You are on page 1of 15

View Article Online / Journal Homepage / Table of Contents for this issue

CRITICAL REVIEW www.rsc.org/metallomics | Metallomics

Interplay of metal ions and urease


Eric L. Carter,a Nicholas Flugga,a Jodi L. Boer,b Scott B. Mulrooneya
and Robert P. Hausinger*abc
Received 19th February 2009, Accepted 23rd March 2009
First published as an Advance Article on the web 9th April 2009
DOI: 10.1039/b903311d

Urease, the first enzyme to be crystallized, contains a dinuclear nickel metallocenter that catalyzes
the decomposition of urea to produce ammonia, a reaction of great agricultural and medical
importance. Several mechanisms of urease catalysis have been proposed on the basis of enzyme
crystal structures, model complexes, and computational efforts, but the precise steps in catalysis
and the requirement of nickel versus other metals remain unclear. Purified bacterial urease is
Published on 09 April 2009. Downloaded on 10/10/2014 08:35:54.

partially activated via incubation with carbon dioxide plus nickel ions; however, in vitro activation
also has been achieved with manganese and cobalt. In vivo activation of most ureases requires
accessory proteins that function as nickel metallochaperones and GTP-dependent molecular
chaperones or play other roles in the maturation process. In addition, some microorganisms
control their levels of urease by metal ion-dependent regulatory mechanisms.

1.0 Introduction to urease agricultural implications.3,4 Urease acts as a virulence factor


in many pathogenic microorganisms, including those associated
Urease holds a prominent place in the history of Science. It with urinary stone formation, pyelonephritis, ammonia
was the first enzyme to be crystallized1 and the first shown to encephalopathy, and hepatic coma;5,6 but perhaps the most
contain nickel.2 The enzyme catalyzes the deceptively simple infamous pathogenic organism that exploits urease is
hydrolysis of urea into ammonia and carbamic acid. The latter Helicobacter pylori, which takes advantage of the increase in
compound spontaneously decomposes into carbonic acid and pH associated with the hydrolysis of urea to colonize the acidic
a second ammonia molecule, as shown in the reactions below: stomach.7–9 Urease also participates in the environmental
cycling of various nitrogen compounds, including urea-based
H2N–C(O)–NH2 + H2O - NH3 + H2N–COOH
fertilizers.4 In addition, the enzyme plays a role in seed
germination by degrading urea formed from arginase
H2N–COOH + H2O - NH3 + H2CO3 activity.10 The multi-faceted significance of this enzyme has
stimulated widespread interest in its characterization.
The increase in ammonia levels and consequent elevation of
Ureases have been isolated and characterized from various
pH associated with this reaction have major medical and
bacteria, fungi, and plants.3,4,11–13 Despite having differences
a
Department of Microbiology and Molecular Genetics, Michigan in their quaternary structures, ureases possess essentially
State University, East Lansing, Michigan 48824-4320, USA. identical folds and are united by a basic trimeric structure
b
E-mail: hausinge@msu.edu containing three catalytic centers.14 Most bacterial ureases,
Department of Biochemistry and Molecular Biology, Michigan State exemplified by those from Klebsiella aerogenes (Fig. 1A)15 and
University, East Lansing, Michigan 48824-1319, USA
c
Quantitative Biology Program, Michigan State University, Bacillus pasteurii,16 possess three distinct types of subunits that
East Lansing, Michigan 48824-4320, USA form a (UreABC)3 structure. Urease from H. pylori has only

Eric L. Carter is a graduate Nicholas Flugga is a graduate


student who received his BS in student who received his BS in
Biomedical Science from Biology at the University of
Western Michigan University Findlay in 2007. His research
in 2005 then worked as focuses on testing whether
a research technician at UreB shifts by a hinge-like
Midwestern University until motion to allow access to the
2007. His area of research nascent active site during
focuses on the involvement of urease activation.
UreD in urease activation and
testing of a putative nickel-
independent urease.

Eric L. Carter Nicholas Flugga

This journal is 
c The Royal Society of Chemistry 2009 Metallomics, 2009, 1, 207–221 | 207
View Article Online

Fig. 1 The structures of three well-characterized ureases. (A) K. aerogenes urease (PDB access code 1fwj) with UreA depicted in blue, UreB in
Published on 09 April 2009. Downloaded on 10/10/2014 08:35:54.

orange, and UreC in yellow, together forming a (UreABC)3 structure. (B) H. pylori urease (1e9z) with UreA (corresponding to a fusion of the two
small subunits in the K. aerogenes enzyme) depicted in blue and UreB (analogous to UreC in the K. aerogenes protein) shown in yellow for one
(UreAB)3 unit, with three more (UreAB)3 units shown in gray included in the biologically relevant [(UreAB)3]4 structure. (C) Jack bean urease with
one subunit (comparable to a fusion of all three K. aerogenes subunits) shown in gold in the otherwise blue hexameric protein (two trimers
interacting in a face-to-face manner and shown after a 90 degree rotation compared to the other ureases). (Copied by permission of the
International Union of Crystallography; http://journals.iucr.org/).

two subunits, the smaller of which is equivalent to a fusion of ((UreAB)3)4 complex as shown in Fig. 1B.17 Eukaryotic
UreA and UreB found in other bacterial enzymes. The two organisms exhibit yet another configuration, as illustrated by
H. pylori urease subunits form a trimer of dimers that closely jack bean (Canavalia ensiformis) urease—the enzyme used in
resembles other ureases, but this unit forms a supramolecular the pioneering studies mentioned above. This plant enzyme
contains a single type of subunit which represents a fusion of
the separate subunits found in bacterial ureases. The subunit
assembles into a trimer analogous to the (UreABC)3 trimer
Jodi L. Boer is a graduate
shown for the K. aerogenes enzyme (Fig. 1A), and two of these
student who received her BS
at Calvin College in 2007. She trimers dimerize in a face-to-face manner to generate the
carries out structural and native hexameric structure (rotated 90 degrees relative to the
biochemical studies of UreG, other enzymes in Fig. 1C).18
required for activation of this The active site of urease contains two nickel atoms bridged
nickel-containing enzyme. by a carboxylated lysine and a solvent molecule, as shown for
the K. aerogenes enzyme (Fig. 2).19 In addition, Ni1 is
coordinated by two His residues plus a terminal water
molecule while Ni2 is coordinated by two His, an Asp, and
a terminal solvent. Also present at the active site are two His
residues thought to participate in substrate binding and/or
catalysis. This active site structure is generally consistent with
Jodi L. Boer results derived from earlier biophysical investigations of the

Scott B. Mulrooney received Robert P. Hausinger received


his PhD from MSU in 1990, his PhD at the University of
was a postdoctoral fellow and Minnesota in 1982. After post-
lecturer at the University of doctoral training at M.I.T.
Michigan, and returned to (1982–1984) he joined the
MSU in 1999 where he is faculty at Michigan State
now an Associate Research University where he is Profes-
Professor. His urease studies sor of Microbiology and
have focused on characteriza- Biochemistry, and Director of
tion of UreE. the Quantitative Biology
Program. His research focuses
on the mechanisms of metallo-
center biosynthesis and the
catalytic mechanisms and ver-
Scott B. Mulrooney Robert P. Hausinger satility of FeII/a-ketoglutarate
hydroxylases.

208 | Metallomics, 2009, 1, 207–221 This journal is 


c The Royal Society of Chemistry 2009
View Article Online

in vitro with nickel and other metal ions, summarize our


current understanding of how urease is activated in vivo in
bacteria, fungi, and plants, and highlight urease genes that
exhibit metal-dependent regulation.

2.0 Role of nickel in urease


Soon after the discovery that urease contains two nickel ions
per active site, Dixon et al. proposed a catalytic mechanism
incorporating both metals.32 These researchers hypothesized
that one metal site binds the urea carbonyl oxygen and
enhances the electrophilicity of the carbon atom, the second
metal increases the nucleophilicity of a water molecule, and
protein side chains serve additional acid/base roles to achieve
urea hydrolysis. Notably, this hydrolysis reaction is distinct
Published on 09 April 2009. Downloaded on 10/10/2014 08:35:54.

from the non-catalyzed decomposition of urea which proceeds


by elimination of ammonia with consequent formation of
cyanic acid (OQCQN–H), a compound that is not observed
Fig. 2 The urease active site. The active site of urease contains two in enzymatic reactions. Informed by the structure of the
nickel atoms (green) bridged by a carboxylated lysine and a hydroxyl K. aerogenes enzyme,15 the initial hydrolytic mechanism has
group. Ni1 is also coordinated by two histidine residues and a solvent been elaborated as shown in Fig. 3A. In this model, the urea
molecule, while Ni2 is coordinated by two histidines, an aspartic acid
carbonyl oxygen is suggested to displace a terminal water
residue, and a water molecule. Waters are red, metal-binding side
molecule from the five-coordinate nickel (Ni1), with additional
chains are shown with white carbon atoms, and two nearby histidine
residues that function in catalysis are shown with orange carbon stabilization offered by interaction with His 219 (K. aerogenes
atoms. numbering). The terminal solvent bound to the hexacoordinate
nickel (Ni2) is thought to be appropriately positioned to
attack the carbonyl oxygen, forming a tetrahedral intermediate
metallocenter. X-Ray absorption spectroscopy (XAS) of jack that decomposes with His 320 acting as a general acid.
bean,20,21 K. aerogenes,22 and B. pasteurii23 ureases provides Support for the binding of the urea carbonyl oxygen to Ni1
evidence for 5–6 coordinate nickel ions with exclusively is derived from the structure of acetohydroxamic acid-bound
O/N ligands, including about two imidazoles per metal, enzyme, where this type of interaction is observed.19 This
and the K. aerogenes data include a scattering component simple scheme nicely accounts for the catalytic properties
indicating a Ni–Ni distance of B3.26 Å.22 The presence of of H219A and H320A variants of the protein,31 and this
b-mercaptoethanol leads to thiolate-to-nickel charge-transfer mechanism accommodates the bell-shaped pH dependence of
transitions,24,25 a Ni–S scattering interaction seen by XAS,21,22 the protein if one invokes a reverse-protonation model in
and a unique variable-temperature magnetic circular which activity requires the protonated state of His 320
dichroism feature26 that are all consistent with the inhibitor (pKa B 6.5) and the deprotonated state of the solvent bound
sulfur atom replacing the bridging solvent molecule in the to Ni2 (pKa B 9).33
dinuclear metallocenter. Structural characterization of the An alternative mechanism for urea hydrolysis was proposed
b-mercaptoethanol-bound enzyme directly demonstrates on the basis of a structure determined for phenylphos-
this binding mode.27 Preliminary studies to characterize the phorodiamidate-inhibited urease of B. pasteurii.16 The inhibitor
magnetic properties of the metals in jack bean urease suggest is in fact a slow substrate of the enzyme,34 with release of
weak antiferromagnetic exchange coupling between the nickel phenol leading to the diamidophosphoric acid-bound form of
atoms,28 but saturation magnetization data collected with the enzyme. An amino group of diamidophosphoric acid is
both jack bean and K. aerogenes ureases fail to confirm bound to Ni2, leading the investigators to suggest that binding
this result. of urea displaces both terminal water molecules from the
Many residues nearby or directly coordinating to the dinuclear center to coordinate in a bidentate fashion
K. aerogenes urease dinuclear active site have been substituted (Fig. 3B). Significantly, the structure is consistent with attack
by mutagenesis approaches, and structures are known for of the initial inhibitor by the bridging (not terminal) water
several of these variants.19,29–31 The H134A variant is notable molecule. The bridging solvent molecule is thus proposed to
because removal of the Ni2 ligand results in formation of a attack the urea carbonyl carbon during normal catalysis.
mononuclear Ni1 site.29 Also of interest are studies where the A merged mechanism has also been considered (Fig. 3C). In
lysine residue that becomes carboxylated in the wild-type this case, the bridging water attacks the substrate, but with His
enzyme is substituted. The K217A and K217C variants are 320 continuing to function as a general acid. This model
inactive as isolated, but activity is generated by incubating the readily accommodates the results of mutagenesis studies,31
proteins with nickel ions plus formic acid. Structures of the whereas large changes in the kinetics of His 320 variants fit
chemically rescued enzymes are known.19 less clearly to the model of Fig. 3B. Fluoride inhibition studies
In this review, we discuss the function of nickel in urease of K. aerogenes urease were interpreted in terms of the halogen
catalysis, describe how urease apoprotein can be reconstituted replacing the bridging water molecule to inhibit the enzyme,

This journal is 
c The Royal Society of Chemistry 2009 Metallomics, 2009, 1, 207–221 | 209
Published on 09 April 2009. Downloaded on 10/10/2014 08:35:54. View Article Online

Fig. 3 Proposals for the urease catalytic mechanism. (A) The hydroxyl group bound to Ni2 attacks urea, whose carbonyl group is polarized by
coordination to Ni1, forming a tetrahedral intermediate that releases ammonia with His 320 (K. aerogenes numbering) acting as a general acid. (B)
The bridging hydroxyl group attacks urea, bound with its carbonyl group coordinated to Ni1 and an amine interacting with Ni2, and the hydroxyl
proton transfers to the released ammonia. (C) A merged mechanism in which the bridging water attacks the substrate, but with His 320 acting as a
general acid. (D) Elimination mechanism to form a cyanic acid (OQCQN–H) intermediate that subsequently becomes hydrated (not depicted) to
form carbamate. In all mechanisms, the carbamate spontaneously decomposes.

thus providing support to the hypothesis that the bridging (bdptz) was used to synthesize [Ni2(m-urea)(bdptz)(urea)-
water serves as the nucleophile.35 (CH3CN)](ClO4)3 as a dinuclear urease mimic.38 Heating of
Examination of biomimetic model compounds has provided this compound results in the elimination of ammonia,
further insight into the chemistry available to urease-like followed by slow hydration of the resulting cyanate, leading
dinuclear nickel sites. A wide assortment of such studies has the investigators to suggest that such a sequence of reactions
been reported, only three of which are briefly highlighted here. could also account for the ammonia and carbamate products
In the first example, the ligand N,N,N 0 ,N 0 -tetrakis[(6-methyl- of urease (Fig. 3D).
2-pyridyl)methyl]-1,3-diaminopropan-2-ol (Me4-Htpdp) was To further define the mechanism of urease, high level
used to prepare [Ni2(Me4-tpdp)(CH3CO2)(ClO4)(CH3OH)]ClO4 computational methods have been investigated. For example,
whose structure is known.36 Urea addition leads to displacement shortly after publication of the structure of diamidophosphoric
of ClO4 as the urea carbonyl oxygen coordinates to one acid-inhibited B. pasteurii urease, ab initio and density
nickel, verified by crystallography, with accompanying functional theory approaches were used to explore the most
changes in the absorption spectrum. Of functional significance, likely pathway of urea hydrolysis.39 These studies suggest that
this species reacts with ethanol to generate ethyl carbamate. the urea carbonyl oxygen first coordinates to Ni1, with a urea
For the second representative example, a pyrazolate-based amine binding to Ni2 concomitantly with, or subsequent to,
ligand (HL2) was used to synthesize the crystallographically attack of the bridging hydroxide on the urea carbonyl. Later
characterized L2Ni2(O2H3)(ClO4)2 species.37 The addition of quantum chemical calculations of dinuclear nickel complexes
urea leads to the displacement of H3O2 and bidentate binding are consistent with the bridging ligand being a hydroxide
with the carbonyl oxygen coordinating to one nickel and an rather than an oxo dianion and suggest that urea can bind
amine binding to the second. Heating of this sample leads to in a monodentate or bidentate fashion.40 Computational
the elimination of ammonia and provides a bridging cyanato analysis of the bdptz complex mentioned above led to the
complex. Finally, 1,4-bis(2,2 0 -dipyridylmethyl)phthalazine suggestion that urea may decompose by both hydrolytic and

210 | Metallomics, 2009, 1, 207–221 This journal is 


c The Royal Society of Chemistry 2009
View Article Online

elimination routes.41 Molecular dynamics simulations were demonstrates that about 50% of the protein is stably carboxylated
used to both assess the likely protonation status of His after activation; notably, this value is significantly larger
219 and His 320 and compare various urea decomposition than the percent of enzyme that is active. When urease
pathways.42 More recent quantum chemical studies extend the apoprotein is first incubated with nickel ions and then exposed
examination of various pathways and come to the same to bicarbonate, no activation is observed.45 This finding
conclusion as above that hydrolysis and elimination reactions indicates that CO2 binding must precede binding of Ni. Metal
may compete in the enzyme.43 Several alternative elimination analysis indicates that this nickel-inhibited species and the
pathways were proposed with the various mechanisms protein subjected to optimal activation conditions both
utilizing different general base and general acid groups contain nearly two bound nickel ions per active site even
(Fig. 3D), but all forming cyanate as a product. Significantly, though only 15% of the latter species is active. The improperly
however, cyanate has not been detected during the urease bound nickel ions are more labile than the active metallocenters,
reaction nor is it a substrate of the enzyme (unpublished which are stable to prolonged incubation with EDTA.
observations). It is feasible that cyanate is not released from Extended exposure of the activated protein to EDTA removes
the enzyme, but rather that the metallocenter-bound cyanate is B75% of the bound nickel without affecting activity, and a
immediately hydrated, analogous to the situation reported for second round of activation provides a further increase in the
the bdptz model complex,38 but calculations related to this levels of activity.45,46 XAS studies show the metallocenter
Published on 09 April 2009. Downloaded on 10/10/2014 08:35:54.

additional step have not been reported. properties of the inactive nickel-containing urease and the
active enzyme are nearly indistinguishable.46 The metallocenter
3.0 In vitro reconstitution and properties of structure of the nickel- and bicarbonate-treated, but still
inactive, protein species remains unclear.
metal-substituted ureases
The interaction of urease apoprotein with other metal ions
Of far-reaching significance, the incubation of purified also has been examined. Pre-treatment of urease apoprotein
K. aerogenes urease apoprotein with nickel ions and carbon with zinc, copper, cobalt, or manganese leads to inhibition of
dioxide results in partially activated enzyme.44 The properties the normal CO2 and nickel-dependent activation, whereas
of this in vitro activation process are extensively characterized magnesium and calcium have no effect on this process.45
and have set the stage for comparison to similar studies with In the absence of nickel, incubation of apoprotein with
various urease apoprotein-containing complexes (section 4.0) manganese ions plus bicarbonate leads to activity levels
and for understanding the in vivo mechanism of enzyme comparable to activation of about 2% of the urease active
maturation. Simple addition of nickel ions to urease apoprotein sites.45 In contrast to the situation with nickel, the active
does not generate detectable activity, whereas the use of mangano-urease loses activity when incubated with EDTA.
buffers containing bicarbonate (which is in equilibrium with The EDTA-treated protein still contains B0.4 manganese
CO2 in solution) plus nickel ions yields active urease in per active site and this inactive protein was able to be
approximately 15% of the protein.44,45 CO2 rather than crystallized.46 The structure of this dinuclear manganese
bicarbonate is the critical reagent required for this process as metallocenter is nearly indistinguishable from that of the
shown by using stock bicarbonate buffers at pH 4 or 8.5 active nickel-containing enzyme and the reason for its
during short-term activation experiments (with the same final inactivity is unknown. In contrast to the situation with
pH 8.3 conditions); the low pH stock solution provides more manganese, incubation of apoprotein with cobalt plus
rapid production of active urease because the more acidic bicarbonate yields no activity; however, the electronic
conditions result in higher CO2 concentrations.44 Inclusion of spectrum of this species possesses a feature that could arise
carbonic anhydrase when using the low-pH-bicarbonate stock from a thiolate-to-cobalt charge-transfer transition.46 When
solution decreases the activation rates, further supporting the the same study is carried out using a variant apoprotein
involvement of CO2 in the activation process. Using buffers of (C319A) lacking a cysteine residue near the active site, no
varied pH, all supplied with 0.3% CO2, the extent of activation charge-transfer band is observed and significant levels of
increases with pH consistent with a process involving a group activity (B2% of similarly treated wild-type apoprotein) are
with a pKa of at least 9. These findings led to the suggestion measured. XAS studies are consistent with partial thiolate
that the deprotonated form of an active site residue of high coordination in the sample generated from wild-type
pKa reacts with CO2 to provide a metal-binding ligand needed protein, and this ligand is missing in the C319A mutant. The
to form active enzyme. Elucidation of the urease crystal manganese and cobalt results demonstrate that urease
structure revealed the carboxylated Lys bridging ligand apoprotein can acquire some activity with non-nickel metal
(Fig. 2), in perfect agreement with these results. ions when they are appropriately incorporated into the
Additional studies have characterized the kinetics of the protein. In contrast, zinc- and copper-bound urease are
activation process and the timing of nickel addition. After inactive under all conditions tested.45,46
identifying optimal concentrations of bicarbonate (100 mM) SO2, CS2, and vanadate were investigated for their capacities
and nickel ions (100 mM) for activation at pH 8.3, the process to substitute for CO2 in serving as lysine modification
still requires about 90 min to reach maximum activity.44 These reagents.47 SO2 neither inhibits the normal activation process
results demonstrate the relative inaccessibility of the nascent nor substitutes for CO2. By contrast, CS2 inhibits standard
active site to bicarbonate and metal ions, and they highlight activation and when incubated with nickel ions plus urease
the importance of additional components in generating urease apoprotein it generates a species containing a sulfur-to-nickel
activity during cell growth. Use of radiolabeled bicarbonate charge-transfer transition. These results are interpreted in

This journal is 
c The Royal Society of Chemistry 2009 Metallomics, 2009, 1, 207–221 | 211
View Article Online

terms of formation of a lysine dithiocarbamate-bridged microorganisms, such as Helicobacter mustelae,60 contain a


metallocenter. Remarkably, when urease apoprotein is complete urease gene cluster as well as a second set of
incubated with nickel ions plus vanadate, activity is generated structural genes not associated with any accessory protein
at levels similar to those obtained with bicarbonate.47 These genes. Furthermore, some bacteria lack one or more of the
results suggest the formation of a vanadylated lysine bridging four typical urease auxiliary genes; this is best exemplified by
two nickel ions at the active site. Bacillus subtilis which lacks any identifiable accessory protein
In contrast to the detailed in vitro activation studies with genes.61 The latter result suggests that accessory proteins are
K. aerogenes urease apoprotein, very limited findings are not always required for in vivo urease activation or that
available with other ureases. As one example, nickel-containing genetically unlinked cellular maturation factors are utilized
jack bean urease was dialyzed at room temperature against in some cases. Below, we summarize evidence demonstrating
buffers containing zinc and cobalt for several weeks.48 After the need for four auxiliary genes in most ureolytic bacteria, we
51 days of dialysis versus zinc ions the nickel content of the describe the properties and functions of UreD, UreF, UreG,
protein reduces from 2 to 0.89 per active site, the zinc content and UreE using the framework illustrated in Fig. 5, and we
increases to 0.83 per active site, and the activity drops to 10% discuss evidence for additional genes needed for urease
of the starting value. Similarly, after 87 days of dialysis versus production in selected systems. Unless clearly indicated
Published on 09 April 2009. Downloaded on 10/10/2014 08:35:54.

cobalt ions the nickel content drops to 1.27 per active site, the otherwise, the results described in the following sections involve
cobalt content increases to 0.75 per active site, and the activity analysis of K. aerogenes urease genes expressed in E. coli.
diminishes to 27%. These results were interpreted in terms of
the production of mixed-metal derivatives, but additional
4.2 In vivo demonstration that accessory proteins are required
metallocenter studies (such as XAS) are needed to confirm
this conclusion. More generally, the very lengthy time The significance of the individual accessory proteins in urease
periods involved in these studies raise questions about the activation has been assessed in the K. aerogenes system by
physiological relevance of these metal substitution studies. using a systematic knockout and complementation approach.
Each auxiliary gene was at least partially deleted in the context
4.0 In vivo maturation of bacterial ureases of the otherwise intact ureDABCEFG gene cluster, and the
urease activities in cell extracts were compared to those in cells
Only a portion of purified urease apoprotein acquires activity expressing the wild-type genes [approximately 200 mmol urea
in vitro (B15% in the case of the K. aerogenes protein; degraded min1 (mg protein)1 or B200 U mg1 in cell
section 3.0), even when using optimized activation conditions. extracts].51,62 It was shown that ureD, ureF, and ureG are all
In the bacterial cell, this limitation is overcome by the concerted essential for the production of functional urease since
effort of a series of accessory proteins.49,50 In this section, we mutations in these genes nearly abolish activity in cell extracts
describe how auxiliary genes encoding accessory proteins are (o1 U mg1).51 In these early studies, partial deletions in ureE
often clustered with the structural genes for urease, we detail the resulted in only B50% lower specific activities; however,
properties of each gene product and its associated protein subsequent efforts with a true ureE deletion mutant show that
complexes, and we briefly mention additional genes that affect urease activity is essentially eliminated.63 Furthermore, E. coli
urease activation in some microorganisms. cells expressing only the K. aerogenes structural genes and
grown in the presence of 5 mM NiCl2 possess only trace levels
4.1 Genetic structure of urease-associated genes
(o0.05 U mg1) of urease activity.61 Significantly, purified
Bacterial urease structural genes often are clustered with genes urease from cells expressing the deletion mutants of ureD,
encoding urease-associated proteins, but their number and ureF, and ureG contain negligible amounts of nickel. Urease
order are not universal across species (Fig. 4). For example, activity is largely restored to cells co-expressing the mutant
the three genes encoding urease (ureABC) of K. aerogenes are gene clusters from one plasmid with a wild-type complement
flanked by four genes (ureD positioned upstream and ureE, of the mutant gene on a second plasmid.51 Taken together
ureF, and ureG found downstream) encoding accessory these data indicate that each of the four K. aerogenes accessory
proteins, each of which facilitates urease activation.51,52 This proteins plays an essential role in urease maturation.
urease gene structure is retained in Rhizobium leguminosarum, Genetic experiments using deletion, insertional inactivation,
but with several uncharacterized open reading frames (ORFs) and complementation approaches have been used to identify
inserted into this cluster.53 Many bacteria position ureD after multiple non-urease-subunit genes required for urease activity
ureG, as shown for thermophilic Bacillus sp. TB-90 which in Proteus mirabilis,64 Klebsiella pneumoniae,65 Providencia
also contains the downstream ureH (likely encoding a stuartii,66 and many other bacteria, including the peptic-ulcer
nickel ion permease) and another ORF.54 In Actinobacillus causing H. pylori. In the latter case, disruptions or deletions in
pleuropneumoniae, the ureABCEFGD cluster is interrupted by ureA, ureB, ureF, ureG, or ureH of the H. pylori urease genes
an ORF and is preceded by a gene cluster encoding a five- expressed in E. coli result in a non-ureolytic phenotype.59
component nickel transporter and a possible urea permease.55 Subsequent studies confirm these results and further show a
The H. pylori urease cluster consists of ureA, a fusion of the significant reduction in urease activity in cells lacking
small subunit genes from other bacteria, and ureB, encoding ureE.67,68 Detailed investigations of the four standard
the large subunit,56,57 along with five downstream genes: ureI accessory proteins are discussed in sections 4.3–4.6, and the
(encoding a proton-gated urea channel),58 ureE, ureF, ureG, products of additional genes found in selected microorganisms
and ureH (homologous to ureD of other bacteria).59 Some are summarized in section 4.7.

212 | Metallomics, 2009, 1, 207–221 This journal is 


c The Royal Society of Chemistry 2009
Published on 09 April 2009. Downloaded on 10/10/2014 08:35:54. View Article Online

Fig. 4 Organization of representative urease gene clusters. The urease gene cluster of K. aerogenes (ureDABCEFG) is compared to the gene
organization found in selected other bacteria: R. leguminosarum, containing insertions of unidentified genes; Bacillus sp. TB-90, which repositions
ureD and adds a likely nickel permease (ureH); A. pleuropneumoniae, which juxtaposes genes encoding a likely nickel transport (cbiKLMO) and
urea permease (utp); Helicobacter species which fuse the two small structural subunits and add a proton-gated urea channel (ureI); H. mustelae
containing a second, incomplete urease cluster; and B. subtilis that lacks urease accessory genes. Genes encoding urease subunits are shown in blue,
the ureE gene encoding a metallochaperone is green, other urease accessory genes are purple, genes encoding proteins involved in nickel uptake are
orange, those encoding proteins related to urea transport are red, and unknown genes are white. The sizes of the arrows do not accurately reflect
the sizes of the genes.

4.3 UreD and the UreABC–UreD complex Following activation, UreD dissociates from the structural
subunits. Taken together these data indicate that UreD plays
Attempts to heterologously express K. aerogenes ureD in
an important, but still undefined, role in incorporating nickel
E. coli lead to the production of inclusion bodies,69 and the
into the urease active site.
resulting absence of a soluble protein prevents characterization
Corroborating the results observed in the K. aerogenes
of the general protein properties. Recently, however, a maltose
system, yeast two-hybrid studies of H. pylori proteins identify
binding protein (MBP)-UreD fusion protein was found to be
interactions between the corresponding UreH and UreA
soluble and its characterization is in progress (Carter and
proteins.67,72 Furthermore, immunoprecipitation of UreC
Hausinger, unpublished observations). No purification of
from E. coli cell extracts expressing the P. mirabilis urease
UreD from another bacterium is reported, and no structure
genes leads to coprecipitation of UreD, consistent with an
of UreD is available. In contrast to the dearth of information
interaction between these two proteins.73
about isolated UreD, the properties of several protein
complexes containing this accessory protein (Fig. 5) are
4.4 UreF and the UreABC–UreDF complex
known, as discussed below.
When ureD is highly expressed along with the genes Similar to the situation for ureD, heterologous expression of
encoding the urease subunits, a UreABC–UreD protein K. aerogenes ureF leads to an insoluble protein.74 In this case,
complex can be purified.69 This complex is known by comparison however, two soluble forms of UreF have been described: an
of native and denaturing gel electrophoresis to be comprised of MBP–UreF fusion75 and a UreE–UreF fusion.76 The general
four species, where the (UreABC)3 apoprotein binds to properties of the MBP–UreF fusion protein and its capacity to
0–3 molecules of UreD. Results from chemical cross-linking interact with other urease proteins are unknown. In contrast,
studies indicate that UreD is in close proximity to both UreB the monomeric UreE–UreF fusion protein is capable of
and UreC,70 and small angle X-ray absorption (SAXS) studies facilitating urease activation in the cell, it binds to the
confirm the binding of UreD near UreB in the UreABC–UreD UreABC–UreD complex in vitro, and it forms an even larger
complex.71 When the K. aerogenes UreABC–UreD complex is complex in vivo.76 No UreF protein has been purified from
subjected to the in vitro activation conditions described any other microorganism, and no structure is reported.
in section 3, about 30% of the nascent active sites are Nevertheless, a homology model was created for a portion
activated,49,69 demonstrating that UreD significantly enhances of UreF from B. pasteurii by using two GTPase activating
the activation competence over that of the apo-protein alone. enzymes as templates, and a similar role is hypothesized for

This journal is 
c The Royal Society of Chemistry 2009 Metallomics, 2009, 1, 207–221 | 213
View Article Online

fragments.70 No UreD cross-links are observed in the


UreABC–UreDF complex whereas distinct cross-links are
detected in the UreABC–UreD species. Further analysis by
this cross-linking/proteolysis/mass spectrometry approach
provides evidence for an altered conformation of the
UreABC–UreDF complex in comparison to UreABC–UreD
or urease apoprotein. In particular, a shift of UreB in a
hinge-like motion is hypothesized in order to account for
cross-linking of a lysine residue in UreB with another lysine
in a distant portion of UreC (located over 50 Å away in the
urease apoprotein). Compatible with this proposal, flexibility
analysis of urease demonstrates that UreB can reasonably shift
to enhance access to the nascent active site.71 SAXS studies
lack sufficient resolution to address the presence of a shifted
UreB domain, but the modeled structure derived from these
Published on 09 April 2009. Downloaded on 10/10/2014 08:35:54.

data places UreD and UreF together in close contact


with UreB.71 In aggregate, these results are consistent
with UreABC–UreDF being capable of undergoing a
conformational change that enhances its competence for
insertion of bicarbonate and Ni into the buried active site by
increasing its exposure. UreABC–UreDF is similar to the
UreABC–UreD complex in allowing B30% of the available
protein to be activated using standard conditions; however,
the UreABC–UreDF complex requires lower concentrations
of bicarbonate for activation because it is less susceptible to
nickel-dependent inactivation. The protection against nickel
inhibition suggests a role for UreF in regulating the sequential
incorporation of bicarbonate before nickel into the active site.
Although UreABC–UreDF complexes have not been
studied from other microorganisms, UreD–UreF interactions
Fig. 5 Postulated interactions among urease-related bacterial have been noted. For example, yeast two-hybrid analysis
proteins. The urease apoprotein sequentially binds UreD, UreF, and reveal the association of UreD with UreF in P. mirabilis.73
UreG, and the in vitro activation properties exhibit differences in each This same approach demonstrates an interaction between
apoprotein species. Alternatively, a preformed UreDFG complex may UreF and UreH, an orthologue of UreD, in the H. pylori
bind the apoprotein. Within the resulting complex, the UreDFG system.67,72
heterotrimer acts as GTP-dependent molecular chaperone to
enhance exposure of the nascent active site. UreE interacts with the
UreABC-UreDFG complex and delivers nickel ions, thus serving a 4.5 UreG and its complexes
metallochaperone role. Carbon dioxide is used to form the
carboxy-lysine metal ligand, and GTP hydrolysis (occurring in UreG) UreG is a soluble protein that has been purified and
drives the metallocenter assembly process to provide active urease, characterized from several microorganisms including
with release of all accessory proteins. K. aerogenes,78 B. pasteurii,79 M. tuberculosis,80 and H. pylori.81
In contrast to the monomeric protein from K. aerogenes, UreG
proteins from B. pasteurii and M. tuberculosis are dimeric with
this protein.77 Like UreD, UreF is a component of a the two monomers said to be joined through a disulfide bridge,
number of urease-related protein complexes (Fig. 5), as and that from H. pylori undergoes zinc-dependent (but not
described below. nickel-dependent) dimerization. The metal-binding properties
Over-expression of ureF and ureD along with ureABC leads of UreG proteins from B. pasteurii and H. pylori are
to the production of a UreABC–UreDF complex.74 In characterized; the former binds two zinc and four nickel ions
contrast, no complex is generated by expressing just ureF with per dimer79 and the latter binds 0.5 zinc and two nickel ions
ureABC. The purified UreABC–UreDF complex is a mixture per monomer.81 Two residues suspected to participate in
of species containing 0–3 UreDF heterodimers per (UreABC)3 binding zinc (Cys 66 and His 68) were mutated individually
apoprotein according to Western blot analysis of native and together; the variant H. pylori UreG proteins bind zinc
gels using anti-urease antibodies.74 Significantly, anti-UreD with a 10-fold larger Kd than wild-type protein and continues
antibodies fail to detect UreABC–UreDF complexes, but do to dimerize, but curiously the double mutant protein binds two
recognize UreABC–UreD, thus suggesting that UreF masks zinc per dimer.81 The ureG sequences from these and other
the UreD epitopes by directly binding to UreD. Support bacteria reveal a nucleotide binding motif (P-loop) consistent
for this proposal is derived from a study involving with a GTPase role, but only very low or no hydrolysis of GTP
chemical cross-linking in conjunction with matrix-associated is detected. Mutation of a single residue in this motif for either
laser desorption-ionization mass spectrometry of proteolytic the K. aerogenes or H. pylori protein leads to the abolishment

214 | Metallomics, 2009, 1, 207–221 This journal is 


c The Royal Society of Chemistry 2009
View Article Online

of urease activity.78,82 The B. pasteurii protein is claimed to be truncated version (H144*UreE) lacking this region still binds
intrinsically disordered on the basis of NMR dispersion and B2 Ni2+ atoms per homo-dimer.92 For both proteins,
fluorescence analysis;79,83 however, circular dichroism studies copper, cobalt, and zinc compete with nickel ions, and the
show substantial secondary structure in some members of this spectroscopic properties of these proteins are known.93 For
protein family. No structure is reported for any UreG, but one cells grown in the presence of nickel and expressing a urease
has been elucidated for the related protein HypB (required cluster with H144* UreE substituting for the wild-type
for biosynthesis of nickel-dependent hydrogenases) from version, active urease is produced, indicating that the
Methanocaldococcus jannaschii.84 This dimeric protein contains C-terminus of wild-type UreE is unnecessary for urease
a GTP binding site and a dinuclear zinc site at the subunit activation.91,94 Additional mutagenesis studies suggest that
interface. Two of the residues serving as ligands to the HypB His 110 and His 112 participate in binding nickel but are not
zinc site coincide with the UreG residues that had been critical to urease activation, whereas His 96 has a role
mutated in the H. pylori protein. in binding the essential metal for transfer to urease.89,94
A UreABC–UreDFG complex (Fig. 5) was identified in Isothermal titration calorimetry studies of the interaction of
E. coli cells expressing a wild-type K. aerogenes urease cluster nickel, copper, and zinc with H144* UreE demonstrate
in the absence of nickel ions.85 UreABC–UreDFG is also remarkable complexity, including a protein concentration
dependence in these thermodynamic interactions.95 At low
Published on 09 April 2009. Downloaded on 10/10/2014 08:35:54.

obtained in vitro by mixing purified UreG with the isolated


UreABC–UreDF complex.86 When activated in the standard protein concentrations, two metal ions bind per dimeric
buffer containing high levels of nickel and bicarbonate, over protein; whereas, at high protein concentrations three nickel
60% of the urease apoprotein in the complex is activated, or copper ions are initially bound per dimer followed by the
leading to the notion that UreDFG functions as a binding of two additional metal ions. The thermodynamics of
urease-specific molecular chaperone.87 Importantly, the nickel ion binding to K. aerogenes H144* UreE is enthalpically
inclusion of GTP, but not other nucleotide tri-phosphates, favored.
results in significant activity when incubated with physiologically The structure of the H144* variant of K. aerogenes UreE
relevant bicarbonate levels, compared to the trace activity dimeric apoprotein is depicted in Fig. 6.96 While the
levels observed in a similar reactions without GTP. Hydrolysis nickel-bound form of the protein is not structurally characterized,
of GTP, and not just binding, is needed for activation as the structure of the copper-bound species is known and the
shown by using a non-hydrolyzable GTP analogue. The site of protein binds three copper ions. One copper binds at the
GTP action is within UreG as shown by studies of the subunit interface via His 96 side chains from each subunit
UreABC–UreDFG complex containing a mutation in the (encircled in red). On the basis of mutational studies, this site
P-loop motif of UreG; this complex exhibits little competence is thought to be critical for nickel ion binding related to urease
for activation.86 A hint of a comparable UreG- and activation.89 Two additional copper ions bind to periphery
urease-containing complex in H. pylori is derived from work sites (encircled in blue) via His 110 and His 112 side
in which UreG purified by use of a tandem affinity tag chains. These sites also bind nickel, but mutational studies
possesses substoichiometric UreB.88 demonstrate these residues are not essential for urease
In addition to being a component of the UreABC–UreDFG activation.89 The peripheral sites are suggested to assist in
complex, UreG forms an insoluble, urease-free, UreDFG nickel delivery to the central site. Likewise, the His-rich
complex (Fig. 5) in cells expressing ureD, ureF, and ureG C-terminus of the full-length protein would be positioned well
and lacking ureE or the structural subunit genes.78 This for feeding nickel ions into the interfacial site. The protein
heterotrimer is solubilized by using low concentrations of domain responsible for binding metal ions resembles a
detergent (0.5% Triton X-100), allowing for isolation via a ferredoxin fold, which also is found in the copper metallo-
combination of anion-exchange chromatography, detergent chaperone Atx1.97 In addition to the metal-binding domain of
removal, and use of an ATP-linked agarose column. A the protein, UreE contains a second domain (shown in purple
UreDFG complex containing a P-loop variant of UreG fails brackets) that resembles a peptide-binding domain of yeast
to bind to the nucleotide resin, indicating that an intact P-loop Hsp40 or Sis1.98 To test whether this domain might function in
is necessary for this interaction. It is unknown whether peptide binding as part of the nickel delivery process, mutants
pre-formed UreDFG interacts with urease apoprotein, and it in this region of the protein were examined. Notably, the loss
is unclear whether this complex is physiologically relevant. of this entire domain along with the C-terminus fails to
Nevertheless, the preformed UreDFG species potentially eliminate its ability to function in urease activation.63
could function as a stable unit during urease activation as an UreE proteins have been purified and characterized to
alternative to the sequential binding of UreD, UreF, and various extents from several other bacteria. P. mirabilis UreE
UreG to urease apoprotein (Fig. 5). contains a shorter His-rich C-terminus (HHHHDHHH) and
also is dimeric.99 The H. pylori UreE protein, which lacks a
His-rich region at its C-terminus, binds one nickel ion per
4.6 UreE and the UreABC–UreDFG–UreE complex
dimer.100 More extensive investigations have been carried out
The metallochaperone UreE (Fig. 5) delivers nickel to the on UreE of B. pasteurii, which binds two nickel ions per
UreABC–UreDFG complex.87,89,90 This protein has been dimer.101–103 Significantly, the crystal structure of the
extensively studied in K. aerogenes where it binds six nickel zinc-bound protein is known,104 but like K. aerogenes UreE
ions per homo-dimer.91 Most of this nickel is bound to the the nickel-bound structure is not available. The overall fold
histidine-rich C-terminus (HGHHHAHHDHHAHSH), but a of B. pasteurii UreE is essentially identical to that from

This journal is 
c The Royal Society of Chemistry 2009 Metallomics, 2009, 1, 207–221 | 215
Published on 09 April 2009. Downloaded on 10/10/2014 08:35:54. View Article Online

Fig. 6 Structure of UreE. The structure of homodimeric K. aerogenes H144* UreE (PDB access code 1gmw), a truncated protein missing the
C-terminal His-rich sequence, is depicted without metal ions. The central metal binding site (encircled in red) occurs at the subunit interface site
and binds two nickel ions using, in part, His 96 residues from each subunit. Auxiliary metal binding sites (encircled in blue), involving the
non-essential His 110 and His 112 residues, are presumed to donate nickel ions to the central site. When present, the C-terminal extension is also
well positioned to provide nickel ions to the interfacial site. In addition to the metal-binding domain, a ‘‘peptide-binding domain’’ is present
(in brackets) but is not essential for UreE function.

K. aerogenes, shown in Fig. 6. The B. pasteurii protein lacks when grown in Luria-Bertani medium, whereas cells containing
residues corresponding to His 110 and His 112, and thus the only the urease genes require supplementation by nickel ions
periphery sites are absent; however, the residue corresponding to achieve full urease activity.55 Yersinia pseudotuberculosis
to His 96 of K. aerogenes is conserved and this protein binds a also possesses a five-gene cluster (yntABCDE) located 5’ of the
single zinc atom at this interface site. Calorimetry studies urease genes, and deletion of this region within the endogenous
provide confirmatory evidence that two nickel ions bind to host abolishes urease activity and reduces the rates of nickel
this protein in an entropically-driven process (differing from uptake by the microorganism.106 Y. pseudotuberculosis also
the situation with K. aerogenes UreE),95 and results from XAS contains a gene, ureH located 3’ of the urease genes, that is
studies are compatible with Ni–Ni scattering from metal ions related in sequence to nickel-cobalt transporter proteins;
separated by 3.4 Å.102 deletion of ureH leads to diminished rates of nickel uptake,
The interaction of UreE with the UreABC–UreDFG com- confirming a function in nickel transport.106 In a similar
plex is of great interest as this most likely represents the manner, Streptococcus salivarius 57.I contains three genes
physiologically relevant activation complex that produces fully (ureMQO) immediately downstream of the urease gene cluster,
active urease. Significantly, incubation of UreABC–UreDFG and insertional inactivation of ureM leads to the inability to
plus purified UreE with physiologically relevant concentrations accumulate nickel ions and the consequent loss of urease
of bicarbonate, nickel, and GTP activates the urease apoprotein activity.107
to wild-type levels.105 E. coli cells expressing an intact Urease activation in H. pylori and related bacteria is further
K. aerogenes urease gene cluster encoding a UreB variant complicated by the involvement of several additional genes
(involving the proposed hinge region mentioned above) exhibit that are not adjacent to the urease gene cluster. For example,
deficiencies in urease activation and appear to have trapped the nixA gene encodes a nickel ion transporter according to
some of the urease apoprotein in a UreABC–UreDFG–UreE uptake studies in recombinant E. coli cells containing this
complex.71 UreE also directly interacts with UreG according gene, and its presence along with the H. pylori urease genes
to analyses of the H. pylori system using yeast two-hybrid significantly enhances urease activity in this heterologous
approaches.67,72 host.108 Furthermore, mutagenesis of nixA in H. pylori reduces
nickel ion transport and decreases urease activity.109 In addition
4.7 Other accessory proteins
to NixA, H. pylori contains the AbcABCD nickel ABC-type
Superimposed on the already complex urease activation transporter.171 This microorganism also possesses an outer
scheme shown in Fig. 5, some bacteria possess additional membrane transporter, energized by the TonB/ExbB/ExbD
accessory proteins encoded within their urease gene clusters. complex, that takes up nickel and enhances urease
As mentioned in section 4.1, genes encoding a proton-gated activity.110,111 Also of note are three cytoplasmic nickel-binding
urea channel58 and suspected nickel ion permeases or proteins: HspA, Hpn, and the Hpn-like protein. HspA is a
multi-component, nickel-specific, ATP-binding cassette-type GroES homologue containing a 27-residue C-terminal extension
transporters have been reported.54,55 Consistent with a that is rich in His and Cys residues. In E. coli cells containing
function in nickel transport, E. coli cells containing the the H. pylori urease gene cluster, co-expression of hspA
putative A. pleuropneumoniae transporter genes (cbiKLMQ) stimulates the level of urease activity about four-fold.112,113
along with this organism’s urease genes are urease positive The extra domain binds two nickel ions and provides some

216 | Metallomics, 2009, 1, 207–221 This journal is 


c The Royal Society of Chemistry 2009
View Article Online

protection against toxic levels of this metal, and it also binds a His-rich N-terminus that is not found in bacterial UreG
two bismuth ions, a component of several anti-ulcer sequences. The fungal UreF protein of S. pombe shares only
drugs.113,114 The Hpn protein is a 60-residue peptide containing 20% sequence identity with the soybean protein, but the plant
28 His residues.115 Deletion of the gene encoding Hpn within gene rescues the corresponding yeast mutant, whereas this
H. pylori either has no effect on urease activity115 or actually situation is not observed for UreD where the proteins share
increases urease activity.116 This metal-binding protein117 is 30% sequence identity.134
hypothesized to play a role in nickel storage that could be Plants can possess multiple urease isozymes as well as genes
utilized for nickel donation while also functioning in metal encoding several maturation proteins. The historically
detoxification.116,118 The Hpn-like protein, a 75-residue interesting jack bean (C. ensiformis) system includes two
peptide containing 19 His and 30 Gln, binds two nickel ions structural urease genes,135,136 but no accessory genes have
per monomer.119 Deletion of the gene leads to an increase in yet been reported. The better-studied soybean (Glycine max)
urease activity while also increasing the sensitivity of cells to system also has two urease isozymes: an embryo-specific form
nickel, cobalt, and cadmium toxicity.116 encoded at the Eu1 locus137,138 and a ubiquitously-expressed
Of greater interest to urease activation are two genes, hypA species encoded at the Eu4 locus.139,140 In addition, this plant
and hypB typically associated with hydrogenase biosynthesis contains several demonstrated urease accessory genes. For
Published on 09 April 2009. Downloaded on 10/10/2014 08:35:54.

in bacteria, that are required for full urease activity in H. pylori example, soybean UreD and UreF are orthologues of the
and H. hepaticus.120,121 H. pylori contains a full complement of bacterial and fungal genes; in the latter case, the plant gene
structural and accessory genes for the production of a [NiFe] complements a mutant involving the corresponding gene in
hydrogenase, an enzyme that serves as an energy source for S. pombe.134 The Eu3 locus was long known to exhibit
this bacterium.122 HypA is a nickel-binding protein that pleiotropic effects on both ureases,141 and more recently was
is proposed to be the nickel chaperone for hydrogenase shown to encode a protein related to bacterial UreG proteins
maturation.123 In contrast, H. pylori HypB does not bind with a conserved P-loop motif and an added His-rich
nickel, but this dimeric protein, which is homologous to UreG, N-terminus that resembles the C-terminus of some bacterial
possesses weak GTPase activity needed for hydrogenase UreE proteins and is probably involved in nickel binding.142
activation.123 Furthermore, chemical cross-linking studies Eu2 encodes another protein necessary for activation of both
demonstrate that HypA and HypB can form a heterodimer. ureases,141 but its sequence and function remain unknown.
In both Helicobacter species mentioned above, mutations in Homologues encoding UreD-, UreF-, and UreG-like proteins
hypA and hypB nearly abolish urease and hydrogenase are now known to exist in many other plant species, including
activity.120,121 The mechanism responsible for the dual tomato (Lycopersicon esculentum), potato (Solanum tuberosum),
functioning of these activation proteins remains unclear, and and Arabidopsis thaliana.134,143,144 Of interest, potato UreG
neither HypA nor HypB are needed for heterologous complements a K. aerogenes ureG mutation143 and insertions
activation of H. pylori urease in E. coli.124 A possible hint of into each of the three accessory genes of A. thaliana abolished
the site of interaction is the HypA–UreE heterodimer complex urease activity.144
captured by chemical cross-linking studies, consistent with
an interaction between these two proteins in the cell.125 6.0 Regulation of urease by metal ions or other
Furthermore, tandem affinity purification experiments
effectors
demonstrate interactions between HypB and UreG as well as
HypB and urease.88 Further efforts are needed to define the Ureolytic organisms utilize a wide variety of regulatory
interplay of these two activation systems in this microorganism. mechanisms to control urease. In bacterial pathogens that
infect the urinary tract, such as P. mirabilis or P. stuartii, the
AraC-like transcriptional activator UreR binds urea as an
5.0 In vivo maturation of eukaryotic ureases
effector molecule and induces urease expression.145,146 A
As detailed in the preceding section 4.0, the mechanism of pH-dependent regulatory mechanism operates through
urease activation has been extensively investigated in the case cis-acting elements in the dental plaque microorganism
of bacterial ureases; however, many fungi and plants also S. salivarius.147,148 In some environmental isolates that utilize
possess this enzyme which must undergo analogous activation urea as a nitrogen source, the NtrC transcriptional activator
processes. Genetic and biochemical studies related to urease binds to the urease promoter and allows for nitrogen-
activation in eukaryotes are summarized below. dependent regulation of urease.149 A cascade version of this
Fungal ureases contain a single type of subunit, but genetic mechanism is utilized by K. aerogenes which uses NtrC to
studies reveal that multiple genes are needed for urease expres- control levels of the nitrogen assimilation control protein that
sion. For example, early studies with Neurospora crassa,126–128 then directly regulates urease expression.150,151 Three factors,
Aspergillus nidulans,129,130 and Schizosaccharomyces pombe,131 CodY, GlnR, and Spo0H, all regulate urease expression in
show that four distinct loci are required for obtaining active response to nitrogen availability in the spore-forming
enzyme. The best studied fungal urease system is that in the bacterium B. subtilis.152 In contrast, urease of B. pasteurii
fission yeast S. pombe where the enzyme was purified,132 appears to be nearly constitutively expressed at high levels.153
the structural gene (ure2) identified,133 and candidate genes Additional levels of complexity are known in eukaryotes. As
encoding UreD (ure4), UreF (ure3), and UreG (ure1) were an illustration, the algal partner in the lichen Evernia prunastri
identified.134 Of interest, the fungal UreG has 61% sequence is suggested to produce a protein that inhibits urease synthesis
identity to soybean UreG (see below) including the presence of in the associated fungus.154 As another example, soybean

This journal is 
c The Royal Society of Chemistry 2009 Metallomics, 2009, 1, 207–221 | 217
View Article Online

regulates its isozymes in a temporal and tissue-specific a nikR/ureB/ureG mutant possesses the same activity levels,
manner.139 Cucumber leaf urease is reported to be induced suggesting that accessory proteins are not required for
by cobalt ions, although the molecular basis of this regulation synthesis of an active form of the second urease. Similarly,
is unknown.155 These and other regulatory mechanisms will hypB was shown not to be required for this activity. In
not be further described here; rather, this section focuses on contrast to the stability observed for most ureases, the activity
metal-dependent regulation of bacterial ureases. associated with UreA2B2 is lost upon disruption of the cells.
The clearest example of urease expression being regulated These results led the researchers to put forth the intriguing
by metal ions has been described in H. pylori. In particular, hypothesis that the second urease may be an oxygen-sensitive,
both Northern hybridization studies and analysis of iron-containing urease. Congruent with this hypothesis,
b-galactosidase activity in a ureA::lacZ transcriptional fusion these Helicobacter species are noted as being associated with
show that supplementation of brucella growth medium with carnivores whose food source is depleted in nickel, but rich in
nickel ions leads to an increase (up to 3.5-fold) in transcription iron. Studies of the second enzyme from H. mustelae are under
of the urease genes.156 Notably, the effects are not observed way in an effort to characterize this potential iron-dependent
upon addition of cadmium, cobalt, copper, iron, manganese, urease.
or zinc, but are specific to nickel. This same report
Published on 09 April 2009. Downloaded on 10/10/2014 08:35:54.

demonstrates a two-fold effect of a fur mutant (lacking a


7.0 Perspectives
functional ferric uptake regulator) in the nickel-supplemented
cells, suggesting that Fur also modulates the levels of urease. Despite extensive efforts to understand the catalysis, nickel
Additional evidence supporting nickel-dependent regulation dependence, and molecular activation mechanism of urease,
of urease activity in H. pylori is derived from studies of a nikR many fundamental questions remain unanswered.
mutant, defective in the corresponding nickel regulatory Structural, biochemical, and computational data provide
protein.157 The mutant cells lack the ability to increase urease us with a wealth of detail about the urease active site;
expression in the presence of nickel. The same phenotype is nevertheless, as illustrated in Fig. 3, the precise reactions
noted when a 19-bp palindromic sequence just upstream of the leading to the final release of carbamic acid and ammonia
urease operon is removed. Furthermore, the nikR mutant cells are unclear. The chemistries inherent to these reactions are not
are growth inhibited by nickel ions at concentrations greater restricted to nickel, yet all ureases characterized to date
than 100 mM whereas the wild-type cells tolerate high possess nickel. Do non-nickel forms of this enzyme exist in
concentrations of nickel ions. These results are consistent with nature? The generation of partially active urease by treatment
nickel-bound NikR acting as an activator of urease and a of apoprotein with bicarbonate plus manganese or cobalt (for
repressor of nickel transport. These findings were extended to C319A protein) lends support to this idea. In addition, the
demonstrate that metal-bound NikR activates or represses discovery that ureA2B2 of H. mustelae is inversely regulated
several genes (ureA, nixA, copA2, hpn, and hpn-like, several by nickel and stimulated by iron further supports this notion.
of which are known to encode proteins that bind nickel ions) The definitive roles of the UreD, UreE, UreF, and UreG
while repressing a large number of other genes that include accessory proteins during urease maturation remain elusive.
nickel uptake factors and fur.158,159,162,172 Crystallographic We discussed earlier the notion that UreDFG, perhaps
studies have elucidated the structure of H. pylori NikR in pre-formed, might serve as a GTP-dependent molecular
the apoprotein and nickel-bound states.160 An elegant model chaperone while UreE functions as a metallochaperone, but
was described for the pH-dependent modulation of the this proposal demands further verification. Using MBP-UreD
NikR-dependent regulation, accounting for acid adaptation or other soluble forms of UreD, we anticipate that biochem-
of H. pylori.161 Additional studies by several laboratories have ical and structural analyses will be carried out on this yet
characterized the binding of nickel to NikR and its interaction uncharacterized protein. Such studies, combined with follow-
with the urease promoter in this microorganism.162–169 up investigations of UreABC–UreD, may shed light on the
Studies carried out with three other species of Helicobacter ability of ureD to increase the in vitro activation competence of
(i.e., H. mustelae, H. acinonychis, and H. felis) highlight the bound apoprotein. Additional biochemical and structural
another intriguing aspect of metal-dependent urease studies will be performed with the UreEF fusion protein
regulation. These microorganisms possess two sets of urease and other soluble forms of UreF, as well as with the
genes, one which contains the complete urease gene cluster and UreABC–UreDF complex. This effort should provide new
one containing only a second copy of the structural genes insight into the structure of UreF and its function in the
(termed ureA2 and ureB2).60,170 In each case, the second copy maturation process. The postulated similarity of UreF to
of each subunit is closely related (more than 50% identical) to known GTPase activating factors is intriguing as this protein
the first copy. In the best studied case of H. mustelae, may act in concert with UreG (a known GTPase) in the
quantitative RT-PCR was used to show that ureAB is induced UreABC–UreDFG activation complex to couple the energy
by nickel ions whereas ureA2B2 is inversely regulated by nickel of GTP hydrolysis for efficient urease activation. The structure
and increases with iron supplementation.60 Immunological of UreG still needs to be determined, its metal-binding
results confirm this inverse regulation of the two proteins in properties more clearly examined, and its interaction with
H. mustelae and demonstrate the same situation exists in the other urease components investigated at greater resolution.
other two species. A nikR/ureB mutant exhibits small amounts Our current model designates UreE as responsible for nickel
of urease activity, consistent with ureA2B2 encoding a delivery; however, the mechanism of delivery is unknown. For
functional enzyme that does not utilize nickel ions. Furthermore, example, is nickel transferred directly from UreE to the

218 | Metallomics, 2009, 1, 207–221 This journal is 


c The Royal Society of Chemistry 2009
View Article Online

structural subunits or do other components function as 18 L. Sheridan, C. M. Wilmont, K. D. Cromie, P. van der Logt and
intermediaries in this process? How GTP hydrolysis drives S. E. V. Phillips, Acta Crystallogr., Sect. D, 2002, D58,
374–376.
this process is also uncertain. 19 M. A. Pearson, L. O. Michel, R. P. Hausinger and P. A. Karplus,
The findings that additional non-contiguous genes are often Biochemistry, 1997, 36, 8164–8172.
encoded near urease gene clusters and the recent discovery that 20 L. Alagna, S. S. Hasnain, B. Piggott and D. J. Williams, Biochem.
hydrogenase maturation factors affect urease activation in at J., 1984, 220, 591–595.
21 P. A. Clark, D. E. Wilcox and R. A. Scott, Inorg. Chem., 1990, 29,
least two strains of Helicobacter are also of interest. The 579–581.
presence of urea and nickel transport/assimilation systems 22 S. Wang, M. H. Lee, R. P. Hausinger, P. A. Clark, D. E. Wilcox
linked to urease activity in other organisms and the lack of and R. A. Scott, Inorg. Chem., 1994, 33, 1589–1593.
23 S. Benini, S. Ciurli, H. F. Nolting and S. Mangani, Eur. J.
accessory proteins in B. subtilis demonstrate that urease Biochem., 1996, 239, 61–66.
maturation may be influenced by other cellular factors. 24 R. L. Blakeley, N. E. Dixon and B. Zerner, Biochim. Biophys.
The convergence of two maturation pathways may increase Acta, 1983, 744, 219–229.
the efficiency of metallocenter assembly by coordinating the 25 M. J. Todd and R. P. Hausinger, J. Biol. Chem., 1989, 264,
15835–15842.
delivery of nickel (using HypA and UreE) or facilitating the 26 M. G. Finnegan, A. Kowal, M. T. Werth, P. A. Clark,
nucleotide dependence (using HypB and UreG). D. E. Wilcox and M. K. Johnson, J. Am. Chem. Soc., 1991,
Published on 09 April 2009. Downloaded on 10/10/2014 08:35:54.

Relatively little is known about urease maturation in 113, 4030–4032.


eukaryotic systems. Parallels between the fungal and plant 27 S. Benini, W. R. Rypniewski, K. S. Wilson, S. Ciurli and
S. Mangani, J. Biol. Inorg. Chem., 1998, 3, 268–273.
activation components with the bacterial systems need to be 28 P. A. Clark and D. E. Wilcox, Inorg. Chem., 1989, 28, 1326–1333.
further explored, and novel features, such as the product of 29 I.-S. Park, L. O. Michel, M. A. Pearson, E. Jabri, P. A. Karplus,
Eu2 in soybean, require additional investigation. Studies to S. Wang, J. Dong, R. A. Scott, B. P. Koehler, M. K. Johnson and
R. P. Hausinger, J. Biol. Chem., 1996, 271, 18632–18637.
uncover cellular urea and nickel homeostasis systems and to 30 M. A. Pearson, R. A. Schaller, L. O. Michel, P. A. Karplus and
explore novel urease maturation pathways in both eukaryotes R. P. Hausinger, Biochemistry, 1998, 37, 6214–6220.
and prokaryotes will surely lead to new discoveries in urease 31 M. A. Pearson, I.-S. Park, R. A. Schaller, L. O. Michel,
biology. P. A. Karplus and R. P. Hausinger, Biochemistry, 2000, 39,
8575–8584.
32 N. E. Dixon, P. W. Riddles, C. Gazzola, R. L. Blakeley and
B. Zerner, Can. J. Biochem., 1980, 58, 1335–1344.
Acknowledgements 33 P. A. Karplus, M. A. Pearson and R. P. Hausinger, Acc. Chem.
Res., 1997, 30, 330–337.
Studies related to this topic in the Hausinger laboratory are 34 R. K. Andrews, A. Dexter, R. L. Blakeley and B. Zerner, J. Am.
supported by NIH grant DK04586. Chem. Soc., 1986, 108, 7124–7125.
35 M. J. Todd and R. P. Hausinger, Biochemistry, 2000, 39,
5389–5396.
References 36 K. Yamaguchi, S. Koshino, F. Akagi, M. Suzuki, A. Uehara and
S. Suzuki, J. Am. Chem. Soc., 1997, 119, 5752–5753.
1 J. B. Sumner, J. Biol. Chem., 1926, 69, 435–441. 37 F. Meyer, E. Kaifer, P. Kircher, K. Heinze and H. Pritzkow,
2 N. E. Dixon, C. Gazzola, R. L. Blakeley and B. Zerner, J. Am. Chem.–Eur. J., 1999, 5, 1617–1630.
Chem. Soc., 1975, 97, 4131–4133. 38 A. M. Barrios and S. J. Lippard, J. Am. Chem. Soc., 2000, 122,
3 H. L. T. Mobley, M. D. Island and R. P. Hausinger, Microbiol. 9172–9177.
Rev., 1995, 59, 451–480. 39 F. Musiani, E. Arnofi, R. Casadio and S. Ciurli, J. Biol. Inorg.
4 H. L. T. Mobley and R. P. Hausinger, Microbiol. Rev., 1989, 53, Chem., 2001, 6, 300–314.
85–108. 40 D. Suárez, N. Diaz and K. M. Merz, Jr, J. Am. Chem. Soc., 2003,
5 R. J. C. McLean, J. C. Nickel, K.-J. Cheng and J. W. Costerton, 125, 15324–15337.
CRC Crit. Rev. Microbiol., 1988, 16, 37–79. 41 G. Estiu and K. M. Merz, Jr, J. Am. Chem. Soc., 2004, 126,
6 C. M. Collins and S. E. F. D’Orazio, Mol. Microbiol., 1993, 9, 11832–11842.
907–913. 42 G. Estiu and K. M. Merz, Jr, Biochemistry, 2006, 45, 4429–4443.
7 J. C. Atherton, Annu. Rev. Pathol. Mech. Dis., 2006, 1, 63–96. 43 G. Estiu and K. M. Merz, Jr, J. Phys. Chem. B, 2007, 111,
8 J. G. Kusters, A. H. M. Van Vliet and E. J. Kuipers, Clin. 10263–10274.
Microbiol. Rev., 2006, 19, 449–490. 44 I.-S. Park and R. P. Hausinger, Science, 1995, 267, 1156–1158.
9 K. Stingl, K. Altendorf and E. P. Bakker, Trends Microbiol., 45 I.-S. Park and R. P. Hausinger, Biochemistry, 1996, 35,
2002, 10, 70–74. 5345–5352.
10 L. E. Zonia, N. E. Stebbins and J. C. Polacco, Plant Physiol., 46 K. Yamaguchi, N. J. Cosper, C. Stalhanske, R. A. Scott,
1995, 107, 1097–1103. M. A. Pearson, P. A. Karplus and R. P. Hausinger, J. Biol. Inorg.
11 R. P. Hausinger, in Biochemistry of Nickel, Plenum Publishing Chem., 1999, 4, 468–477.
Corp., New York, 1993, pp. 23–57. 47 K. Yamaguchi and R. P. Hausinger, Biochemistry, 1997, 36,
12 R. P. Hausinger and P. A. Karplus, in Handbook of Metallo- 15118–15122.
proteins, ed. K. Wieghardt, R. Huber, T. L. Poulos and 48 G. J. King and B. Zerner, Inorg. Chim. Acta, 1997, 255, 381–388.
A. Messerschmidt, John Wiley & Sons Ltd., West Sussex, UK, 49 R. P. Hausinger, G. J. Colpas and A. Soriano, ASM News, 2001,
2001, pp. 867–879. 67, 78–84.
13 C. Follmer, Phytochemistry, 2008, 69, 18–28. 50 S. B. Mulrooney and R. P. Hausinger, FEMS Microbiol. Rev.,
14 S. Ciurli, in Metal Ions in Life Sciences, ed. A. Sigel, H. Sigel and 2003, 27, 239–261.
R. K. O. Sigel, John Wiley & Sons, Ltd., Chichester, UK, 2007, 51 M. H. Lee, S. B. Mulrooney, M. J. Renner, Y. Markowicz and
vol. 2, pp. 241–278. R. P. Hausinger, J. Bacteriol., 1992, 174, 4324–4330.
15 E. Jabri, M. B. Carr, R. P. Hausinger and P. A. Karplus, Science, 52 S. B. Mulrooney and R. P. Hausinger, J. Bacteriol., 1990, 172,
1995, 268, 998–1004. 5837–5843.
16 S. Benini, W. R. Rypniewski, K. S. Wilson, S. Miletti, S. Ciurli 53 A. Toffanin, E. Cadahia, J. Imperial, T. Ruiz-Argüeso and
and S. Mangani, Structure, 1999, 7, 205–216. J. M. Palacios, Arch. Microbiol., 2002, 177, 290–298.
17 N.-C. Ha, S.-T. Oh, J. Y. Sung, K.-A. Cha, M. H. Lee and 54 M. Maeda, M. Hidaka, A. Nakamura, H. Masaki and
B.-H. Oh, Nat. Struct. Biol., 2001, 8, 505–509. T. Uozumi, J. Bacteriol., 1994, 176, 432–442.

This journal is 
c The Royal Society of Chemistry 2009 Metallomics, 2009, 1, 207–221 | 219
View Article Online

55 J. T. Bossè, H. D. Gilmour and J. I. MacInnes, J. Bacteriol., 2001, 89 G. J. Colpas, T. G. Brayman, L.-J. Ming and R. P. Hausinger,
183, 1242–1247. Biochemistry, 1999, 38, 4078–4088.
56 C. L. Clayton, M. J. Pallen, H. Kleanthous, B. W. Wren and 90 F. Musiani, B. Zambelli, M. Stola and S. Ciurli, J. Inorg.
S. Tabaqchali, Nucleic Acids Res., 1990, 18, 362. Biochem., 2004, 98, 803–813.
57 A. Labigne, V. Cussac and P. Courcoux, J. Bacteriol., 1991, 173, 91 M. H. Lee, H. S. Pankratz, S. Wang, R. A. Scott,
1920–1931. M. G. Finnegan, M. K. Johnson, J. A. Ippolito,
58 D. L. Weeks, S. Eskandar, D. R. Scott and G. Sachs, Science, D. W. Christianson and R. P. Hausinger, Protein Sci., 1993, 2,
2000, 287, 482–485. 1042–1052.
59 V. Cussac, R. L. Ferrero and A. Labigne, J. Bacteriol., 1992, 174, 92 T. G. Brayman and R. P. Hausinger, J. Bacteriol., 1996, 178,
2466–2473. 5410–5416.
60 J. Stoof, S. Breijer, R. G. J. Pot, D. van der Neut, E. J. Kuipers 93 G. J. Colpas, T. G. Brayman, J. McCracken, M. A. Pressler,
and A. H. M. van Vliet, Environ. Microbiol., 2008, 10, 2586–2597. G. T. Babcock, L.-J. Ming, C. M. Colangelo, R. A. Scott and
61 J. K. Kim, S. B. Mulrooney and R. P. Hausinger, J. Bacteriol., R. P. Hausinger, J. Biol. Inorg. Chem., 1998, 3, 150–160.
2005, 187, 7150–7154. 94 G. J. Colpas and R. P. Hausinger, J. Biol. Chem., 2000, 275,
62 S. B. Mulrooney, H. S. Pankratz and R. P. Hausinger, J. Gen. 10731–10737.
Microbiol., 1989, 135, 1769–1776. 95 N. E. Grossoehme, S. B. Mulrooney, R. P. Hausinger and
63 S. B. Mulrooney, S. K. Ward and R. P. Hausinger, J. Bacteriol., D. E. Wilcox, Biochemistry, 2007, 46, 10506–10516.
2005, 187, 3581–3585. 96 H. K. Song, S. B. Mulrooney, R. Huber and R. P. Hausinger,
64 S. E. Walz, S. K. Wray, S. I. Hull and R. E. Hull, J. Bacteriol., J. Biol. Chem., 2001, 276, 49359–49364.
Published on 09 April 2009. Downloaded on 10/10/2014 08:35:54.

1988, 170, 1027–1033. 97 A. C. Rosenzweig, D. L. Huffman, M. Y. Hou, A. K. Wernimont,


65 G.-F. Gerlach, S. Clegg and W. A. Nichols, FEMS Microbiol. R. A. Pufahl and T. V. O’Halloran, Structure, 1999, 7, 605–617.
Lett., 1988, 50, 131–135. 98 B. Sha, S.-K. Lee and D. M. Cyr, Structure, 2000, 8, 799–807.
66 S. B. Mulrooney, M. J. Lynch, H. L. T. Mobley and 99 B. Sriwanthana, M. D. Island, D. Maneval and H. L. T. Mobley,
R. P. Hausinger, J. Bacteriol., 1988, 170, 2202–2207. J. Bacteriol., 1994, 176, 6836–6841.
67 P. Voland, D. L. Weeks, E. A. Marcus, C. Prinz, G. Sachs and 100 S. Benoit and R. J. Maier, J. Bacteriol., 2003, 185, 4787–4795.
D. Scott, Liver Physiol., 2003, 284, G96–G106. 101 S. Ciurli, N. Safarof, S. Miletti, A. Dikiy, S. K. Christensen,
68 J.-U. Park, J.-Y. Song, Y.-C. Kwon, M.-J. Chung, J.-S. Jun, K. Kornetzky, D. A. Bryant, I. Vandenberghe, B. Devreese,
J.-W. Park, S.-G. Park, H.-R. Hwang, S.-H. Choi, S.-C. Baik, B. Samyn, H. Remaut and J. Van Beeumen, J. Biol. Inorg. Chem.,
H.-L. Kang, H.-S. Youn, W.-K. Lee, M.-J. Cho and K.-H. Rhee, 2002, 7, 623–631.
Mol. Cell., 2005, 20, 371–377. 102 M. Stola, F. Musiani, S. Mangani, P. Turano, N. Safarov,
69 I.-S. Park, M. B. Carr and R. P. Hausinger, Proc. Natl. Acad. Sci. B. Zambelli and S. Ciurli, Biochemistry, 2006, 45, 6495–6509.
U. S. A., 1994, 91, 3233–3237. 103 H.-S. Won, Y.-H. Lee, J.-H. Kim, I. S. Shin, M. H. Lee and
70 Z. Chang, J. Kuchar and R. P. Hausinger, J. Biol. Chem., 2004, B.-J. Lee, J. Biol. Chem., 2004, 279, 17466–17472.
279, 15305–15313. 104 H. Remaut, N. Safarof, S. Ciurli and J. Van Beeumen, J. Biol.
71 S. Quiroz-Valenzuela, S. C. K. Sukuru, R. P. Hausinger, L. A. Kuhn Chem., 2001, 276, 49365–49370.
and W. T. Heller, Arch. Biochem. Biophys., 2008, 480, 51–57. 105 A. Soriano, G. J. Colpas and R. P. Hausinger, Biochemistry,
72 J.-C. Rain, L. Selig, H. de Reuse, V. Battaglia, C. Reverdy, 2000, 39, 12435–12440.
S. Simon, G. Lenzen, F. Petel, J. Wojcik, V. Schächter, 106 F. Sebbane, M. A. Mandrand-Bethelot and M. Simonet,
Y. Chemama, A. Labigne and P. Legrain, Nature, 2001, 409, J. Bacteriol., 2002, 184, 5706–5713.
211–215. 107 Y.-Y. M. Chen and R. A. Burne, J. Bacteriol., 2003, 185,
73 S. R. Heimer and H. L. Mobley, J. Bacteriol., 2001, 183, 6773–6779.
1423–1433. 108 H. L. T. Mobley, R. M. Garner and P. Bauerfeind, Mol.
74 M. B. C. Moncrief and R. P. Hausinger, J. Bacteriol., 1996, 178, Microbiol., 1995, 16, 97–109.
5417–5421. 109 P. Bauerfiend, R. M. Garner and H. L. T. Mobley, Infect.
75 K. Y. Kim, C. H. Yang and M. H. Lee, Arch. Pharm. Res., 1999, Immun., 1996, 64, 2877–2880.
22, 274–278. 110 G. S. Davis, E. L. Flannery and H. L. T. Mobley, Infect. Immun.,
76 J. K. Kim and R. P. Hausinger, J. Bacteriol., 2006, 188, 2006, 74, 6811–6820.
8413–8420. 111 K. Schauer, B. Gouget, M. Carrière, A. Labigne and H. de Reuse,
77 M. Salomone-Stagni, B. Zambelli, F. Musiani and S. Ciurli, Mol. Microbiol., 2007, 63, 1054–1068.
Proteins, 2007, 68, 749–761. 112 S. Suerbaum, J.-M. Thiberge, I. Kansau, R. L. Ferrero and
78 M. B. C. Moncrief and R. P. Hausinger, J. Bacteriol., 1997, 179, A. Labigne, Mol. Microbiol., 1994, 14, 959–974.
4081–4086. 113 I. Kansau, F. Guillain, J.-M. Thiberge and A. Labigne, Mol.
79 B. Zambelli, M. Stola, F. Musiani, K. De Vriendt, B. Samyn, Microbiol., 1996, 22, 1013–1023.
B. Devreese, J. Van Beeumen, A. Dikiy, D. A. Bryant and 114 S. Cun, H. Li, R. Ge, M. C. M. Lin and H. Sun, J. Biol. Chem.,
S. Ciurli, J. Biol. Chem., 2005, 280, 4684–4695. 2008, 283, 15142–15151.
80 B. Zambelli, F. Musiani, M. Savini, P. Tucker and S. Ciurli, 115 J. V. Gilbert, J. Ramakrishna, F. W. Sunderman Jr, A. Wright
Biochemistry, 2007, 46, 3171–3182. and A. G. Plaut, Infect. Immun., 1995, 63, 2682–2688.
81 B. Zambelli, P. Turano, F. Musiani, P. Neyroz and S. Ciurli, 116 S. Seshadri, S. L. Benoit and R. J. Maier, J. Bacteriol., 2007, 189,
Proteins, 2008, 74, 222–239. 4120–4126.
82 N. Mehta, S. Benoit and R. J. Maier, Microb. Pathog., 2003, 35, 117 R. Ge, Y. Zhang, X. Sun, R. M. Watt, Q.-Y. He, J.-D. Huang,
229–234. D. E. Wilcox and H. Sun, J. Am. Chem. Soc., 2006, 128,
83 P. Neyroz, B. Zambelli and S. Ciurli, Biochemistry, 2006, 45, 11330–11331.
8918–8930. 118 R. Ge, R. M. Watt, X. Sun, J. A. Tanner, Q.-Y. He, J.-D. Huang
84 R. Gasper, A. Scrima and A. Wittinghofer, J. Biol. Chem., 2006, and H. Sun, Biochem. J., 2006, 393, 285–293.
281, 27492–27502. 119 Y.-B. Z. Zeng, D.-M. H. Li and H. Sun, J. Biol. Inorg. Chem.,
85 I.-S. Park and R. P. Hausinger, J. Bacteriol., 1995, 177, 2008, 13, 1121–1131.
1947–1951. 120 J. W. Olson, N. S. Mehta and R. J. Maier, Mol. Microbiol., 2001,
86 A. Soriano and R. P. Hausinger, Proc. Natl. Acad. Sci. U. S. A., 39, 176.
1999, 96, 11140–11144. 121 S. L. Benoit, A. L. Zbell and R. J. Maier, Microbiology, 2007, 153,
87 S. Quiroz, J. K. Kim, S. B. Mulrooney and R. P. Hausinger, in 3748–3756.
Metal Ions in Life Sciences, ed. A. Sigel, H. Sigel and R. K. O. 122 J. W. Olson and R. J. Maier, Science, 2002, 298, 1788–1790.
Sigel, John Wiley & Sons, New York, 2007, vol. 2, pp. 519–544. 123 N. S. Mehta, J. W. Olson and R. J. Maier, J. Bacteriol., 2003, 185,
88 K. Stingl, K. Schauer, C. Ecobichon, A. Labigne, P. Lenormand, 726–734.
J.-C. Rousselle, A. Namane and H. de Reuse, Mol. Cell. 124 L.-T. Hu and H. L. T. Mobley, Infect. Immun., 1993, 61,
Proteom., 2008, 7, 2429–2441. 2563–2569.

220 | Metallomics, 2009, 1, 207–221 This journal is 


c The Royal Society of Chemistry 2009
View Article Online

125 S. L. Benoit, N. Mehta, M. V. Weinberg, C. Maier and 151 Q. Liu and R. A. Bender, J. Bacteriol., 2007, 189, 7593–7599.
R. J. Maier, Microbiology, 2007, 153, 1474–1482. 152 L. V. Wray, Jr, A. E. Ferson and S. H. Fisher, J. Bacteriol., 1997,
126 H. G. Kolmark, Mutat. Res., 1969, 8, 51–63. 179, 5494–5501.
127 P. Haysman and H. B. Howe, Jr, Can. J. Genet. Cytol., 1971, 13, 153 A. D. Larson and R. E. Kallio, J. Bacteriol., 1954, 68, 67–73.
256–269. 154 E. Perez-Urria, M. Rodriguez and C. Vicente, Plant Mol. Biol.,
128 E. W. Benson and H. B. Howe, Jr, Mol. Gen. Genet., 1978, 165, 1989, 13, 665–672.
277–282. 155 Y. Watanabe, T. Iizuka and N. Shimada, Soil Sci. Plant Nutr.,
129 E. M. Mackay and J. A. Pateman, J. Gen. Microbiol., 1980, 116, 1994, 40, 545–548.
249–251. 156 A. H. M. van Vleit, E. J. Kuipers, B. Waidner, B. J. Davies, N. de
130 E. M. Mackay and J. A. Pateman, Biochem. Genet., 1982, 20, Vries, C. W. Penn, C. M. J. E. Vandenbroucke-Grauls, M. Kist,
763–776. S. Bereswill and J. G. Kusters, Infect. Immun., 2001, 69,
131 J. R. Kinghorn and R. Fluri, Curr. Genet., 1984, 8, 99–105. 4891–4897.
132 M. W. Lubbers, S. B. Rodriquez, N. K. Honey and 157 A. H. M. van Vliet, S. W. Poppelaars, B. J. Davies, J. Stoof,
R. J. Thornton, Can. J. Microbiol., 1996, 42, 132–140. S. Bereswill, M. Kist, C. W. Penn, E. J. Kuipers and
133 Y. Tange and O. Niwa, Curr. Genet., 1997, 32, 244–246. J. G. Kusters, Infect. Immun., 2002, 70, 2846–2852.
134 M. Bacanamwo, C.-P. Witte, M. W. Lubbers and J. C. Polacco, 158 M. Contreras, J.-M. Thiberg, M.-A. Mandrand-Berthelot and
Mol. Gen. Genet., 2002, 268, 525–534. A. Labigne, Mol. Microbiol., 2003, 49, 947–963.
135 P. W. Riddles, V. Whan, R. L. Blakeley and B. Zerner, Gene, 159 F. D. Ernst, J. Stoof, W. M. Horrevoets, E. J. Kuipers,
1991, 108, 265–267. J. G. Kusters and A. H. M. Van Vliet, Infect. Immun., 2006, 74,
Published on 09 April 2009. Downloaded on 10/10/2014 08:35:54.

136 M. Pires-Alves, Grossi-de-Sá, G. B. S. Barcellos, C. R. Carlini 6821–6828.


and M. G. Moraes, Plant Cell Physiol., 2003, 44, 139–145. 160 C. Dian, K. Schauer, U. Kapp, S. M. McSweeney, A. Labigne
137 L. E. Meyer-Bothling and J. E. Polacco, Mol. Gen. Genet., 1987, and L. Terradot, J. Mol. Biol., 2006, 361, 715–730.
209, 439–444. 161 A. H. M. van Vliet, F. D. Ernst and J. G. Kusters, Trends
138 R. W. Krueger, M. A. Holland, D. Chisholm and J. C. Polacco, Microbiol., 2004, 12, 489–494.
Gene, 1987, 54, 41–50. 162 F. D. Ernst, E. J. Kuipers, A. Heijens, R. Sarwari, J. Stoof,
139 M. A. Holland, J. D. Griffin, L. E. Meyer-Bothling and C. W. Penn, J. G. Kusters and A. H. van Vliet, Infect Immun,
J. C. Polacco, Dev. Genet., 1987, 8, 375–387. 2005, 73, 7252–7258.
140 R. S. Torisky, J. D. Griffin, R. L. Yenofsky and J. E. Polacco, 163 L. O. Abraham, Y. Li and D. B. Zamble, J. Inorg. Biochem., 2006,
Mol. Gen. Genet., 1994, 242, 404–414. 100, 1005–1014.
141 L. E. Meyer-Bothling, J. C. Polacco and S. R. Cianzio, Mol. Gen. 164 C. Fauquant, R. E. Diederix, A. Rodrigue, C. Dian, U. Kapp,
Genet., 1987, 209, 432–438. L. Terradot, M. A. Mandrand-Berthelot and I. Michaud-Soret,
142 S. K. Freyermuth, M. Bacanamwo and J. C. Polacco, Plant J., Biochimie, 2006, 88, 1693–1705.
2000, 21, 53–60. 165 N. S. Dosanjh, N. A. Hammerbacher and S. L. Michel, Biochemistry,
143 C.-P. Witte, E. Isadore, S. A. Tiller, H. V. Davies and 2007, 46, 2520–2529.
M. A. Taylor, Plant Mol. Biol., 2001, 45, 169–179. 166 B. Zambelli, M. Belluci, A. Danielli, V. Scarlato and S. Ciurli,
144 C.-P. Witte, M. G. Rosso and T. Romeis, Plant Physiol., 2005, Chem. Commun., 2007, 3649–3651.
139, 1155–1162. 167 E. L. Benati and P. T. Chivers, J. Biol. Chem., 2007, 282,
145 S. E. F. D’Orazio and C. M. Collins, J. Bacteriol., 1993, 175, 20365–20375.
3459–3467. 168 B. Zambelli, A. Danielli, S. Romagnoli, P. Neyroz, S. Ciurli and
146 I. Gendlina, D. M. Gutman, V. Thomas and C. M. Collins, V. Scarlato, J. Mol. Biol., 2008, 383, 1129–1143.
J. Biol. Chem., 2002, 277, 37349–37358. 169 N. S. Dosanjh, A. L. West and S. L. Michel, Biochemistry, 2009,
147 C. H. Sisson, H. E. R. Perinpanayagam and E. M. Hancock, Oral 48, 527–536.
Microbiol. Immunol., 1992, 7, 159–164. 170 R. G. J. Pot, J. Stoof, P. J. M. Nuijten, L. A. M. de Haan,
148 Y.-Y. M. Chen, M. J. Betzenhauser and R. A. Burne, Microbiology, P. Loeffen, E. J. Kuipers, A. H. M. Van Vliet and J. G. Kusters,
2002, 148, 3599–3608. FEMS Immunol. Med. Microbiol., 2007, 50, 273–279.
149 B. Masepohl, B. Kaiser, N. Isakovic, C. L. Richard, R. G. Kranz 171 J. K. Hendricks and H. L. Mobley, J. Bacteriol., 1997, 179,
and W. Klipp, J. Bacteriol., 2001, 183, 637–643. 5892–5902.
150 A. Macaluso, E. A. Best and R. A. Bender, J. Bacteriol., 1990, 172 L. Wolfram, E. Haas and P. Bauerfeind, J. Bacteriol., 2006, 188,
172, 7249–7255. 1245–1250.

This journal is 
c The Royal Society of Chemistry 2009 Metallomics, 2009, 1, 207–221 | 221

You might also like