You are on page 1of 7

Bacterial enhancer-binding proteins: unlocking s54-dependent

gene transcription
Mathieu Rappas, Daniel Bose and Xiaodong Zhang

Bacterial transcription relies on the binding of dissociable (remodelling) involves losing this repressive structure.
sigma (s) factors to RNA polymerase (RNAP) for promoter Remodelling the closed complex into a transcriptionally
specificity. The major variant sigma factor (s54) forms a stable competent open complex requires energy derived from
closed complex with RNAP bound to DNA that rarely ATP hydrolysis catalysed by transcriptional activators.
spontaneously isomerises to an open complex. ATP hydrolysis Activators of closed s54 complexes are members of the
by bacterial enhancer-binding proteins is used to remodel the AAA+ (ATPases associated with various cellular activ-
RNAP–s54–DNA closed complex. Recently, a wealth of ities) protein family [6–10]. They bind DNA sites located
structural information on bacterial enhancer-binding proteins upstream of the promoter, known as upstream activation
has enabled unprecedented insights into their mechanism. sequences (UASs), and contact the closed complex
These data provide a structural basis for nucleotide binding and through DNA looping, often facilitated by DNA-bending
hydrolysis, oligomerisation and the conversion of ATPase proteins. This mode of interaction between the activator
activity into remodelling events within the RNAP–s54 closed and RNAP is similar to that of eukaryotic enhancer-
complex, and represent advances towards a complete binding proteins (EBPs), hence s54 activators are also
understanding of the s54-dependent transcription activation known as bacterial EBPs (bEBPs) [4,11–13].
mechanism.
bEBPs typically consist of three domains: an N-terminal
Addresses
Centre for Structural Biology, Division of Molecular Biosciences, Faculty regulatory (R) domain, a central AAA+ (C) domain and a
of Natural Sciences, Imperial College London, London SW7 2AZ, UK C-terminal DNA-binding (D) domain [14]. Recently,
several high- and low-resolution structures of bEBPs
Corresponding author: Zhang, Xiaodong bound to various nucleotide analogues (AMPPNP,
(xiaodong.zhang@imperial.ac.uk)
ATPgS, ADPAlFx, ADP) have become available (see
Table 1), unveiling the details of the s54-dependent
Current Opinion in Structural Biology 2007, 17:110–116 transcription activation mechanism. These include
high-resolution crystal structures of the NtrC1 R and C
This review comes from a themed issue on
Protein–nucleic acid interactions
domains (nitrogen regulation protein C1, NtrC1R+C)
Edited by James M Berger and Christoph W Müller [15], the PspF C domain (phage shock protein F,
PspFC) [16,17] and the ZraR C and D domains (hydro-
Available online 6th December 2006 genase regulatory protein G, ZraRC+D) [18]. In addition,
0959-440X/$ – see front matter
lower resolution electron microscopy (EM) reconstruc-
# 2006 Elsevier Ltd. All rights reserved. tions of NtrCR+C+D [19] and PspFC in complex with s54
[17] are now available. Finally, small/wide-angle X-ray
DOI 10.1016/j.sbi.2006.11.002 scattering (SAXS/WAXS) models of NtrCR+C+D have
been obtained [19]. ADPAlFx, which mimics the
ATP hydrolysis transition state, allows PspFC to form a
Introduction stable oligomer capable of interacting with RNAP–s54–
Regulated transcription is essential for cellular adapta- DNA. Spectroscopic analysis of PspFC–RNAP–s54–DNA
tion, differentiation and growth. In bacteria, transcription in the presence of ADPAlFx confirms that it is a bona fide
regulation is achieved through the use of dissociable intermediate ‘en route’ to open complex formation [20].
sigma (s) factors, which bind and direct RNA polymerase
(RNAP) to specific promoter DNA. Predominant s70 and In this review, we summarise recent structural data on
the alternative s54 form two distinct classes of s factors in bEBPs and propose a mechanistic model for s54-depen-
bacteria. The binding of s70–RNAP holoenzyme to pro- dent transcription activation, from initial binding of UAS
moter DNA is sufficient for promoter melting and tran- DNA to transcription initiation. In particular, we focus on
scription initiation — a process collectively known as how the recent structural data have advanced our under-
open complex formation [1–3]. The alternative s54 class standing of nucleotide binding and hydrolysis, oligomer-
is functionally distinct, as the binding of a s54–RNAP isation, and the interactions between bEBPs and s54.
holoenzyme to promoter DNA results in a stable closed
promoter complex, unable to initiate transcription [4,5]. Nucleotide binding and hydrolysis
This closed complex is stabilised by the interaction of s54 AAA+ proteins are characterised by the highly conserved
with a repressive fork junction DNA structure near –12, Walker A (GxxxxGK[T/S], where x represents any amino
with respect to the transcription start site (+1); activation acid) and B (hhhhDE, where h represents hydrophobic

Current Opinion in Structural Biology 2007, 17:110–116 www.sciencedirect.com


Bacterial enhancer-binding proteins Rappas, Bose and Zhang 111

Table 1

Recent structures of bacterial enhancer-binding proteins (determined since 2003).

Structure Organism Technique Resolution (Å) PDB/EMD code References


NtrC R domain (BeF complex) Salmonella typhimurium NMR – 1J56, 1KRX, 1KRW [25]
NtrC1 R and C domains Aquifex aeolicus X-ray 2.4 1NY5 [15]
NtrC1 C domain Aquifex aeolicus X-ray 3.1 1NY6 [15]
PspF1–275 C domain (apo, apo Escherichia coli X-ray 1.75, 1.7 2BJW, 2BJV [17]
R168A mutant)
NtrC1 R domain Aquifex aeolicus X-ray 3.03 1ZY2 [33]
PspF1–275 C domain (AMPPNP, Escherichia coli X-ray 1.9, 1.8, 1.9, 2.1 2C99, 2C96, 2C98, [16]
ATP, ADP, MgATP R227A mutant 2C9C
complexes)
ZraR C and D domains Salmonella typhimurium X-ray 3.0 1OJL [18]
s54–PspF C domain (ADPAlFx Sinorhizobium meliloti and Cryo-EM 20 EMD-1109 [17]
complex) Escherichia coli
NtrC R and C and D domains Salmonella typhimurium SAXS/WAXS – – [19]
(BeF and ADP complex)
NtrC R and C domains (ADP, Salmonella typhimurium Negative-stain EM 25, 28 EMD-1218 [19]
ADPAlFx complexes)

amino acids) motifs, responsible for nucleotide binding functional oligomeric state of bEBPs has been a matter of
and hydrolysis, respectively, and the sensor I and sensor II debate. Whereas the first X-ray structure of an oligomeric
motifs, responsible for sensing nucleotide states [6–10]. AAA+ domain (NtrC1C) was heptameric [15], the crystal
Several medium- and high-resolution structures of bEBPs structure of ZraRC+D revealed a hexameric structure [18]
confirm that, in common with all other AAA+ proteins, the (Figure 2), and nano-ESI (electrospray ionisation) mass
C domains of bEBPs fold into an a/b subdomain followed spectrometry and three-dimensional cryo-EM reconstruc-
by an a-helical subdomain. Nucleotide binds in the cleft tion revealed that PspFC is a hexamer when in complex
between these two subdomains (Figure 1) and between with s54 at the point of ATP hydrolysis [17]. Moreover,
two adjacent protomers (Figure 2). The C domain of the studies of ZraR and the three-dimensional cryo-EM
bEBPs is distinguished from other AAA+ family members reconstruction of the PspF–s54 complex suggest that
by the signature motif GAFTGA, located within surface bEBPs form non-planar asymmetric hexamers, indicating
loop L1 of the a/b subdomain [4,15,17,18]. Another that they may follow a non-concerted nucleotide binding/
surface loop, called L2, is also found in some AAA+ DNA hydrolysis mechanism. This is consistent with recent
helicases, including RuvB [17,21]. biochemical studies [26], and is similar to that exhibited
by other AAA+ proteins, including ClpX [27], papilloma-
Crystal structures of PspFC in complex with different virus E1 replicative helicase [28] and FtsH [29].
nucleotides have shed light on the conformational
changes that occur in the nucleotide-binding pocket Recent work on NorR (nitric oxide regulatory protein R)
during the hydrolysis cycle. Comparison of the different suggests that three inactive NorR dimers bind to three
nucleotide-bound structures identified regions and resi- adjacent UAS sites, resulting in an increased local con-
dues important for nucleotide binding and hydrolysis, as centration of NorR and facilitating oligomerisation upon
illustrated in Figure 1 [16]. Most interestingly, during activation of the regulatory GAF domain [30–32]. Oligo-
the early stages of ATP hydrolysis, a newly formed merisation of NorR would also promote DNA bending
interaction between a catalytic residue (E108 of Walker and DNA looping, essential for closed complex interac-
B) and a highly conserved structural residue (N64) acts as tion. Binding to DNA as dimers is supported by the
an ‘atomic switch’, enabling the bEBP to sense changes dimeric DNA-binding domains observed in the structure
within the catalytic site and transmit them to the L1 and of ZraR and the recently published negative-stain EM
L2 loops (Figure 1). reconstruction of NtrC, which reveals a symmetry mis-
match between the hexameric arrangement of both the C
Oligomerisation of the bEBP and R domains and the ‘trimer of dimers’ arrangement of
bEBPs are typically dimeric in their inactive state, with the D domains [18,19] (Figure 2).
the major dimerisation determinants existing either in the
R domain, as in DctD (C4-dicarboxylic acid transport The R domains sense specific environmental cues and
protein D) and NtrC1 [13,22,23], or in the D domain, adjust accordingly the activity of the AAA+ domain [10].
as in NtrC [15,24,25]. However, in response to stimu- One way of modulating AAA+ domain activity is to
latory signals, bEBPs form higher order oligomers via regulate oligomerisation, either negatively (as in NtrC1
their C AAA+ domain, which in turn stimulates their and DctD) or positively (as in NtrC and ZraR). The
ATPase activity and transcription activation. The exact inactive NtrC1 dimer is maintained by a tight interaction

www.sciencedirect.com Current Opinion in Structural Biology 2007, 17:110–116


112 Protein–nucleic acid interactions

Figure 1

Nucleotide binding and hydrolysis. (a) High-resolution crystal structures of PspFC in complex with ATP, ADP and, shown, MgATP (using
hydrolysis-deficient mutant R227A) reveal distinct changes during the ATP hydrolysis cycle. Nucleotide exchange induces changes in the
Walker A motif, loops L1 and L2, sensor II and the linker region between the two subdomains (blue). Nucleotide hydrolysis induces changes in
linker 1 and linker 2, L1 and L2, Walker B, sensor I and the linker region between the two subdomains (red). Regions that are affected by both
nucleotide exchange and hydrolysis are highlighted in magenta. (b) Enlarged view of the nucleotide-binding pocket. Residues involved in
nucleotide binding and hydrolysis are labelled.

Figure 2

Functional oligomerisation. (a) NtrC1R+C inactive dimer. (b) Schematic oligomeric arrangement of bEBPs. (c) Side and top views of an X-ray
crystallographic structure of the ZraRC+D hexamer. Side and top views of (d) SAXS/WAXS and (e) 3D EM models of activated (ADPAlFx-bound)
NtrCR+C+D, showing R domains on the periphery and D domains beneath the plane of the hexameric C domain.

Current Opinion in Structural Biology 2007, 17:110–116 www.sciencedirect.com


Bacterial enhancer-binding proteins Rappas, Bose and Zhang 113

between R domains that locks the AAA+ domains in a nificant roles in oligomerisation. This is consistent with
‘front to front’ orientation, thus preventing oligomerisa- crystal structures in which the nucleotide binds between
tion (Figure 2). Phosphorylation of the R domain alters its protomers. In AAA+ proteins, R fingers are proposed to be
conformation, causing greater conformational flexibility involved in intersubunit catalysis and nucleotide sensing
of the C domains and allowing repacking in the ‘front to [6–9]. In bEBPs, two highly conserved R fingers and
back’ orientation necessary for oligomerisation [15,33]. sensor II residues are indeed located at the protomer
In NtrC, phosphorylation exposes a hydrophobic patch in interface, ideally situated to sense changes in the nucleo-
the R domain, which allows the R domain of one protomer tide-binding pocket and correspondingly affect the oli-
to bind to the outside edge of the adjacent protomer, gomeric organisation.
thereby actively facilitating oligomerisation [19]. The
resulting conformation, with the R domains positioned on Interactions with s54
the periphery of the hexameric ring, would prevent steric To remodel the closed promoter complex, the UAS-
clashes when contacting the s54 holoenzyme [19]. bound bEBP must loop out the intervening DNA and
make contact with the complex at the promoter. Studies
Stable hexamers have been observed for PspF, NtrC, using variable operator lengths have revealed the impor-
NtrC1 and ZraR in the presence of different nucleotides tance of the geometry of this process, showing that bEBPs
[15,18,19,26,34], implying that nucleotides play sig- must bind to the opposite face of the promoter DNA to

Figure 3

Mechanistic model of s54-dependent transcription activation. (a) Inactive dimers bind to the UAS on the opposite DNA face to the RNAP–s54
complex, promoting oligomerisation. (b) Upon stimuli and through DNA looping, hexameric bEBP contacts RNAP–s54. (c) Role of the proposed
atomic switch pair during the ATP hydrolysis cycle. (i) Locked state of GAFTGA (ADP state); there is no interaction between N64 and E108.
(ii) Released state of GAFTGA (apo state). (iii) ‘Open’ state of GAFTGA (ATP state); interaction between N64 and E108 is established. (iv) ‘Engaged’
state of GAFTGA. Upon nucleotide binding and initial hydrolysis (ii and iii), changes in the nucleotide-binding pocket are sensed by the atomic
switch and transmitted through a conformational signalling pathway to allow a productive interaction between the L1 and L2 loops of bEBP and
s54 (iv). g-Phosphate release changes L1 and L2 into the locked conformation (i). (d) Combining available structural information, the transcriptional
intermediate complex shown induces changes in RNAP–s54–DNA that result in open complex formation.

www.sciencedirect.com Current Opinion in Structural Biology 2007, 17:110–116


114 Protein–nucleic acid interactions

that bound by the s54 holoenzyme to efficiently initiate we present the current mechanistic model of bEBP-
transcription [35]. The bEBP GAFTGA signature motif dependent transcription activation (Figure 3).
in the AAA+ domain forms the main contact between
bEBPs and s54; mutations within this motif generally Dimeric bEBPs bind to the UAS DNA distantly and to
abolish s54 interaction and transcription initiation [36,37]. the opposite face of DNA to that bound by RNAP–s54,
As revealed by the crystal structures of NtrC1 and ZraR, thereby increasing the local concentration of bEBPs.
GAFTGA is located at the tip of the L1 loop, which is Upon receipt of an appropriate stimulus, the R domain
inserted into helix 3 located at the surface of the a/b facilitates formation of an unstable hexamer, capable of
subdomain and near the hexameric pore. binding and hydrolysing ATP. The oligomerisation of
the bEBPs and other DNA-bending proteins would
From these structures, it is not obvious how the then facilitate DNA looping between the UAS and
GAFTGA motif could interact with s54. Hydrophobic the promoter, bringing the bEBP into close proximity
interactions involving the phenylalanine of GAFTGA, a with RNAP–s54. Nucleotide binding stabilises the hex-
valine from the adjacent L2 loop (Figure 1) and residues amer and initial hydrolysis within certain subunits is
from helix 3 lock GAFTGA in a conformation pointing sensed by the atomic switch pair (PspF E108–N64)
towards the central pore of the ring [15]. This con- and transmitted through the conformational signalling
formation is unfavourable for stable s54 interaction, as pathway to release the locked L1 and L2 loops. Exposure
the GAFTGA motif is buried and not readily accessible. of one or more GAFTGA motifs within the hexamer
This is supported by biochemical studies indicating leads to a stable interaction with s54. Completion of
that the s54-binding site may be masked before ATP hydrolysis and g-phosphate release (from ATP to
hydrolysis and that the threonine of GAFTGA might ADP) disrupts the atomic switch pair interaction, releas-
make a direct interaction with s54 [36]. Fitting the crystal ing the conformational strain that caused the exposure of
structure of PspFC into the low-resolution three-dimen- the L1 and L2 loops and allowing the L1-locking
sional cryo-EM map of the PspFC–s54 complex revealed mechanism to be re-engaged [15,16,17]. Changes
that the L1 loop (with GAFTGA at its tip) has to project in L1 conformation will cause changes in RNAP–s54
upwards from the plane of the hexameric PspF ring that result in the release of fork junction DNA repression,
to contact s54 [17]. A raised ‘rim’ corresponding to leading to open complex formation and transcription
the L1 and L2 loops also appears in NtrC in the presence initiation.
of the ATP hydrolysis transition state analogue
ADPAlFx and is absent in the presence of ADP Our current data provide evidence of the origin of con-
[19]. The raised conformation of L1 and L2 is thought formational changes in the activator (Figure 3) and a
to happen at the point of ATP hydrolysis and to be part structural model for the transcriptional intermediate com-
of a coordinated series of movements that occurs in plex, as shown in Figure 3. However, we still lack a
response to changing nucleotide states during the hydro- detailed understanding of how the energy released from
lysis cycle. nucleotide hydrolysis in the activator is used to remodel
the RNAP–s54–DNA closed complex, resulting in DNA
The chemical energy released by ATP hydrolysis in the melting and open complex formation. This requires a
active site is converted into conformational changes that mechanistic understanding of how the nucleotide activity
ultimately lead to the relocation of the L1 and L2 loops. of one subunit influences the activity and functionality of
Recent studies on PspFc in complex with different other subunits within the hexameric bEBP. In addition,
nucleotides provided a structural basis for this conversion structures and dynamics of bEBP in complex with
[16]. The changes in the nucleotide-binding pocket are RNAP–s54–DNA are necessary for a complete under-
sensed by the atomic switch pair (E108–N64) and com- standing of the s54-dependent transcription activation
municated through a conformational signalling pathway, process.
resulting in the rotation of helix 3, where L1 is inserted
(Figure 3). This disrupts the above-mentioned hydro- Acknowledgements
phobic interactions that lock L1 and the GAFTGA motif, We are indebted to many useful discussions with Martin Buck, Paul
Freemont, Tillmann Pape, Ramesh Wignerawaraj, Nicolas Joly and Jorg
and moves L1 into the raised conformation. The inter- Schumacher. This work is supported by the Wellcome Trust and the
action between a residue in L1 (E81) and a residue in L2 Biotechnology and Biological Sciences Research Council.
(R131) also ensures the coordinated movement of these
two loops. References and recommended reading
Papers of particular interest, published within the period of review,
have been highlighted as:
Conclusions: from binding to initiation
Recent structural studies have provided a detailed under-  of special interest
 of outstanding interest
standing of bEBP nucleotide binding and hydrolysis,
functional oligomerisation and interactions with s54. 1. Busby S, Ebright RH: Transcription activation by catabolite
Combining the structural information discussed above, activator protein (CAP). J Mol Biol 1999, 293:199-213.

Current Opinion in Structural Biology 2007, 17:110–116 www.sciencedirect.com


Bacterial enhancer-binding proteins Rappas, Bose and Zhang 115

2. Browning DF, Busby SJ: The regulation of bacterial 19. De Carlo S, Chen B, Hoover TR, Kondrashkina E, Nogales E,
transcription initiation. Nat Rev Microbiol 2004, 2:57-65.  Nixon BT: The structural basis for regulated assembly and
function of the transcriptional activator NtrC. Genes Dev 2006,
3. Ishihama A: Functional modulation of Escherichia coli RNA 20:1485-1495.
polymerase. Annu Rev Microbiol 2000, 54:499-518. The authors present EM and SAXS/WAXS studies of full-length NtrC in
complex with ADP and ADPAlFx. They also provide evidence of the
4. Zhang X, Chaney M, Wigneshweraraj SR, Schumacher J, relocation of the R domains, GAFTGA motifs and D domains.
Bordes P, Cannon W, Buck M: Mechanochemical ATPases and
transcriptional activation. Mol Microbiol 2002, 45:895-903. 20. Leach RN, Gell C, Wigneshweraraj SR, Buck M, Smith A,
Stockley PG: Mapping ATP-dependent activation at a sigma54
5. Buck M, Gallegos MT, Studholme DJ, Guo Y, Gralla JD: The promoter. J Biol Chem 2006, 281:33717-33726.
bacterial enhancer-dependent sigma(54) (sigma(N))
transcription factor. J Bacteriol 2000, 182:4129-4136. 21. Yamada K, Kunishima N, Mayanagi K, Ohnishi T, Nishino T,
Iwasaki H, Shinagawa H, Morikawa K: Crystal structure of the
6. Erzberger JP, Berger JM: Evolutionary relationships and Holliday junction migration motor protein RuvB from Thermus
structural mechanisms of AAA+ proteins. Annu Rev Biophys thermophilus HB8. Proc Natl Acad Sci USA 2001, 98:1442-1447.
Biomol Struct 2006, 35:93-114.
22. Park S, Meyer M, Jones AD, Yennawar HP, Yennawar NH,
7. Hanson PI, Whiteheart SW: AAA+ proteins: have energy, will Nixon BT: Two-component signaling in the AAA+ ATPase
work. Nat Rev Mol Cell Biol 2005, 6:519-529. DctD: binding Mg2+ and BeF3- selects between alternate
dimeric states of the receiver domain. FASEB J 2002,
8. Lupas AN, Martin J: AAA proteins. Curr Opin Struct Biol 2002, 16:1964-1966.
12:746-753.
23. Meyer MG, Park S, Zeringue L, Staley M, McKinstry M,
9. Ogura T, Wilkinson AJ: AAA+ superfamily ATPases: common Kaufman RI, Zhang H, Yan D, Yennawar N, Yennawar H et al.:
structure–diverse function. Genes Cells 2001, 6:575-597. A dimeric two-component receiver domain inhibits the
sigma54-dependent ATPase in DctD. FASEB J 2001,
10. Neuwald AF, Aravind L, Spouge JL, Koonin EV: AAA+: a class of 15:1326-1328.
chaperone-like ATPases associated with the assembly,
operation, and disassembly of protein complexes. Genome 24. Pelton JG, Kustu S, Wemmer DE: Solution structure of the
Res 1999, 9:27-43. DNA-binding domain of NtrC with three alanine substitutions.
J Mol Biol 1999, 292:1095-1110.
11. Hahn S: Structure and mechanism of the RNA polymerase II
transcription machinery. Nat Struct Mol Biol 2004, 11:394-403. 25. Hastings CA, Lee SY, Cho HS, Yan D, Kustu S, Wemmer DE: High-
resolution solution structure of the beryllofluoride-activated
12. Schumacher J, Joly N, Rappas M, Zhang X, Buck M: Structures NtrC receiver domain. Biochemistry 2003, 42:9081-9090.
and organisation of AAA+ enhancer binding proteins in
transcriptional activation. J Struct Biol 2006, 156:190-199. 26. Joly N, Schumacher J, Buck M: Heterogeneous nucleotide
 occupancy stimulates functionality of PspF, an AAA+
13. Wigneshweraraj S, Burrows PC, Bordes P, Schumacher J, transcriptional activator. J Biol Chem 2006, 281:34997-35007.
Rappas M, Finn RD, Cannon WV, Zhang X, Buck M: The second The authors present evidence of heterogeneous occupancy of nucleotide
paradigm for activation of transcription. Prog Nucleic Acid Res in PspF, and argue in support of stochastic nucleotide binding and a non-
Mol Biol 2005, 79:339-369. concerted molecular mechanism.
14. Studholme DJ, Dixon R: Domain architectures of sigma54- 27. Martin A, Baker TA, Sauer RT: Rebuilt AAA+ motors reveal
dependent transcriptional activators. J Bacteriol 2003, operating principles for ATP-fuelled machines. Nature 2005,
185:1757-1767. 437:1115-1120.
15. Lee SY, De La Torre A, Yan D, Kustu S, Nixon BT, Wemmer DE: 28. Enemark EJ, Joshua-Tor L: Mechanism of DNA translocation in
 Regulation of the transcriptional activator NtrC1: structural a replicative hexameric helicase. Nature 2006, 442:270-275.
studies of the regulatory and AAA+ ATPase domains. Genes
Dev 2003, 17:2552-2563. 29. Suno R, Niwa H, Tsuchiya D, Zhang X, Yoshida M, Morikawa K:
The authors present the high-resolution structure of the NtrC1 C and R Structure of the whole cytosolic region of ATP-dependent
domains — the first high-resolution structure of a bEBP C domain. The protease FtsH. Mol Cell 2006, 22:575-585.
paper also proposes a mechanism for negative regulation of oligomer-
isation by the R domain. 30. Tucker NP, D’Autreaux B, Studholme DJ, Spiro S, Dixon R: DNA
binding activity of the Escherichia coli nitric oxide sensor NorR
16. Rappas M, Schumacher J, Niwa H, Buck M, Zhang X: Structural suggests a conserved target sequence in diverse
 basis of the nucleotide driven conformational changes in the proteobacteria. J Bacteriol 2004, 186:6656-6660.
AAA+ domain of transcription activator PspF. J Mol Biol 2006,
357:481-492. 31. D’Autreaux B, Tucker NP, Dixon R, Spiro S: A non-haem iron
Transiently stable crystals of PspF in the presence of ATP pin down the centre in the transcription factor NorR senses nitric oxide.
origin of conformational changes within the nucleotide pocket. An atomic Nature 2005, 437:769-772.
switch pair, responsible for sensing changes during the nucleotide 32. Tucker N, D’Autreaux B, Spiro S, Dixon R: DNA binding
hydrolysis cycle and transmitting them through a conformational signal- properties of the Escherichia coli nitric oxide sensor NorR:
ling pathway to the s54-interacting determinants, is proposed in this work. towards an understanding of the regulation of flavorubredoxin
expression. Biochem Soc Trans 2005, 33:181-183.
17. Rappas M, Schumacher J, Beuron F, Niwa H, Bordes P,
 Wigneshweraraj S, Keetch CA, Robinson CV, Buck M, Zhang X: 33. Doucleff M, Chen B, Maris AE, Wemmer DE, Kondrashkina E,
Structural insights into the activity of enhancer-binding Nixon BT: Negative regulation of AAA+ ATPase assembly by
proteins. Science 2005, 307:1972-1975. two component receiver domains: a transcription activation
The first structural evidence that hexameric PspF interacts with s54. mechanism that is conserved in mesophilic and extremely
Fitting the high-resolution structure of PspF into a cryo-EM reconstruction hyperthermophilic bacteria. J Mol Biol 2005, 353:242-255.
of the PspF–s54 complex reveals an engaged (projecting) GAFTGA
conformation. The study also proposes a conformational signalling path- 34. Chaney M, Grande R, Wigneshweraraj SR, Cannon W, Casaz P,
way that transmits changes in the nucleotide-binding pocket to the s54 Gallegos MT, Schumacher J, Jones S, Elderkin S, Dago AE et al.:
interaction. Binding of transcriptional activators to sigma 54 in the
presence of the transition state analog ADP-aluminum
18. Sallai L, Tucker PA: Crystal structure of the central and fluoride: insights into activator mechanochemical action.
 C-terminal domain of the sigma(54)-activator ZraR. Genes Dev 2001, 15:2282-2294.
J Struct Biol 2005, 151:160-170.
The authors present the first hexameric crystal structure of bEBP C and D 35. Huo YX, Tian ZX, Rappas M, Wen J, Chen YC, You CH, Zhang X,
domains. The structure suggests a non-symmetric hexamer with hetero-  Buck M, Wang YP, Kolb A: Protein-induced DNA bending
geneous nucleotide binding and shows the D domains located beneath clarifies the architectural organization of the sigma54-
the plane of the hexameric ring in a dimeric arrangement. dependent glnAp2 promoter. Mol Microbiol 2006, 59:168-180.

www.sciencedirect.com Current Opinion in Structural Biology 2007, 17:110–116


116 Protein–nucleic acid interactions

An elegant demonstration that bEBP binds to the opposite face of DNA to a surface that binds sigma 54. Proc Natl Acad Sci USA 2003,
that bound by RNAP–s54, and that promoter DNA is sandwiched between 100:2278-2283.
RNAP–s54 and activator.
37. Gallegos MT, Cannon WV, Buck M: Functions of the sigma(54)
36. Bordes P, Wigneshweraraj SR, Schumacher J, Zhang X, region I in trans and implications for transcription activation.
Chaney M, Buck M: The ATP hydrolyzing transcription J Biol Chem 1999, 274:25285-25290.
activator phage shock protein F of Escherichia coli: identifying

Current Opinion in Structural Biology 2007, 17:110–116 www.sciencedirect.com

You might also like