913–R914 (2007). of an archaeal chromatin protein Alba from the
4. Postow, L., Hardy, C. D., Arsuaga, J. & Cozzarelli, N. R. This article and reference 11 propose a role for H‑NS hyperthermophilic archaeon Pyrococcus horikoshii OT3.
Topological domain structure of the Escherichia coli in chromosomal loop formation and maintenance on Biosci. Biotechnol. Biochem. 72, 749–58 (2008).
chromosome. Genes Dev. 18, 1766–1779 (2004). the basis of a genetic analysis and whole‑genome 21. Luijsterburg, M. S., White, M. F., van Driel, R. &
5. Stein, R. A., Deng, S. & Higgins, N. P. Measuring protein‑binding data, respectively. Dame, R. T. The major architects of chromatin:
chromosome dynamics on different time scales using 13. Grainger, D. C., Hurd, D., Goldberg, M. D. & architectural proteins in bacteria, archaea and
resolvases with varying half-lives. Mol. Microbiol. 56, Busby, S. W. J. Association of nucleoid proteins with eukaryotes. Crit. Rev. Biochem. Mol. Biol. 43,
1049–1061 (2005). coding and non-coding segments of the Escherichia coli 393–418 (2008).
6. Hillebrand, A., Wurm, R., Menzel, A. & Wagner, R. genome. Nucleic Acids Res. 34, 4642–4652 (2006). 22. Dame, R. T., Wyman, C., Wurm, R., Wagner, R. &
The seven E. coli rRNA operon upstream regulatory 14. Oshima, T., Ishikawa, S., Kurokawa K., Aiba, H. & Goosen, N. Structural basis of H-NS-mediated
regions differ in structure and transcription factor Ogasawara, N. Escherichia coli histone-like protein trapping of RNA polymerase in the open initiation
binding efficiencies. Biol. Chem. 386, 523–534 H-NS binds to horizontally acquired DNA in association complex at rrnB P1. J. Biol. Chem. 277, 2146–2150
(2005). with RNA polymerase. DNA Res. 13, 141–153 (2006). (2002).
7. Potrykus, K. & Cashel, M. (p)ppGpp, still magical? 15. Lucchini, S. et al. H-NS mediates the silencing of 23. Stoebel, D. M., Free, A. & Dorman, C. J. Anti-silencing:
Annu. Rev. Microbiol. 62, 35–51 (2008). laterally acquired genes in bacteria. PLoS Pathog. overcoming H-NS-mediated repression of transcription
8. Rutherford, S. T., Villers, C. L., Lee, J. H., Ross, W. & 2, e81 (2006). in Gram-negative bacteria. Microbiol. 154,
Gourse, R. L. Allosteric control of Escherichia coli 16. Navarre, W. W. et al. Selective silencing of foreign DNA 2533–2545 (2008).
rRNA promoter complexes by DksA. Genes Dev. with low GC content by the H-NS protein in Salmonella. 24. Gordon, B. R., Imperial, R., Wang, L., Navarre, W. W. &
23, 236–248 (2009). Science 313, 236–238 (2006). Liu, J. Lsr2 of Mycobacterium represents a novel class
9. Jin, D. J. & Cabrera, J. E. Coupling the distribution This work and that in reference 15 provide of H-NS-like proteins. J. Bacteriol. 190, 7052–7059
of RNA polymerase to global gene regulation and evidence that H‑NS participates in a process called (2008).
the dynamic structure of the bacterial nucleoid in xenogeneic silencing to down‑regulate horizontally 25. Nye, M. B. & Taylor, R. K. Vibrio cholerae H-NS
Escherichia coli. J. Struct. Biol. 156, 284–291 acquired genes. domain structure and function with respect to
(2006). 17. Schneider, R. et al. An architectural role of the transcriptional repression of ToxR regulon genes
This paper describes the use of imaging techniques Escherichia coli chromatin protein FIS in organizing reveals differences among H-NS family members.
to monitor the formation of transcription factories DNA. Nucleic Acids Res. 29, 5107–5114 (2001). Mol. Microbiol. 50, 427–444 (2003).
in the E. coli nucleoid. 18. Zimmerman, S. B. Cooperative transitions of isolated 26. Castang, S., McManus, H. R., Turner, K. H. &
10. Marenduzzo, D., Faro-Trindade, I. & Cook, P. R. Escherichia coli nucleoids: implications for the Dove, S. L. H-NS family members function coordinately
What are the molecular ties that maintain genomic nucleoid as a cellular phase. J. Struct. Biol. 153, in an opportunistic pathogen. Proc. Natl Acad. Sci.
loops? Trends Genet. 23, 126–133 (2007). 160–175 (2006). USA 105, 18947–18952 (2008).

NATuRE REvIEWs | Microbiology voluME 8 | MARcH 2010 | 193

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

27. Timchenko, T., Bailone, A. & Deverot, R. BtcD, a 49. Santero, E. et al. Role of integration host factor in in Salmonella enterica serovar Typhimurium via the
mouse protein that binds to curved DNA, can stimulating transcription from the σ54-dependent nifH fimZ regulatory gene. J. Bacteriol. 190, 602–612
substitute in Escherichia coli for H-NS, a bacterial promoter. J. Mol. Biol. 227, 602–620 (1992). (2008).
nucleoid protein. EMBO J. 15, 3986–3992 (1996). 50. Sheridan, S. D., Benham, C. J. & Hatfield, G. W. 71. Beloin, C. et al. Contribution of DNA conformation
28. Hinton, J. C. D. et al. Expression and mutational Activation of gene expression by a novel DNA and topology in right-handed DNA wrapping by the
analysis of the nucleoid-associated protein H-NS of structural transmission mechanism that requires Bacillus subtilis LrpC protein. J. Biol. Chem. 278,
Salmonella typhimurium. Mol. Microbiol. 6, supercoiling-induced DNA duplex destabilization in 5333–5342 (2003).
2327–2337 (1992). an upstream activating sequence. J. Biol. Chem. 273, 72. Tapias, A., Lopez, G. & Ayora, S. Bacillus subtilis
29. Zhang, A., Rimsky, S., Reaban, M. E., Buc, H. & 21298–21308 (1998). LrpC is a sequence-independent DNA-binding and
Belfort, M. Escherichia coli protein analogs StpA and 51. Benham, C. J. Sites of predicted stress-induced DNA DNA-bending protein which bridges DNA. Nucleic
H-NS: regulatory loops, similar and disparate effects duplex destabilization occur preferentially at regulatory Acids Res. 28, 552–559 (2000).
on nucleic acid dynamics. EMBO J. 15, 1340–1349 loci. Proc. Natl Acad. Sci. USA 90, 2999–3003 (1993). 73. Pul, U., Wurm, R. & Wagner, R. The role of LRP and
(1996). 52. Opel, M. L. et al. Activation of transcription initiation H-NS in transcription regulation: involvement of
30. Maurer, S., Fritz, J. & Muskhelishvili, G. A systematic from a stable RNA promoter by a Fis protein-mediated synergism, allostery and macromolecular crowding.
in vitro study of nucleoprotein complexes formed by DNA structural transmission mechanism. Mol. J. Mol. Biol. 366, 900–915 (2007).
bacterial nucleoid-associated proteins revealing novel Microbiol. 53, 655–674 (2004). 74. Landgraf, J. R., Wu, J. & Calvo, J. M. Effects of
types of DNA organization. J. Mol. Biol. 387, This article and reference 50 describe promoter nutrition and growth rate on Lrp levels in Escherichia
1261–1276 (2009). activation at a distance through a mechanism coli. J. Bacteriol. 178, 6930–6936 (1996).
This study uses protein–DNA imaging techniques involving transmission of DNA twist energy 75. Bradley, M. D., Beach, M. B., de Koning, A. P.,
to analyze different NAP–DNA structures in vitro. following NAP binding. Pratt, T. S. & Osuna, R. Effects of Fis on Escherichia
31. Higgins, C. F. et al. A physiological role for DNA 53. Muskhelishvili, G., Buckle, M., Heumann, H., coli gene expression during different growth stages.
supercoiling in the osmotic regulation of gene Kahmann, R. & Travers, A. A. FIS activates sequential Microbiology 153, 2922–2944 (2007).
expression in S. typhimurium and E. coli. Cell 52, steps during transcription initiation at a stable RNA 76. Kelly, A. et al. A global role for Fis in the
569–584 (1988). promoter. EMBO J. 16, 3655–3665 (1997). transcriptional control of metabolism and type III
32. Mayer, O., Rajkowitsch, L., Lorenz, C., Konrat, R. & 54. Leonard, A. C. & Grimwade, J. E. Building a bacterial secretion in Salmonella enterica serovar Typhimurium.
Schröder, R. RNA chaperone activity and RNA binding orisome: emergence of new regulatory features for Microbiology 150, 2037–2053 (2004).
properties of the E. coli protein StpA. Nucleic Acids replication origin unwinding. Mol. Microbiol. 55, 77. Cho, B. K., Knight, E. M. Barrett, C. L. & Palsson, B. Ø.
Res. 35, 1257–1269 (2007). 978–985 (2005). Genome-wide analysis of Fis binding in Escherichia coli
33. Dorman, C. J. H-NS, a universal regulator for a 55. Nozaki, S., Yamada, Y. & Ogawa, T. Initiator titration indicates a causative role for A-/AT-tracts. Genome Res.
dynamic genome. Nature Rev. Microbiol. 2, 391–400 complex formed at datA with the aid of IHF regulates 18, 900–910 (2008).
(2004). replication timing in Escherichia coli. Genes Cells 78. Pan, C. Q. et al. Variable structures of Fis-DNA
34. Müller, C. M. et al. Role of histone-like proteins H-NS 14, 329–341 (2009). complexes determined by flanking DNA-protein
and StpA in expression of virulence determinants of 56. Haniford, D. B. Transpososome dynamics and contacts. J. Mol. Biol. 264, 675–695 (1996).
uropathogenic Escherichia coli. J. Bacteriol. 188, regulation in Tn10 transposition. Crit. Rev. Biochem. 79. Skoko, D. et al. Mechanism of chromosome
5428–5438 (2006). Mol. Biol. 41, 407–424 (2006). compaction and looping by the Escherichia coli
35. Madrid, C., Balsalobre, C., Garcia, J. & Juárez, A. 57. Niki, H., Jaffé, A., Imamura, R., Ogura, T. & Hiraga, S. nucleoid protein Fis. J. Mol. Biol. 364, 777–798
The novel Hha/YmoA family of nucleoid-associated The new gene mukB codes for a 177 kD protein with (2006).
proteins: use of structural mimicry to modulate the coiled-coil domains involved in chromosome 80. Dorman, C. J. Nucleoid-associated proteins and
activity of the H-NS family of proteins. Mol. Microbiol. partitioning of E. coli. EMBO J. 10, 183–193 (1991). bacterial physiology. Adv. Appl. Microbiol. 67, 47–64
63, 7–14 (2007). 58. Sawitzke, J. A. & Austin, S. Suppression of (2009).
36. Claret, L. & Rouvière-Yaniv, J. Variation in HU chromosome segregation defects of Escherichia coli 81. Pedersen, A. G., Jensen, L. J., Brunak, S.,
composition during growth of Escherichia coli: the muk mutants by mutations in topoisomerase I. Staerfeldt, H. H. & Ussery, D. W. A DNA structural
heterodimer is required for long term survival. J. Mol. Proc. Natl Acad. Sci. USA 97, 1671–1676 (2000). atlas for Escherichia coli. J. Mol. Biol. 299, 907–930
Biol. 273, 93–104 (1997). 59. Petrushenko, Z. M., Lai, C.-H., Rai, R. & Rybenkov, V. V. (2000).
37. Vis, H. et al. Solution structure of the HU protein from DNA reshaping by MukB: right-handed knotting, left- 82. Grainger, D. C., Goldberg, M. D., Lee, D. J. & Busby,
Bacillus stearothermophilus. J. Mol. Biol. 254, handed supercoiling. J. Biol. Chem. 281, 4606–4615 S. J. W. Selective repression by Fis and H-NS at the
692–703 (1995). (2006). Escherichia coli dps promoter. Mol. Microbiol. 68,
38. Mukherjee, A., DiMario, P. J. & Grove, A. 60. Weitao, T., Nordstrom, K. & Dasgupta, S. Escherichia 1366–1377 (2008).
Mycobacterium smegmatis histone-like protein is coli cell cycle control genes affect chromosome This paper elegantly illustrates the complex
nucleoid associated. FEMS Microbiol. Lett. 291, superhelicity. EMBO Rep. 1, 494–499 (2000). transcriptional control achieved by NAPs.
232–240 (2009). 61. Hsu, Y.-H., Chung, M.-W. & Li, T.-K. Distribution of 83. Schneider, R., Travers, A., Kutateladze, T. &
39. Castaing, B., Zelwer, C., Laval, J. & Bioteux, S. gyrase and topoisomerase IV on bacterial nucleoid: Muskhelishvili, G. A DNA architectural protein couples
HU protein of Escherichia coli binds specifically to implications for nucleoid organization. Nucleic Acids cellular physiology and DNA topology in Escherichia
DNA that contains single-strand breaks or gaps. Res. 34, 3128–3138 (2006). coli. Mol. Microbiol. 34, 953–964 (1999).
J. Biol. Chem. 270, 10291–10296 (1995). 62. Cho, B.-W., Barrett, C. L. Knight, E. M., Park, Y. S. & 84. McLeod, S. M., Aiyar, S. E., Gourse, R. L. &
40. Broyles, S. S. & Pettijohn, D. E. Interaction of the Palsson, B. Ø. Genome scale reconstruction of the Lrp Johnson, R. C. The C-terminal domains of the RNA
Escherichia coli HU protein with DNA. Evidence for regulatory network in Escherichia coli. Proc. Natl polymerase α subunits: contact site with Fis and
formation of nucleosome-like structures with altered Acad. Sci. USA 105, 19462–19467 (2008). localization during co-activation with CRP at the
DNA helical pitch. J. Mol. Biol. 187, 47–60 (1986). A very thorough genome‑wide analysis of a NAP Escherichia coli proP P2 promoter. J. Mol. Biol.
41. Oberto, J., Nabti, S., Jooste, V., Mignot, H. & regulon. 316, 517–529 (2002).
Rouvière-Yaniv, J. The HU regulon is composed of 63. Cui, Y., Wang, Q., Stormo, G. D. & Calvo, J. M. 85. Auner, H. et al. Mechanism of transcriptional
genes responding to anaerobiosis, acid stress, high A consensus sequence for binding of Lrp to DNA. activation by FIS: role of core promoter structure and
osmolarity and SOS induction. PLoS ONE 4, e4367 J. Bacteriol. 177, 4872–4880 (1995). DNA topology. J. Mol. Biol. 331, 331–344 (2003).
(2009). 64. Wang, Q. & Calvo, J. M. Lrp, a major regulatory 86. Mallik, P. et al. Growth phase-dependent regulation
42. Guo, F. & Adhya, S. Spiral structure of Escherichia coli protein in Escherichia coli, bends DNA and can and stringent control of fis are conserved processes
HUαβ provides foundation for DNA supercoiling. Proc. organize the assembly of a higher order nucleoprotein in enteric bacteria and involve a single promoter
Natl. Acad. Sci. USA 104, 4309–4314 (2007). structure. EMBO J. 12, 2495–2501 (1993). (fisP) in Escherichia coli. J. Bacteriol. 186, 122–135
This paper discusses how higher‑order forms of HU 65. Chen, S. & Calvo, J. M. Leucine-induced dissociation (2004).
can direct the path of DNA in the nucleoid. of Escherichia coli Lrp hexadecamers to octamers. 87. Dennis, P. P., Ehrenberg, M. & Bremer, H. Control of
43. Becker, N. A., Kahn, J. D. & Maher, L. M. III. Effects of J. Mol. Biol. 318, 1031–1042 (2002). rRNA synthesis in Escherichia coli: a systems biology
nucleoid proteins on DNA repression loop formation in 66. Willins, D. A., Ryan, C. W., Platko, J. V. & Calvo, J. M. approach. Microbiol. Mol. Biol. Rev. 68, 639–668
Escherichia coli. Nucleic Acids Res. 35, 3988–4000 Characterization of Lrp, an Escherichia coli regulatory (2004).
(2007). protein that mediates a global response to leucine. 88. Weinstein-Fischer, D. & Altuvia, S. Differential
44. Mumm, J. P., Landy, A. & Gelles, J. Viewing single λ J. Biol. Chem. 266, 10768–10774 (1991). regulation of Escherichia coli topoisomerase I
site-specific recombination events from start to finish. 67. Leonard, P. M. et al. Crystal structure of the Lrp by Fis. Mol. Microbiol. 63, 1131–1144 (2007).
EMBO J. 25, 4586–4595 (2006). like transcriptional regulator from the archaeon 89. Rochman, M. et al. Buffering of stable RNA promoter
45. Swinger, K. K. & Rice, P. A. IHF and HU: flexible Pyrococcus furiosus. EMBO J. 20, 990–997 (2001). activity against DNA relaxation requires a far upstream
architects of bent DNA. Curr. Opin. Struct. Biol. 68. Thaw, P. et al. , Structural insight into gene sequence. Mol. Microbiol. 53, 143–152 (2004).
14, 28–35 (2004). transcriptional regulation and effector binding by the 90. Hirsch, M. & Elliott, T. Fis regulates transcriptional
46. Mangan, M. W. et al. The integration host factor (IHF) Lrp/AsnC family. Nucleic Acids Res. 34, 1439–1449 induction of RpoS in Salmonella enterica. J. Bacteriol.
integrates stationary-phase and virulence gene (2006). 187, 1568–1580 (2005).
expression in Salmonella enterica serovar Typhimurium. 69. de los Rios, S. & Perona, J. J. Structure of the 91. Ó Cróinín, T. & Dorman, C. J. Expression of the Fis
Mol. Microbiol. 59, 1831–1847 (2006). Escherichia coli leucine-responsive regulatory protein protein is sustained in late exponential and early
47. Arfin, S. M. et al. Global gene expression profiling in Lrp reveals a novel octameric assembly. J. Mol. Biol. stationary phase cultures of Salmonella enterica
Escherichia coli K-12. The effects of integration host 366, 1589–1602 (2007). serovar Typhimurium grown in the absence of
factor. J. Biol. Chem. 275, 29672–29684 (2000). This study determines the higher‑order aeration. Mol. Microbiol. 66, 237–251 (2007).
48. Macchi, R. et al. Recruitment of σ54-RNA polymerase organization of the Lrp protein and shows the 92. Chenoweth, M. R. & Wickner, S. Complex regulation of
to the Pu promoter of Pseudomonas putida through sensitivity of this structure to leucine. the DnaJ homolog CbpA by the global regulators σs
integration host factor-mediated positioning switch 70. McFarland, K. A., Lucchini, S., Hinton, J. C. D. & and Lrp, by the specific inhibitor CbpM, and by the
of α subunit carboxyl-terminal domain on an UP-like Dorman, C. J. The leucine-responsive regulatory proteolytic degradation of CbpM. J. Bacteriol. 190,
element. J. Biol. Chem. 278, 27695–27702 (2003). protein, Lrp, activates transcription of the fim operon 5153–5161 (2008).

194 | MARcH 2010 | voluME 8 www.nature.com/reviews/micro

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

93. Riley, S. P. et al. Borrelia burgdorferi EbfC defines Escherichia coli. J. Mol. Microbiol. Biotechnol. 3, 125. Guo, L. et al. Biochemical and structural
a newly-identified, widespread family of bacterial 329–346 (2001). characterization of Cren7, a novel chromatin protein
DNA-binding proteins. Nucleic Acids Res. 37, 109. Liang, W., Pascual-Montano, A., Silva, A. J. & conserved among Crenarchaea. Nucleic Acids Res.
1973–1983 (2009). Benitez, J. A. The cyclic AMP receptor protein 36, 1129–1137 (2008).
94. Almiron, M., Link, A. J., Furlong, D. & Kolter, R. modulates quorum sensing, motility and multiple 126. Robinson, H. et al. The hyperthermophile
A novel DNA-binding protein with regulatory and genes that affect intestinal colonization in Vibrio chromosomal protein Sac7d sharply kinks DNA.
protective roles in starved Escherichia coli. Genes cholerae. Microbiology 153, 2964–2975 (2007). Nature 392, 202–205 (1998).
Dev. 6, 2646–2654 (1992). 110. Stavans, J. & Oppenheim, A. B. DNA-protein 127. Edmondson, S. P., Kahsai, M. A., Gupta, R. &
95. Gottesman, S. The small RNA regulators of interactions and bacterial chromosome architecture. Shriver, J. W. Characterization of Sac10a, a
Escherichia coli: roles and mechanisms. Annu. Phys. Biol. 3, R1–R10 (2006). hyperthermophile DNA-binding protein from
Rev. Microbiol. 58, 303–328 (2004). 111. Travers, A. & Muskhelishvili, G. Bacterial chromatin. Sulfolobus acidocaldarius. Biochemistry 43,
96. Deighan, P., Beloin, C. & Dorman, C. J. Three-way Curr. Opin. Genet. Dev. 15, 507–514 (2005). 13026–13036 (2004).
interactions among the Sfh, StpA and H-NS nucleoid- 112. Dorman, C. J. DNA supercoiling and bacterial gene 128. Elie, C., Baucher, M. F., Fondrat, C. & Forterre, P.
structuring proteins of Shigella flexneri 2a strain expression. Sci. Prog. 89, 151–166 (2006). A protein related to eucaryal and bacterial
2457T. Mol. Microbiol. 48, 1401–1416 (2003). 113. Rost, B., Yachdav, G. & Liu, J. The PredictProtein DNA-motor proteins in the hyperthermophilic
97. Falconi, M., Brandi, A., La Teana, A., Gualerzi, C. O. & Server. Nucleic Acids Res. 32, W321–W326 archaeon Sulfolobus acidocaldarius. J. Mol. Evol.
Pon, C. L. Antagonistic involvement of FIS and H-NS (2004). 45, 107–114 (1997).
proteins in the transcriptional control of hns 114. Finn, R. D. et al. The Pfam protein families database. 129. Luo, X. et al. CC1, a novel crenarchaeal DNA binding
expression. Mol. Microbiol. 189, 965–975 (1996). Nucleic Acids Res. 36, D281–D288 (2008). protein. J. Bacteriol. 189, 403–409 (2007).
98. McFarland, K. A. & Dorman, C. J. Autoregulated 115. Richmond, T. J. & Davey, C. A. The structure of DNA in 130. Shimizu, M., Miyake M., Kanke, F., Matsumoto, U. &
expression of the gene coding for the leucine- the nucleosome core. Nature 423, 145–150 (2003). Shindo, H. Characterization of the binding of HU and
responsive protein, Lrp, a global regulator in 116. Zhou, Y. B., Gerchman, S. E., Ramakrishnan, V., IHF, homologous histone-like proteins of Escherichia
Salmonella enterica serovar Typhimurium. Microbiol. Travers, A. & Muyldermans, S. Position and coli, to curved and uncurved DNA. Biochim. Biophys.
154, 2008–2016 (2008). orientation of the globular domain of linker histone H5 Acta 1264, 330–336 (1995).
99. Free, A. & Dorman, C. J. The Escherichia coli stpA on the nucleosome. Nature 395, 402–405 (1998). 131. Britton, R. A., Lin, D. C. & Grossman, A. D.
gene is transiently expressed during growth in rich 117. Ivanov, D. & Nasmyth, K. A topological interaction Characterization of a prokaryotic SMC protein
medium and is induced in minimal medium and by between cohesin rings and a circular minichromosome. involved in chromosome partitioning. Genes Dev.
stress conditions. J. Bacteriol. 179, 909–918 (1997). Cell 122, 849–860 (2005). 12, 1254–1259 (1998).
100. Browning, D. F. & Busby, S. J. W. The regulation of 118. Paull, T. T., Haykinson, M. J. & Johnson, R. C. 132. Morikawa, K. et al. Bacterial nucleoid dynamics:
bacterial transcription initiation. Nature Rev. The nonspecific DNA-binding and -bending proteins oxidative stress response in Staphylococcus aureus.
Microbiol. 2, 57–65 (2004). HMG1 and HMG2 promote the assembly of complex Genes Cells 11, 409–423 (2006).
101. Grainger, D. C., Hurd, D., Harrison, M., Holdstock, J. nucleoprotein structures. Genes Dev. 7, 1521–1534 133. Liu, Y., Chen, H., Kenney, L. J. & Yan, J. A divalent
& Busby, S. J. W. Studies of the distribution of (1993). switch drives H-NS/DNA binding conformations
Escherichia coli cAMP-receptor protein and RNA 119. Decanniere, K., Babu, A. M., Sandman, K., between stiffening and bridging modes. Genes Dev.
polymerase along the E. coli chromosome. Proc. Natl Reeve, J. N. & Heinemann, U. Crystal structures of (in the press).
Acad. Sci. USA 102, 17693–17698 (2005). recombinant histones HMfA and HMfB from the
102. Azam, T. A. & Ishihama, A. Twelve species of the hyperthermophilic archaeon Methanothermus Acknowledgements
nucleoid-associated protein from Escherichia coli. fervidus. J. Mol. Biol. 303, 35–47 (2000). We thank N. Ní Bhriain for critical comments on the manu-
Sequence recognition specificity and DNA binding 120. Leonard, P. M. et al. Crystal structure of the Lrp-like script. Work in the authors’ laboratory is supported by grants
affinity. J. Biol. Chem. 274, 33105–33113 (1999). transcriptional regulator from the archaeon from Science Foundation Ireland and the Wellcome Trust.
103. Lu M., Campbell, J. L., Boye, E. & Kleckner, N. SeqA: Pyrococcus furiosus. EMBO J. 20, 990–997
a negative modulator of replication initiation in E. coli. (2001). Competing interests statement
Cell 77, 413–426 (1994). 121. Cam, E. L., Culard, F., Larquet, E., Delain, E. & The authors declare no competing financial interests.
104. Prieto, A. et al. The GATC-binding protein SeqA is Cognet, J. A. DNA bending induced by the
required for bile resistance and virulence in Salmonella archaebacterial histone-like protein MC1. J. Mol. Biol.
enterica serovar Typhimurium. J. Bacteriol. 189, 285, 1011–1021 (1999). DATABASES
8496–8502 (2007). 122. Esser, D., Rudolph, R., Jaenicke, R. & Bohm, G. The Entrez Genome Project: http://www.ncbi.nlm.nih.gov/
105. Hwang, D. S. & Kornberg, A. Opposed actions of HU protein from Thermotoga maritima: recombinant genomeprj
regulatory proteins, DnaA and IciA, in opening the expression, purification and physicochemical Bacillus subtilis | Borrelia burgdorferi | Escherichia coli |
replication origin of Escherichia coli. J. Biol. Chem. characterization of an extremely hyperthermophilic Geobacillus stearothermophilus | Haemophilus influenzae |
267, 23087–23091 (1992). DNA-binding protein. J. Mol. Biol. 291, 1135–1146 Mycobacterium tuberculosis | Pseudomonas aeruginosa |
106. Free, A. & Dorman, C. J. Coupling of Escherichia coli (1999). Salmonella enterica | Vibrio cholerae
hns mRNA levels to DNA synthesis by autoregulation: 123. Herrmann, U. & Soppa, J. Cell cycle-dependent UniProtKB: http://www.uniprot.org
implications for growth phase control. Mol. Microbiol. expression of an essential SMC-like protein and CbpA | Crp | DksA | DnaA | Dps | EbfC | Fis | Hlp | H-NS | HUα |
18, 101–113 (1995). dynamic chromosome localization in the archaeon HUβ | IciA | LeuO | Lrp | MukB | RelA | RpoS | SeqA | StpA
107. Doyle, M. & Dorman, C. J. Reciprocal transcriptional Halobacterium salinarum. Mol. Microbiol. 46,
and posttranscriptional growth-phase-dependent 395–409 (2002). FURTHER INFORMATION
expression of sfh, a gene that encodes a paralogue 124. Peeters, E., Willaert, R., Maes, D. & Charlier, D. Author’s homepage:
of the nucleoid-associated protein H-NS. J. Bacteriol. Ss-LrpB from Sulfolobus solfataricus condenses about http://www.tcd.ie/Microbiology/research/cj_dorman.php
188, 7581–7591 (2006). 100 base pairs of its own operator DNA into globular Pfam: http://pfam.sanger.ac.uk
108. Tchieu, J. H., Norris, V., Edwards, J. S. & Saier, M. H. Jr. nucleoprotein complexes. J. Biol. Chem. 281, All liNks Are Active iN the oNliNe PDf
The complete phosphotransferase system in 11721–11728 (2006).

NATuRE REvIEWs | Microbiology voluME 8 | MARcH 2010 | 195

© 2010 Macmillan Publishers Limited. All rights reserved

You might also like