You are on page 1of 11

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/51703770

Copper toxicity and the origin of bacterial resistance - New insights and
applications

Article  in  Metallomics · November 2011


DOI: 10.1039/c1mt00107h · Source: PubMed

CITATIONS READS

120 865

3 authors, including:

Gregor Grass Christopher Rensing


Bundeswehr Institute of Microbiology Chinese Academy of Sciences
105 PUBLICATIONS   5,214 CITATIONS    203 PUBLICATIONS   8,234 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Bacteria-Nematode Interactions: Bacterial Roles on Pine Wilt Disease and Plant Parasitic Nematodes View project

Comparative reproduction View project

All content following this page was uploaded by Christopher Rensing on 01 June 2014.

The user has requested enhancement of the downloaded file.


View Online
Metallomics Dynamic Article Links

Cite this: DOI: 10.1039/c1mt00107h

www.rsc.org/metallomics MINIREVIEW
Copper toxicity and the origin of bacterial resistance—new insights and
applicationsw
Christopher L. Dupont,a Gregor Grassb and Christopher Rensing*c
Received 18th July 2011, Accepted 27th September 2011
Published on 10 October 2011 on http://pubs.rsc.org | doi:10.1039/C1MT00107H

DOI: 10.1039/c1mt00107h

The bioavailability of different metals has likely changed over the course of Earth’s history. Based
on geochemical models, copper became much more bioavailable with the advent of an oxidizing
atmosphere. This posed both a challenge and an opportunity for the organisms at that time.
Specifically, copper resistance mechanisms were required first and to do this Bacteria appear to
Downloaded by RSC Internal on 18 October 2011

have modified already existing protein structures. Later, Cu-utilizing proteins evolved and
continue to be used sparingly, at least relative to later evolving Eukarya, by Bacteria but with
significant biogeochemical consequences. Copper is a strong soft metal that can attack
intracellular iron–sulfur centers of various proteins under primarily anoxic conditions. In oxic
conditions, copper can catalyze a Fenton-like reaction that may cause lipid peroxidation and
protein damage. The inherent ability of copper to inflict damage upon multiple cellular functions
has been harnessed by macrophages and perhaps amoeba to kill and later digest bacteria and
other microorganisms. Notably, these organisms, unlike Bacteria, most likely evolved after
increases in copper availability, implying that Eukarya utilized their own trafficking and resistance
mechanisms, in addition to the natural toxicity of copper, as leverage in interactions with
Bacteria. In an ‘‘arms race,’’ some pathogenic bacteria have evolved new mechanisms for copper
resistance, which is relevant given renewed interest in the use of copper surfaces due to their
antimicrobial properties.
a
Microbial and Environmental Genomics, J. Craig Venter Institute, San Diego, CA, USA
b
Bundeswehr Institute of Microbiology, Munich, Germany
c
Department of Soil, Water, and Environmental Science, University of Arizona, Shantz Bld #38 Rm 429, Tucson, AZ 85721, USA.
E-mail: rensingc@ag.arizona.edu; Fax: +1 (520) 621-1647; Tel: +1 (520) 626-8482
w This article is published as part of a themed issue on Metal Toxicity, Guest Edited by Gregor Grass and Christopher Rensing.

Christopher L. Dupont, Gregor Grass, Oberregierungs-


Assistant Professor in rat, Bundeswehr Institut of
Microbial and Environmental Microbiology. Gregor Grass
Genomics, J. Craig Venter works as a tenured senior
Institute. Chris L. Dupont research scientist at the Bundes-
received a MS in Environmental wehr Institut of Microbiology
Engineering from Cornell Uni- (Munich, Germany) since 2011.
versity and a PhD in Marine He is interested in how bacteria
Biology from the Scripps Insti- acquire essential transition
tution of Oceanography. Chris metals and in systems involved
joined the J. Craig Venter in metal detoxification. Cur-
Institute in 2008, where he is rently, his research focuses on
an assistant professor in Micro- the antimicrobial mode-of-action
bial and Environmental Geno- of metallic copper and biodefense
Christopher L. Dupont mics. His research focuses on Gregor Grass issues. Gregor studied Biology at
microbial physiology and the the Martin-Luther-University
role of environment in shaping molecular adaptations and (MLU, Halle, Germany). He completed his Ph.D. in microbiology
diversification. in 2000 and then worked as a postdoctoral fellow with Chris Rensing
in Tucson, AZ. In 2002, Gregor become an independent non tenure-
track group leader at MLU before temporarily accepting a position as
assistant professor at the University of Nebraska-Lincoln in 2008.

This journal is c The Royal Society of Chemistry 2011 Metallomics


View Online

Introduction Cu(I)-translocating P-type ATPases. Interestingly, Cu(I) is also


coordinated in trigonal planar geometry by three Mets in the
The transition metal copper (Cu) occurs as Cu(I) and Cu(II) in eukaryotic Cu uptake transporter Ctr15 that seems to be absent
nature. Cu(I) is a strong soft metal whereas Cu(II) is borderline in bacteria.
according to the Hard and Soft Acids and Bases (HSAB)
classification.1 Therefore, Cu binding sites are dominated by
amino acids with soft (sulfur donor atoms) and borderline Evolutionary perspectives on Cu utilization and
(nitrogen donor atoms) ligands. These are cysteine and resistance in bacteria
methionines as soft ligands and histidine as a borderline The so-called Great Oxidation Event (GOE), which occurred
ligand. Cu(I) [Ar]3d10 can be found coordinated by 2, 3 or between 2.4–2.7 billion years ago, refers to an increase in
4 ligands and showing linear, trigonal planar, or tetrahedral atmospheric oxygen that potentially changed the bioavailability
geometries. Cu(II) [Ar]3d9 can be found coordinated by 4, 5, or of metals including Cu.6 Specifically, according to geochemical
6 ligands in geometries such as square planar, square pyramidal,
Published on 10 October 2011 on http://pubs.rsc.org | doi:10.1039/C1MT00107H

models, in an anoxic or euxinic (anoxic and sulfidic) ocean, a


or distorted octahedral. Proteins using Cu as a cofactor have chalcophilic metal like Cu would be insoluble.6 Thus increasing
positive reduction potentials, as do some iron-containing global oxygen should augment a source of Cu, specifically,
proteins that evolved after the rise in oxygen. Since Cu
continental weathering, and also increase the solubility of Cu in
proteins first appeared under oxidizing conditions, most
the ocean. Phylogenomic reconstructions have recently been
evolved functions related to oxygen handling and are involved
used to determine the temporal evolution of the Structural
in electron transfer, oxygen transport and in redox reactions Classification of Proteins (SCOP) fold superfamilies,7 which
Downloaded by RSC Internal on 18 October 2011

of a broad substrate range such as multicopper oxidases are evolutionarily related protein structures. These were
(laccases, Fet3, CueO). Most of these proteins bind Cu tightly combined with a genome census for these fold superfamilies
with high coordination binding sites of 4 to 5 to prevent loss of and aligned to the modeled geological record using several
Cu during redox cycling. Cu trafficking proteins naturally transitional arguments.8 These aligned genome and geological
contain low affinity binding sites or have flexible geometries, records suggest that Cu-binding enzymes evolved after critical
so that metal transfer is possible between two binding sites. oxidative defense enzymes like superoxide dismutase and
The coordination numbers here would be 2 to 3. Recently catalase (Fig. 1).8a These enzymes are essential for oxygen
available protein structures confirm this general principle. In
tolerance in bacteria, thus their evolution must have preceded
the central transport protein CusA Cu(I) is coordinated in a
or coincided with the invention of oxygenic photosynthesis
trigonal planar geometry to three methionines. CusA is part of
and subsequent accumulation of oxygen in the atmosphere.
the CusCBA RND-type transport complex common in Gram Just as with oxygen itself, the increased bioavailability of Cu
negative bacteria.2 In the unrelated Cu(I)-translocating P-type posed both an opportunity and a challenge for all organisms;
ATPases CopA from Legionella pneumophila, Cu(I) also as a cofactor it participates in new biochemical enzymes and
appears to be coordinated in a trigonal planar geometry at pathways, as a free ion it acted as poison. In the period
both binding sites along the transport pathway, first by two Cys following the GOE, new Cu binding proteins with metabolic
and Tyr in transmembrane (TM) binding site I, then by Asn, roles in aerobic and in facultative aerobic bacteria evolved,
Met and Ser in TM binding site II.3 This has also been predicted including the Cu binding cupredoxin fold superfamily, which
in homologous proteins such as CopA from Archaeglobus encompasses the Cu binding proteins plastocyanin, nitrosocyanin,
fulgidus4 and might be universally applicable for all
azurin, and the periplasmic domain of cytochrome c oxidase
subunit II (Fig. 1). Imposed over this timeline of protein
evolution is organismal evolution. Many bacterial lineages,
Christopher Rensing, Associ- including cyanobacteria, were already greatly diversified at the
ate Professor of Microbiol- GOE. Given that phylogenetic speciation prior to Cu-protein
ogy, University of Arizona.
evolution would hinder widespread usage, it is not surprising
Chris Rensing studied and
obtained his PhD at the Freie that none of the new Cu-binding proteins are found in all
Universität Berlin and later extant bacteria. That is, new Cu proteins were likely invented
Martin-Luther Universität by specific lineages, with sparing horizontal gene transfer later.
Halle-Wittenberg supervised Overall, incorporation of genes of Cu-binding proteins into
by Dietrich Nies in 1996. He bacterial genomes is rare, with less than 0.3% of the annotated
then joined Barry Rosen’s lab proteome being Cu-binding in Bacteria (Fig. 2). Later-
at Wayne State University in evolving Cu-binding proteins are found in small numbers of
Detroit as a postdoctoral fel- modern bacterial genomes. Consistent with Cu utilization
low mainly working on the being tied to aerobic metabolism, obligate anaerobic bacteria
biochemical characterization have even fewer Cu binding proteins (Fig. 2). In contrast, the
Christopher Rensing of two metal-transporting
genomes of modern Eukarya appear to reflect an evolution in
P-type ATPases, ZntA and
CopA. In 1999, he accepted a faculty position at the University an environment with elevated Cu concentrations; a greater
of Arizona and in 2007 was promoted to associate professor. His percentage of Eukaryotic genomes code for Cu binding proteins
research is looking at different aspects of metal–microbe inter- (Fig. 2), particularly late-evolving Cu-binding proteins.
actions, encompassing both environmental as well as medical While increasing Cu concentrations provided a redox-facile
microbiology. ligand for metabolic enzymes, it also posed a toxic threat.

Metallomics This journal is c The Royal Society of Chemistry 2011


View Online
Published on 10 October 2011 on http://pubs.rsc.org | doi:10.1039/C1MT00107H
Downloaded by RSC Internal on 18 October 2011

Fig. 1 The evolution of Cu utilization and modern day genomic incorporation of Cu proteins. Shown are parallel timelines of the evolution of
oxygen in the atmosphere, trace metal chemistry in the ocean, and the evolution of Cu binding protein structures. Several key events, like the Great
Oxidation Event and the first physical Eukaryotic fossils are shown for references. For each noted protein fold superfamily, the histograms show
the percent of Bacterial and Eukaryotic genomes that contain them. Data is compiled and replotted from ref. 8a,71.

What would have been the initial manifestation of Cu toxicity transport in modern bacteria (Fig. 1). Finally, the ‘‘heavy
to early life? One way that Cu can be toxic is through the metal associated domain’’ (SCOP id 55008) includes the metal
generation of free radicals, though this would have been binding domains of Zn and Cu transporting ZntA and CopA,
minimal during the early stages of the planetary oxidation the mercury binding protein MerP, and Cu chaperones found
(See O2 trace on Fig. 1). As discussed in more detail below,
Fe–S cluster -containing proteins are a target of Cu(I) based
toxicity. At the time of the GOE, at least over half of the
known Fe–S cluster binding structures had evolved,8a as had
the structures involved in Fe–S biosynthesis, thus damage
and inhibition of Fe–S metabolism is the more likely initial
manifestation of Cu toxicity.
Within the same phylogenies, it appears that ancient
bacteria co-opted already existing protein families to develop
Cu resistance. In modern bacteria, intracellular Cu concentrations
are carefully controlled by a system of Cu-sensing regulatory
proteins, Cu transporters, and possibly Cu chelators. While
metal homeostasis proteins might appear divergent in terms of
amino acid sequence, from a structural standpoint, many of
the proteins involved in sensing and trafficking numerous
metals are quite similar and belong to one of three fold
superfamilies. One is the Ferric Uptake Regulatory (FUR)
protein family, which includes sensors for Fe, Zn, and Ni. At
the even grander structural scale, the FUR protein family is Fig. 2 The average number of Cu binding domains in the genomes of
related to 83 protein families with a ‘‘winged helix DNA- the three superkingdoms of life (A) and the percent of predicted
binding’’ domain (SCOP id 46785), including the arsenic and proteins in those superkingdoms that are Cu binding (B). In B, the
cadmium binding ArsR and CadC.9 The ‘‘helical backbone black line in the Bacterial column shows the percent from obligate
metal receptor domain’’ (SCOP id 53807) is involved in metal anaerobic bacteria.

This journal is c The Royal Society of Chemistry 2011 Metallomics


View Online

in Eukaryotes like ATX1. On structural protein phylogenies, Haber–Weiss reactions (Cu+ + H2O2 - HO + HO +
these protein families evolved prior to the GOE (Fig. 1), Cu2+, O2 + Cu2+ - Cu+ + O2).13 That highly reactive
though the ‘‘parent’’ metal bound at that time is unknown. oxygen intermediates are responsible for lipid peroxidation,
They are also nearly ubiquitous and abundant in bacterial oxidation of proteins and damage to nucleic acids was
genomes. proposed as well.13b,14
In summary, the combination of phylogenies of protein The effect of Cu(I) on generation of reactive oxygen
architectures and surveys of extant bacteria genomes suggests species can also be indirect. Free Cu ions are able to oxidize
that the protein architecture required to traffic Cu arose prior sulfhydryl-groups, such as cysteine in proteins or the cellular
to the GOE, though it might not have been used for Cu at redox-buffer glutathione.15 Degradation of Fe–S clusters
that time. As changes in global redox chemistry increased would lead to the release of iron. An increase in unbound free
environmental Cu availability, our interpretation is that iron can then lead to oxidative damage through iron-based
bacteria tuned existing metal sensing, transporter, and storage Fenton chemistry.16 All of these events can cause oxidative
Published on 10 October 2011 on http://pubs.rsc.org | doi:10.1039/C1MT00107H

proteins. Concomitantly, this facilitated new metabolic damage but it appears only indirectly through Cu. Moreover,
utilizations of Cu, but only by select bacterial lineages. it was believed previously that Cu ion toxicity in bacteria was
mediated by oxidative DNA damage. However, under
anaerobic conditions Cu ions suppressed the growth rate of
Copper toxicity
E. coli even more strongly than under aerobic conditions.17
A question that is of interest in this review is how Cu becomes More importantly, addition of Cu ions to E. coli cells
toxic to cells. What are the targets of Cu? Many studies decreased oxidative DNA damage when cells were challenged
Downloaded by RSC Internal on 18 October 2011

focused on the ability of Cu ions to redox-cycle between with hydrogen peroxide.18 Table 1 summarizes the conse-
Cu(I)/Cu(II). This Fenton-like reaction would then generate quences of contact of microbes to different forms of Cu focusing
reactive oxygen species leading to lipid peroxidation and on aerobic conditions.
protein oxidation and DNA damage. However, as shown
quite impressively in ref. 10, Cu under anaerobic, reducing
Cu homeostasis in bacteria
conditions was most toxic. Cu(I) is a strong soft metal with high
affinity for thiolates and can destabilize iron–sulfur clusters. Little is known about how most bacteria acquire Cu. Much
This is often overlooked as toxicologists and others focus on of what is known about Cu homeostasis in prokaryotes
oxidative damage in many diseases where Cu plays a role. originated from the bacterial model organism Escherichia coli.
Excess Cu led to an increase in iron acquisition and sulfur This enterobacterium probably does not require cytoplasmic
assimilation both in E. coli and in B. subtilis.10,11 This is due to Cu, and it is believed that Cu-requiring proteins are exclusively
decreased iron–sulfur cluster stability both during their located in the periplasm or within the cytoplasmic membrane
biogenesis and when bound to their target proteins. Genes (reviewed in ref. 19) with the Cu center facing the periplasm and
encoding enzymes containing iron sulfur clusters that are finally some in intracellular membranes (e.g. cyanobacterial
induced during Cu stress in B. subtilis include biotin (BioB), photosynthesizers and methanotrophs).
molybdopterin (MoaA), pyrimidin and branched-chain amino In contrast, defense systems against Cu-overload are
acid synthesis. In E. coli dihydroxy-acid dehydratase (IlvD) in conserved in most Gram-negative and even Gram-positive
the common branched-chain amino acid synthesis pathway, organisms.
isopropylmalate dehydratase (LeuC) in the leucine-specific Detoxification of superfluous cytoplasmic Cu is accomplished
branch, fumarase A (FumA) in the tricarboxylic acid cycle by P-type ATPases that efficiently pump out Cu(I).20 Periplasmic
and 6-phosphogluconate dehydratase in the pentose phos- Cu is effluxed by huge multi-component protein complexes
phate pathway (Edd) were found to be inactivated by Cu ions; such as CusCBA of E. coli.17,21 CusA is a resistance/nodulation/
thus, these proteins constitute targets for lethal damage.10 cell division (RND) protein, driven by the proton motive force,
Since these enzymes fulfill vital cell functions, it is essential that together with CusB and CusC transport Cu to the
to keep Cu at very low concentrations in the cytoplasm. In extracellular space. Mechanistically, CusCBA functions
fact, the P-type ATPase regulated by CueR in E. coli allows similar to a peristaltic pump with three alternating enzyme
only extremely low concentrations of free Cu in the cell.12 In conformations, as was shown for a related system.22 Three
effect, there is almost no free Cu in cells. In addition to conserved methionines in the periplasmic cleft of CusA are
degradation of iron–sulfur clusters of vital enzymes, excess responsible for specifically binding and releasing Cu(I).2 The
Cu would effectively lead to an intracellular sink for iron CusCBA complex can sense periplasmic Cu concentrations
making increased iron uptake necessary.11 This would put cells at through three conserved N-terminal residues and adjust
a serious disadvantage in an environment with increasing transport efficiency accordingly.23 The third component of Cu
concentrations of bioavailable Cu and diminishing concentrations homeostasis is a multicopper oxidase (MCO). In E. coli the
of soluble iron. Most bacteria solved this situation by having Cu periplasmic MCO CueO oxidizes Cu(I) to Cu(II), thus protecting
containing enzymes only in the periplasm or facing out in the periplasmic proteins from Cu(I)-mediated toxicity.24 CueO might
cytoplasmic membrane. In the few bacteria that need Cu delivered also oxidize the siderophore enterobactin when Cu levels
to intracellular compartments (e.g. photosynthetic bacteria), are high, providing a Cu-binding oxidation product diminishing
chaperones guide Cu through the cytoplasm as in eukaryotes. Cu-uptake into the cytoplasm.25
Aerobically, Cu readily catalyzes reactions that result in the Surprisingly, a close relative of E. coli, Salmonella enterica
production of hydroxyl radicals through the Fenton and features a variation of this prototypical three-legged Cu

Metallomics This journal is c The Royal Society of Chemistry 2011


View Online

Table 1 Consequences of diverse kinds of aerobic Cu stress

Planktonic culture = Biofilms culture = Cells exposed to moist Cells exposed to dry
growth with Cu salts growth on wet Cu surface Cu surfaces Cu surfaces
Stress exposure Chronic (cells grow Chronic (cells adapt to Acute (hybrid of planktonic Acute
under stress) stress and grow) culture and dry surfaces)
Cu speciation Cu(I), Cu(II) Cu(0), Cu(I), Cu(II) Cu(0), Cu(I), Cu(II) Cu(0), Cu(I), Cu(II)
Cell growth under Yes (up to mM Cu) Yes (live cells on top of No significant growth No
stress dead cell barrier)
Mechanism of cell Fe–S cluster in Probably Fe–S cluster in Unknown, probably Cu ion and Unknown, probably Cu
damage hydratases and other hydratases and other factors ROS related, targets unknown ion and ROS related,
factors (ROS) (ROS) targets unknown
Inactivation of sensitive Slow, bacteriostatic, Initially fast, negligible Fast (min–h) Very fast (min)
cells concentration after biofilm formation
dependent
Published on 10 October 2011 on http://pubs.rsc.org | doi:10.1039/C1MT00107H

Specific Cu resistance Common, well Overlap with planktonic Overlap with planktonic culture Unknown—resistance
mechanisms studied mechanism culture but probably but probably also overlap with mechanisms different
additional unknown dry surface resistance mechanisms from that of planktonic
specialized mechanisms cells

homeostasis system. While CopA and CueO (aka CuiD) are to infection. Further, deletion of both copA and golT resulted in
present, the Cus system is not. In contrast, another periplasmic significantly fewer cells than the wild-type control in infected
Downloaded by RSC Internal on 18 October 2011

Cu defense protein, termed CueP has been identified.26 Similar macrophages, clearly demonstrating a necessity of Cu resistance
CueP proteins are not only restricted to Salmonella but can be for survival of infecting Salmonella in a macrophage model.
found in other, often pathogenic bacteria as well. The highly This observation is in good agreement with a previous study
abundant CueP in the periplasm of Salmonella is no transport showing that Cu-transporting P-type ATPases from both host
protein but the function of CueP is thought to be related to Cu and pathogen play a role during infection.30 When the host
sequestration particularly under anaerobiosis. Here, CueP Golgi Cu exporter ATP7A was expressionally silenced, killing
constitutes a major cellular Cu pool.27 Analogous to the Cus of invading E. coli was attenuated. Conversely, if these E. coli
system, CueP is also most important under anaerobic growth cells lacked CopA they were hypersensitive to macrophage-
conditions. Thus, CueP might replace the Cus system by a mediated killing.
functionally distinct mechanism. However, important parts of the Cu-resistance puzzle
Though the RND-type Cus system is lacking in Salmonella, during host–pathogen interaction are still missing. In a murine
there is an additional unrelated tripartite RND efflux pump model of infection no significant differences in colonization
encoded on the Salmonella genome. This Ges system was between wild-type Salmonella and the copA/golT double
initially identified and characterized as a gold-responsive mutant were observed.27 This might be a consequence of the
operon with a MerR-type regulator, GolS, encoded by the animal model selected or caused by more subtle differences
neighboring Gol system.28 Ges does apparently not transport between the two strains during the course of infection not yet
Cu but the adjacently encoded Gol system does. In addition to elucidated. Also, in these mutant cells the CueO protein was
the regulator GolS, the small metallochaperone GolB and the still present. It would be interesting to include CueO in such
CopA-like P-type ATPase GolT are also present.29 Though studies as well and test a triple mutant for survival in different
Gol was identified by its ability to confer gold-resistance, in models of infection. Recently, a study indicated a role for
the absence of CopA Gol also confers resistance to Cu. CueO in a murine model of systemic infection by Salmonella.31
Interestingly, while the secondary function of Gol for Thus, new surprises regarding Cu homeostasis can certainly be
Cu homeostasis was recently corroborated, gold-resistance expected. For example, cells of E. coli or Salmonella with a
conferred by the system could not be confirmed.27 defect in CueO exhibit a mucoid phenotype and aggregative
Salmonella does not only exhibit an interesting variation of behavior when Cu-stressed.32 It appears that under these
the prototype Cu homeostasis of E. coli, it also offered the conditions cells produce the extracellular polysaccharide
opportunity to study Cu homeostasis in an intracellular colonic acid, and lack of this compound increased Cu toxicity.
pathogen and the role proper Cu-handling plays during It is an open question if colonic acid production has a practical
infection. For this it was first established that the expression relevance during infection such as production of a bacterial
of copA was only dependent on Cu ions and no other relevant capsule for host-defense evasion.
factors potentially encountered in macrophages altered copA- Clearly, more research is needed in addition to these few
levels. Next macrophages were infected with a Salmonella piloting studies concerned with this subject in the past including
strain expressing a reporter comprising the copA promoter these in Legionella pneumophila,33 Pseudomonas aeruginosa34 or
and the gene for beta-galactosidase (copAp-lacZ). It was Mycobacterium tuberculosis.35 The latter example offers yet
known from previous studies that such reporters sensitively another new variation on an old theme. M. tuberculosis harbors
and dose-dependently respond to environmental Cu ions. eleven metal-translocating PIB-type ATPases. An earlier study
Expression of copAp-lacZ also occurred during the course has revealed that only one, CtpV, can be directly associated
of macrophage infection with increasing reporter activities27 with Cu homeostasis in this organism. Further, absence of
indicating that within host cells the bacteria encountered CtpV resulted in altered cellular transcription as a response to
increasing Cu ion concentrations maybe as a defense response increased Cu stress. Similar to the case of Salmonella, animal

This journal is c The Royal Society of Chemistry 2011 Metallomics


View Online

models of tuberculosis infection including mouse and guinea pig Copper and innate immunity
did not reveal an obvious decline of fitness of the mutant.
However, in contrast to infection with wild-type M. tuberculosis, One of the hallmarks of multicellular organisms is the presence
infection with ctpV mutant cells resulted in less lung damage, of an immune system. Multicellular eukaryotes evolved
attenuated immune response and increased survival times of entirely under conditions of relatively high concentrations of
the host. Cu (Fig. 1) and zinc.8a It is therefore not surprising that these
Surprisingly, M. tuberculosis possesses a novel unexpected Cu metals were highly utilized for many functions including
homeostasis component that seems to be essential for virulence development, the nervous system and immunity.39 The role
in this organism. This MctB (mycobacterial Cu transport of Cu in innate immunity is becoming better understood as of
protein B) is located in the outer membrane equivalent of the late. Cu is taken up through Ctr1 and delivered to the Golgi
mycobacterial cell wall.36 There were no homologs of MctB network by the Cu chaperone Atox1. From here the P-type
with known roles but MctB likely functions analogously to ATPase ATP7A delivers it to the phagosome where Cu(I) is
actively pumped across the membrane (Fig. 3). Macrophages
Published on 10 October 2011 on http://pubs.rsc.org | doi:10.1039/C1MT00107H

porins as outer membrane channel for Cu and silver ions.


However, MctB seems to be specifically responsible for Cu use Cu to kill off infectious agents such as microbes in
efflux but not for uptake or facilitated diffusion across the combination with production of reactive oxygen and nitric oxide
membrane, because a deletion of mctB rendered cells more species.27,30 Therefore, inflammation is often characterized by an
susceptible to Cu toxicity and cells accumulated approximately increase in Cu concentration in the affected tissue.40 Since Cu is
100-times more Cu than wild-type cells when challenged even used to actively kill bacterial cells, pathogens had to find
with low mM external Cu ion concentrations. It was suggested effective Cu resistance strategies in order to insure survival.
Downloaded by RSC Internal on 18 October 2011

that MctB and the Cu ATPase CtpV interact constituting a Consequently, copper resistance determinants are important
novel dipartite Cu efflux system.36 It remains to be seen if this for virulence in Mycobacterium tuberculosis, Salmonella enterica
interaction is physical or rather functional in nature. Notwith- and Streptococcus pneumoniae27,36,41 as described above.
standing, it appears that lack of MctB in M. tuberculosis elicits However, the mechanism used by macrophages to
the most severe Cu-dependent phenotype both in growth kill infectious agents appears to be even more widespread.
experiments and animal models when compared with strains Phagocytosis first evolved in primitive unicellular eukaryotes
lacking one of the other two major Cu homeostasis factors, such as amoeba and ciliates.42 Amoeba are known to occur
CtpV and the small metallothionein MymT that may bind in aquatic environments,43 but also appear to use simple
up to six Cu ions.37 A deletion of mctB resulted in a agricultural methods to maintain bacterial food populations
100-fold reduction in infection with M. tuberculosis in guinea in soil.44 Essentially, in addition to the actual environmental
pig lymph nodes and even 1000-fold reduction in lungs.36 This concentrations of Cu, bacteria in diverse environments are
result argues against a strict interdependence of CtpV and likely to encounter predators that utilize the inherent toxicity
MctB but it would be interesting to study M. tuberculosis of Cu. For example, most Vibrio species and the Deep Sea
mutants only harboring each one of its three Cu homeostasis bacterium Photobacterium profundum contain an operon
systems. encoding CusCBA but also a blue Cu protein in an operon
Overall, findings from recent years clearly point to a more regulated by a two component system. It is conceivable, that
prominent role of the biometal Cu for host–pathogen interaction the presence of these operons is in part protection against
than previously thought. Two bacterial model systems are most predatory amoeba since these organisms have also been found
advanced, S. enterica and M. tuberculosis each featuring unique in many marine environments including the Deep Sea.45
Cu homeostasis systems. In case of M. tuberculosis, a model of Moreover, there is strong evidence Acanthamoeba polyphaga
the events occurring during infection was suggested.36 Herein, is host to Vibrio cholera.46 Other operons prevalent in many
host and invader Cu homeostasis systems battle for the upper bacteria including various rhizobia encode an outer membrane
hand with the host also employing the adventitious properties of protein, a multicopper oxidase, an azurin-like blue Cu protein
redox-cycling between Cu’s different oxidation states. The and a CusF-like Cu chaperone.47 Interestingly, many of these
generated toxic reactive oxygen species would thus additionally bacteria such as Mesorhizobium amorphae and Bradyrhizobium
support killing of the bacterial pathogen. It appears that Cu japonicum are resistant to predation by amoeba.48
offers a weak point of defense in M. tuberculosis. Even though Since many of the processes involved in killing bacteria in
this bacterium expresses three potentially efficient Cu home- macrophages and amoeba are similar, the defense mechanisms
ostasis factors, its tolerance for Cu ions is rather low, about a that developed are manifold but there probably was extensive
factor of ten lower than in most bacteria including E. coli or pressure to possess an effective copper resistance system to
Salmonella.32,36 It is tempting to speculate that inhibition of a prolong survival and possibly overcome killing by amoeba and
factor such as MctB, which might be reached from the outside other predatory protozoa. In contrast to macrophages, it is at
of the cell, could lead to new strategies for anti-tuberculosis present not known whether amoeba use copper in their arsenal
chemotherapy. Recently, however, a potential exit strategy for to kill microorganisms, however, in our opinion it is likely
M. tuberculosis from such approaches was offered. A deletion that at least some predatory protozoa will also use copper,
of ricR, a paralog of the long known Cu regulator CsoR, since well studied Dictyostellium discoideum contains both
caused overproduction of MymT with a concomitant highly P80, a Ctr-type copper transporter localized in endocytic
increased resistance of M. tuberculosis against Cu ions.38 compartments involved in maturation of the phagosome,
However, it remains to be tested if such mutant cells can still and a Cu(I)-translocating P-type ATPase with a function in
efficiently invade and infect host cells. copper resistance.49

Metallomics This journal is c The Royal Society of Chemistry 2011


View Online
Published on 10 October 2011 on http://pubs.rsc.org | doi:10.1039/C1MT00107H
Downloaded by RSC Internal on 18 October 2011

Fig. 3 Role of Cu in the respiratory burst reaction within phagosomes. The respiratory burst reaction in activated macrophages and neutrophils
is a reactive oxygen species (ROS)-mediated antibacterial response. ROS production is metal-catalysed, with Fe(II) being considered the important
metal ion. Compelling new evidence indicates that Cu(I) also has an important role in this reaction. Cu(I) is trafficked in the cytoplasm of
eukaryotic cells by metallochaperone proteins to various intracellular sites of utilization. One such metallochaperone, Atox1, shuttles Cu(I) to a
Cu(I) transporter, ATP7A, which resides within the trans-Golgi network, where it provides Cu(I) for the maturation of secreted cuproenzymes. An
elevation in Cu levels within macrophages is achieved by enhanced activity of the high-affinity plasma membrane Cu(I) permease Ctr1. This results
in the relocation of ATP7A to both perinuclear vesicles and the phagosome. Delivery of Cu(I) to the phagosome might, in turn, enhance
bactericidal activity during the phagocytosis of bacteria by catalysing the formation of ROS (with permission from ref. 72).

Since Cu is such an effective means to kill bacteria not only cross-infection opportunities for pathogenic bacteria. Thus,
in macrophages but perhaps also in amoeba and in plants,50 contaminated surfaces contribute to an increase in healthcare-
it is perhaps not surprising that Cu has surfaced as new associated infections and costs.56 Furthermore, some patho-
antibacterial material as described in the following chapter. genic micro-organisms, such as the bacteria Staphylococcus
aureus or Acinetobacter ssp. and the yeast Candida albicans,
can contaminate and persist on surfaces in the hospital
Metallic copper as an antimicrobial agent
environment for months.57 Frequent and efficient removal of
Many historic cultures around the globe explored the utility of such organisms from surfaces combined with hand hygiene
Cu compounds for hygiene and health problems such as ulcer practice reduces such transmission opportunities.58 Problems
treatment, wound dressings or water preservation, taking arise when these active procedures are not properly followed.59
advantage of the antimicrobial properties of Cu without Self-sterilizing surfaces can be envisioned to help counter such
knowing the basis of these effects. This empirical knowledge shortcomings. In 2008 the Environmental Protection Agency
of certain transition metal cations, including Cu when in (EPA) registered Cu surfaces as antimicrobial (http://www.
surplus, exerting antibacterial properties was later described epa.gov/pesticides/factsheets/copper-alloy-products.htm).
as the ‘‘Oligodynamic Effect’’.51 In general, single-celled Thus, the use of anti-microbial metallic Cu surfaces is likely to
organisms are especially prone to Cu ion toxicity, enabling provide protection from infectious microbes by reducing
use of Cu as a selective agent against microbes without surface contamination as was recently shown in successful
harming animals or man.52 Even today, our understanding hospital trials.60 Importantly, bacteria in contact with dry Cu
of the molecular mode of action of Cu ions against bacteria is surfaces, in contrast to those exposed to Cu ions, do not
still limited and best understood in Escherichia coli.10,19,53 proliferate and most are killed within minutes.55
The antimicrobial efficacy of metallic Cu surfaces has been Unsurprisingly, there are similarities between the antimicrobial
established for a variety of bacteria.54 In these studies, cells in properties of ionic and metallic Cu: recent studies have
buffer were applied to Cu surfaces, incubated under ambient demonstrated that Cu ions are released from metallic Cu upon
conditions and cells were killed within hours. Recently, a contact with bacteria.53,55b,61 Further, extracellular supple-
method was established that aimed to mimic contact of mentation with protective substances against oxidative stress
microbes to dry Cu touch surfaces. Under these conditions such as catalase, superoxide dismutase or mannitol, a hydroxyl
most microbes are killed within minutes.55 Surfaces can be an radical quencher, increased the time needed to kill Cu surface-
important reservoir of bacterial contamination; especially the exposed E. coli cells.55b Similarly, Cu surfaces protected by
hospital environment provides multiple transmission and thermal oxide layers or the application of corrosion inhibitors

This journal is c The Royal Society of Chemistry 2011 Metallomics


View Online

prolonged the time needed for complete inactivation of 3 P. Gourdon, X. Y. Liu, T. Skjorringe, J. P. Morth, L. B. Moller,
Cu surface-exposed bacteria.62 We are only beginning to B. P. Pedersen and P. Nissen, Crystal structure of a copper-
transporting PIB-type ATPase, Nature, 2011, 475, 59–64.
comprehend the molecular mode-of-action exerted by Cu ions 4 (a) A. K. Mandal and J. M. Arguello, Functional roles of metal
on microbes.10 However, the specific kinds of stress exerted by binding domains of the Archaeoglobus fulgidus Cu(+)-ATPase
metallic Cu surfaces and the identity of sensitive cellular CopA, Biochemistry, 2003, 42, 11040–7; (b) M. Gonzalez-
targets are less understood and still under investigation. Recent Guerrero, E. Eren, S. Rawat, T. L. Stemmler and J. M. Arguello,
Structure of the two transmembrane Cu+ transport sites of the
studies suggest that cells are not killed via accumulation of Cu+ -ATPases, J. Biol. Chem., 2008, 283, 29753–9.
lethal mutations but rather that initially membranes are 5 C. J. De Feo, S. Mootien and V. M. Unger, Tryptophan scanning
damaged.53,63 This, in turn, also leads to DNA damage in analysis of the membrane domain of CTR-copper transporters,
J. Membr. Biol., 2010, 234, 113–23.
killed cells giving the impression of genotoxicity as the
6 M. A. Saito, D. M. Sigman and F. M. M. Morel, The bioinorganic
underlying mechanism of kill. Conversely, it is more likely chemistry of the ancient ocean: the co-evolution of cyanobacterial
that lipid peroxidation or irreversible inactivation of essential metal requirements and biogeochemical cycles at the Archean-
Published on 10 October 2011 on http://pubs.rsc.org | doi:10.1039/C1MT00107H

membrane proteins compromises membranes of Cu surface- Proterozoic boundary?, Inorg. Chim. Acta, 2003, 356, 308.
7 (a) A. G. Murzin, S. E. Brenner, T. Hubbard and C. Chothia,
exposed cells beyond the point of recovery. SCOP: a structural classification of proteins database for the
Cu nanoparticles are another type of antimicrobial metallic investigation of sequences and structures, J. Mol. Biol., 1995,
Cu surface that can, however, enter cells. Different species 247, 536–40; (b) M. Wang, L. S. Yafremava, D. Caetano-Anolles,
of bacteria64 have been subjected to nano Cu previously. J. E. Mittenthal and G. Caetano-Anolles, Reductive evolution of
architectural repertoires in proteomes and the birth of the tripartite
Concentrations of Cu nanoparticles (40–100 nm, Ø) required world, Genome Res., 2007, 17, 1572–85.
for 90% antimicrobial efficiency were in the low, microgram- 8 (a) C. L. Dupont, A. Butcher, R. E. Valas, P. E. Bourne and
Downloaded by RSC Internal on 18 October 2011

per-milliliter range for both E. coli and Bacillus subtilis,65 but the G. Caetano-Anolles, History of biological metal utilization
inferred through phylogenomic analysis of protein structures, Proc.
mechanism of action of nano Cu remains unknown. Natl. Acad. Sci. U. S. A., 2010, 107, 10567–72; (b) M. Wang,
Use of ionic Cu as a feed-supplement in piggeries66 has Y. Y. Jiang, K. M. Kim, G. Qu, H. F. Ji, J. E. Mittenthal,
resulted in the emergence of high-level Cu ion resistance H. Y. Zhang and G. Caetano-Anolles, A universal molecular clock
systems.67 Similarly, application of Cu-containing antifungal of protein folds and its power in tracing the early history of aerobic
metabolism and planet oxygenation, Mol. Biol. Evol., 2011, 28,
and antibacterial sprays in agriculture has selected for similar 567–82.
Cu ion resistance systems in plant-pathogenic bacteria.68 9 L. S. Busenlehner, M. A. Pennella and D. P. Giedroc, The SmtB/
Importantly, cells exposed to dry Cu surfaces are not in an ArsR family of metalloregulatory transcriptional repressors:
environment in which they can proliferate, in contrast, for Structural insights into prokaryotic metal resistance, FEMS
Microbiol. Rev., 2003, 27, 131–43.
example, to a Cu ion-treated animal gut,67a Cu catheters,69 Cu 10 L. Macomber and J. A. Imlay, The iron-sulfur clusters of
implants70 and Cu in other environments where biofilms may dehydratases are primary intracellular targets of copper toxicity,
form. Comparable studies on the long-term consequences of Proc. Natl. Acad. Sci. U. S. A., 2009, 106, 8344–9.
11 S. Chillappagari, A. Seubert, H. Trip, O. P. Kuipers,
exposure to metallic Cu on bacterial communities, however, M. A. Marahiel and M. Miethke, Copper stress affects iron
are lacking. homeostasis by destabilizing iron-sulfur cluster formation in
Most bacteria54 are metallic Cu surface sensitive and only a Bacillus subtilis, J. Bacteriol., 2010, 192, 2512–24.
few of the bacteria isolated from Cu coins survived prolonged 12 A. Changela, K. Chen, Y. Xue, J. Holschen, C. E. Outten,
T. V. O’Halloran and A. Mondragon, Molecular basis of
exposure to pure Cu.55a Remarkably, some coin-isolates metal-ion selectivity and zeptomolar sensitivity by CueR, Science,
survived from 16 times (Micrococcus luteus) up to 5760 times 2003, 301, 1383–7.
(Pseudomonas oleovorans) longer on pure Cu than their type 13 (a) B. Halliwell and J. M. Gutteridge, Oxygen toxicity, oxygen
radicals, transition metals and disease, Biochem. J., 1984, 219,
strains or controls but were not especially resistant to ionic Cu.
1–14; (b) B. Halliwell and J. M. Gutteridge, Role of free radicals
It is thus unlikely that Cu ion and Cu surface resistance are and catalytic metal ions human disease: an overview, Methods
governed by the same mechanisms because metallic Cu resistant Enzymol., 1990, 186, 1–85.
isolates from coins were sensitive to dissolved Cu ions.55a 14 (a) E. R. Stadtman, Protein oxidation and aging, Science, 1992,
257, 1220–4; (b) J. A. Imlay and S. Linn, DNA damage and oxygen
Further research is also required to investigate and address radical toxicity, Science, 1988, 240, 1302–9.
the likelihood of the possible emergence and spread of Cu 15 (a) K. Helbig, C. Bleuel, G. J. Krauss and D. H. Nies, Glutathione
surface resistant bacteria. and transition-metal homeostasis in Escherichia coli, J. Bacteriol.,
2008, 190, 5431–8; (b) S. J. Stohs and D. Bagchi, Oxidative
mechanisms in the toxicity of metal ions, Free Radical Biol.
Acknowledgements Med., 1995, 18, 321–36.
16 K. Keyer and J. A. Imlay, Superoxide accelerates DNA damage by
CLD was supported by a NASA Astrobiology Institute DDF. elevating free-iron levels, Proc. Natl. Acad. Sci. U. S. A., 1996, 93,
13635–40.
Research in the laboratories of CR and GG was supported by 17 F. W. Outten, D. L. Huffman, J. A. Hale and T. V. O’Halloran,
grants from the International Copper Association. The independent cue and cus systems confer copper tolerance
during aerobic and anaerobic growth Escherichia coli, J. Biol.
Chem., 2001, 276, 30670–7.
References 18 L. Macomber, C. Rensing and J. A. Imlay, Intracellular copper
does not catalyze the formation of oxidative DNA damage in
1 R. G. Pearson, Hard and soft acids and bases, J. Am. Chem. Soc., Escherichia coli, J. Bacteriol., 2007, 189, 1616–26.
1963, 85, 3533–3539. 19 C. Rensing and G. Grass, Escherichia coli mechanisms of copper
2 F. Long, C. C. Su, M. T. Zimmermann, S. E. Boyken, homeostasis in a changing environment, FEMS Microbiol. Rev.,
K. R. Rajashankar, R. L. Jernigan and E. W. Yu, Crystal 2003, 27, 197–213.
structures of the CusA efflux pump suggest methionine-mediated 20 (a) A. Odermatt, H. Suter, R. Krapf and M. Solioz, Primary
metal transport, Nature, 2010, 467, 484–8. structure of two P-type ATPases involved in copper homeostasis

Metallomics This journal is c The Royal Society of Chemistry 2011


View Online

in Enterococcus hirae, J. Biol. Chem., 1993, 268, 12775–9; 38 R. A. Festa, M. B. Jones, S. Butler-Wu, D. Sinsimer, R. Gerads,
(b) C. Rensing, B. Fan, R. Sharma, B. Mitra and B. P. Rosen, W. R. Bishai, S. N. Peterson and K. H. Darwin, A novel copper-
CopA: An Escherichia coli Cu(I)-translocating P-type ATPase, responsive regulon in Mycobacterium tuberculosis, Mol. Microbiol.,
Proc. Natl. Acad. Sci. U. S. A., 2000, 97, 652–6. 2011, 79, 133–48.
21 S. Franke, G. Grass, C. Rensing and D. H. Nies, Molecular 39 (a) H. Fischer, M. Behrens, M. Bock, U. Richter, J. Schmitt,
analysis of the copper-transporting efflux system CusCFBA of L. Loulergue, J. Chappellaz, R. Spahni, T. Blunier,
Escherichia coli, J. Bacteriol., 2003, 185, 3804–12. M. Leuenberger and T. F. Stocker, Changing boreal methane
22 M. A. Seeger, A. Schiefner, T. Eicher, F. Verrey, K. Diederichs and sources and constant biomass burning during the last termination,
K. M. Pos, Structural asymmetry of AcrB trimer suggests a Nature, 2008, 452, 864–7; (b) L. Rink and P. Gabriel, Extracellular
peristaltic pump mechanism, Science, 2006, 313, 1295–8. and immunological actions of zinc, BioMetals, 2001, 14, 367–83.
23 E. H. Kim, D. H. Nies, M. M. McEvoy and C. Rensing, Switch or 40 H. W. Kim, Q. Chan, S. E. Afton, J. A. Caruso, B. Lai,
funnel: how RND-type transport systems control periplasmic N. L. Weintraub and Z. Qin, Human macrophage ATP7A is
metal homeostasis, J. Bacteriol., 2011, 193, 2381–7. localized in the trans-Golgi apparatus, controls intracellular
24 (a) S. K. Singh, G. Grass, C. Rensing and W. R. Montfort, copper levels, and mediates macrophage responses to dermal
Cuprous oxidase activity of CueO from Escherichia coli, wounds, Inflammation, 2011, DOI: 10.1007/s10753-011-9302-z.
Published on 10 October 2011 on http://pubs.rsc.org | doi:10.1039/C1MT00107H

J. Bacteriol., 2004, 186, 7815–7; (b) G. Grass and C. Rensing, 41 S. Shafeeq, H. Yesilkaya, T. G. Kloosterman, G. Narayanan,
CueO is a multi-copper oxidase that confers copper tolerance in P. W. Andrew, O. P. Kuipers and J. A. Morrissey, The cop operon
Escherichia coli, Biochem. Biophys. Res. Commun., 2001, 286, is required for copper homeostasis and contributes to virulence in
902–8. Streptococcus pneumoniae, Mol. Microbiol., 2011, DOI: 10.1111/
25 G. Grass, K. Thakali, P. E. Klebba, D. Thieme, A. Müller, j.1365-2958.2011.07758.x.
G. F. Wildner and C. Rensing, Linkage between catecholate 42 E. B. Sherr and B. F. Sherr, Significance of predation by protists in
siderophores and the multicopper oxidase CueO in Escherichia aquatic microbial food webs, Antonie van Leeuwenhoek, 2002, 81,
coli, J. Bacteriol., 2004, 186, 5826–33. 293–308.
26 L. B. Pontel and F. C. Soncini, Alternative periplasmic copper- 43 A. Rogerson, O. R. Anderson and C. Vogel, Are planktonic naked
Downloaded by RSC Internal on 18 October 2011

resistance mechanisms in Gram negative bacteria, Mol. Microbiol., amoeba predominantly floc associated or free in the water column,
2009, 73, 212–25. J. Plankton Res., 2003, 25, 1359.
27 D. Osman, K. J. Waldron, H. Denton, C. M. Taylor, A. J. Grant, 44 D. A. Brock, T. E. Douglas, D. C. Queller and J. E. Strassmann,
P. Mastroeni, N. J. Robinson and J. S. Cavet, Copper homeostasis Primitive agriculture in a social amoeba, Nature, 2011, 469, 393–6.
in Salmonella is atypical and copper-CueP is a major periplasmic 45 P. Lopez-Garcia, H. Philippe, F. Gail and D. Moreira, Auto-
metal complex, J. Biol. Chem., 2010, 285, 25259–68. chthonous eukaryotic diversity in hydrothermal sediment and
28 L. B. Pontel, M. E. Audero, M. Espariz, S. K. Checa and experimental microcolonizers at the Mid-Atlantic Ridge, Proc.
F. C. Soncini, GolS controls the response to gold by the hierarchical Natl. Acad. Sci. U. S. A., 2003, 100, 697–702.
induction of Salmonella-specific genes that include a CBA 46 G. Sandstrom, A. Saeed and H. Abd, Acanthamoeba polyphaga is a
efflux-coding operon, Mol. Microbiol., 2007.
possible host for Vibrio cholerae in aquatic environments, Exp.
29 S. K. Checa, M. Espariz, M. E. Audero, P. E. Botta, S. V. Spinelli
Parasitol., 2010, 126, 65–8.
and F. C. Soncini, Bacterial sensing of and resistance to gold salts,
47 E. H. Kim, C. Rensing and M. M. McEvoy, Chaperone-mediated
Mol. Microbiol., 2007, 63, 1307–18.
copper handling in the periplasm, Nat. Prod. Rep., 2010, 27, 711–9.
30 C. White, J. Lee, T. Kambe, K. Fritsche and M. J. Petris, A role for
48 G. Greub and D. Raoult, Microorganisms resistant to free-living
the ATP7A copper-transporting ATPase in macrophage bactericidal
amoebae, Clin. Microbiol. Rev., 2004, 17, 413–33.
activity, J. Biol. Chem., 2009, 284, 33949–56.
49 (a) B. Burlando, V. Evangelisti, F. Dondero, G. Pons,
31 M. E. Achard, J. J. Tree, J. A. Holden, K. R. Simpfendorfer,
J. Camakaris and A. Viarengo, Occurrence of Cu-ATPase in
O. L. Wijburg, R. A. Strugnell, M. A. Schembri, M. J. Sweet,
Dictyostelium: possible role in resistance to copper, Biochem.
M. P. Jennings and A. G. McEwan, The multi-copper-ion oxidase
Biophys. Res. Commun., 2002, 291, 476–83; (b) M. Hagedorn and
CueO of Salmonella enterica serovar Typhimurium is required for
systemic virulence, Infect. Immun., 78, 2312–9. T. Soldati, Flotillin and RacH modulate the intracellular immunity
32 (a) G. Grass and C. Rensing, Genes involved in copper homeostasis of Dictyostelium to Mycobacterium marinum infection, Cell.
in Escherichia coli, J. Bacteriol., 2001, 183, 2145–7; (b) J. J. Tree, Microbiol., 2007, 9, 2716–33; (c) K. Ravanel, B. de Chassey,
G. C. Ulett, J. L. Hobman, C. Constantinidou, N. L. Brown, S. Cornillon, M. Benghezal, L. Zulianello, L. Gebbie,
C. Kershaw, M. A. Schembri, M. P. Jennings and A. G. McEwan, F. Letourneur and P. Cosson, Membrane sorting in the endocytic
The multicopper oxidase (CueO) and cell aggregation in Escherichia and phagocytic pathway of Dictyostelium discoideum, Eur. J. Cell
coli, Environ. Microbiol., 2007, 9, 2110–6; (c) L. B. Pontel, A. Pezza Biol., 2001, 80, 754–64.
and F. C. Soncini, Copper stress targets the Rcs system to induce 50 M. Yuan, Z. Chu, X. Li, C. Xu and S. Wang, The bacterial
multiaggregative behavior in a copper-sensitive Salmonella strain, pathogen Xanthomonas oryzae overcomes rice defenses by regulating
J. Bacteriol., 2010, 192, 6287–90. host copper redistribution, Plant Cell, 2010, 22, 3164–76.
33 S. Rankin, Z. Li and R. R. Isberg, Macrophage-induced genes of 51 C. W. von Nägeli, Über oligodynamische Erscheinungen in
Legionella pneumophila: protection from reactive intermediates and lebenden Zellen, Neue Denkschriften der schweizerischen naturforschenden
solute imbalance during intracellular growth, Infect. Immun., 2002, Gesellschaft, 1893, 33, 1–51.
70, 3637–48. 52 H. Kraemer, The oligodynamic action of copper foil on certain
34 W. R. Schwan, P. Warrener, E. Keunz, C. K. Stover and K. R. Folger, intestinal organisms, Proc. Amer. Philos. Soc., 1905, 44, 51–65.
Mutations in the cueA gene encoding a copper homeostasis P-type 53 C. Espirito Santo, E. W. Lam, C. G. Elowsky, D. Quaranta,
ATPase reduce the pathogenicity of Pseudomonas aeruginosa in mice, D. W. Domaille, C. J. Chang and G. Grass, Bacterial killing by
Int. J. Med. Microbiol., 2005, 295, 237–42. dry metallic copper surfaces, Appl. Environ. Microbiol., 2011, 77,
35 S. K. Ward, B. Abomoelak, E. A. Hoye, H. Steinberg and 794–802.
A. M. Talaat, CtpV: a putative copper exporter required for full 54 (a) J. O. Noyce, H. Michels and C. W. Keevil, Potential use of
virulence of Mycobacterium tuberculosis, Mol. Microbiol., 2010, 77, copper surfaces to reduce survival of epidemic meticillin-resistant
1096–110. Staphylococcus aureus in the healthcare environment, J. Hosp.
36 F. Wolschendorf, D. Ackart, T. B. Shrestha, L. Hascall-Dove, Infect., 2006, 63, 289–97; (b) S. Mehtar, I. Wiid and
S. Nolan, G. Lamichhane, Y. Wang, S. H. Bossmann, S. D. Todorov, The antimicrobial activity of copper and copper
R. J. Basaraba and M. Niederweis, Copper resistance is essential alloys against nosocomial pathogens and Mycobacterium tuberculosis
for virulence of Mycobacterium tuberculosis, Proc. Natl. Acad. Sci. isolated from healthcare facilities in the Western Cape: an in vitro
U. S. A., 2011, 108, 1621–6. study, J. Hosp. Infect., 2007, 68, 45–51.
37 B. Gold, H. Deng, R. Bryk, D. Vargas, D. Eliezer, J. Roberts, 55 (a) C. Espirito Santo, P. V. Morais and G. Grass, Isolation and
X. Jiang and C. Nathan, Identification of a copper-binding characterization of bacteria resistant to metallic copper surfaces,
metallothionein in pathogenic mycobacteria, Nat. Chem. Biol., Appl. Environ. Microbiol., 2010, 76, 1341–1348; (b) C. Espirito
2008, 4, 609–16. Santo, N. Taudte, D. H. Nies and G. Grass, Contribution of

This journal is c The Royal Society of Chemistry 2011 Metallomics


View Online

copper ion resistance to survival of Escherichia coli on metallic and copper nanoparticles, Acta Biomater., 2008, 4, 707–716;
copper surfaces, Appl. Environ.Microbiol., 2008, 74, 977–86. (b) F. Rispoli, A. Angelov, D. Badia, A. Kumar, S. Seal and
56 (a) B. Hota, Contamination, disinfection, and cross-colonization: V. Shah, Understanding the toxicity of aggregated zero valent
are hospital surfaces reservoirs for nosocomial infection?, Clin. copper nanoparticles against Escherichia coli, J. Hazard. Mater.,
Infect. Dis., 2004, 39, 1182–9; (b) S. S. Huang, R. Datta and 2010, 180, 212–216.
R. Platt, Risk of acquiring antibiotic-resistant bacteria from prior 65 K. Y. Yoon, J. Hoon Byeon, J. H. Park and J. Hwang, Susceptibility
room occupants, Arch. Intern. Med., 2006, 166, 1945–51. constants of Escherichia coli and Bacillus subtilis to silver and copper
57 A. Kramer, I. Schwebke and G. Kampf, How long do nosocomial nanoparticles, Sci. Total Environ., 2006, 373, 572–5.
pathogens persist on inanimate surfaces? A systematic review, 66 G. C. Shurson, P. K. Ku, G. L. Waxler, M. T. Yokoyama and
BMC Infect. Dis., 2006, 6, 130. E. R. Miller, Physiological relationships between microbiological
58 D. Pittet, B. Allegranzi and J. Boyce, The World Health Organization status and dietary copper levels in the pig, J. Anim. Sci., 1990, 68,
guidelines on hand hygiene in health care and their consensus 1061–71.
recommendations, Infect. Control Hosp. Epidemiol., 2009, 30, 611–22. 67 (a) T. J. Tetaz and R. K. Luke, Plasmid-controlled resistance to
59 S. J. Dancer, Importance of the environment in meticillin-resistant copper in Escherichia coli, J. Bacteriol., 1983, 154, 1263–8;
Staphylococcus aureus acquisition: the case for hospital cleaning, (b) S. M. Lee, G. Grass, C. Rensing, S. R. Barrett, C. J. Yates,
Lancet Infect. Dis., 2008, 8, 101–13. J. V. Stoyanov and N. L. Brown, The Pco proteins are involved in
Published on 10 October 2011 on http://pubs.rsc.org | doi:10.1039/C1MT00107H

60 (a) A. L. Casey, D. Adams, T. J. Karpanen, P. A. Lambert, periplasmic copper handling in Escherichia coli, Biochem. Biophys.
B. D. Cookson, P. Nightingale, L. Miruszenko, R. Shillam, Res. Commun., 2002, 295, 616–20.
P. Christian and T. S. J. Elliott, Role of copper in reducing hospital 68 A. E. Voloudakis, C. L. Bender and D. A. Cooksey, Similarity
environment contamination, J. Hosp. Infect., 2010, 74, 72–7; between copper resistance genes from Xanthomonas campestris and
(b) A. Mikolay, S. Huggett, L. Tikana, G. Grass, J. Braun and Pseudomonas syringae, Appl. Environ. Microbiol., 1993, 59,
D. H. Nies, Survival of bacteria on metallic copper surfaces in a 1627–1634.
hospital trial, Appl. Microbiol. Biotechnol., 2010, 87, 1875–1879. 69 R. J. McLean, A. A. Hussain, M. Sayer, P. J. Vincent,
61 C. Molteni, H. K. Abicht and M. Solioz, Killing of bacteria by D. J. Hughes and T. J. Smith, Antibacterial activity of multilayer
Downloaded by RSC Internal on 18 October 2011

copper surfaces involves dissolved copper, Appl. Environ. Micro- silver-copper surface films on catheter material, Can. J. Microbiol.,
biol., 2010, 76, 4099–101. 1993, 39, 895–9.
62 J. Elguindi, S. Moffitt, H. Hasman, C. Andrade, S. Raghavan and 70 T. Shirai, H. Tsuchiya, T. Shimizu, K. Ohtani, Y. Zen and
C. Rensing, Metallic copper corrosion rates, moisture content, and K. Tomita, Prevention of pin tract infection with titanium-copper
growth medium influence survival of copper ion-resistant bacteria, alloys, J. Biomed. Mater. Res. B Appl. Biomater., 2009, 91, 373–80.
Appl. Microbiol. Biotechnol., 2011, 89, 1963–70. 71 C. L. Dupont, S. Yang, B. Palenik and P. E. Bourne, Modern
63 D. Quaranta, T. Krans, C. Espirito Santo, C. G. Elowsky, proteomes contain putative imprints of ancient shifts in trace metal
D. W. Domaille, C. J. Chang and G. Grass, Mechanisms of yeast geochemistry, Proc. Natl. Acad. Sci. U S A, 2006, 103, 17822–7.
contact-killing on dry metallic copper surfaces, Appl. Environ. 72 S. C. Leary and D. R. Winge, The Janus face of copper: its
Microbiol., 2011, 77, 416–426. expanding roles in biology and the pathophysiology of disease.
64 (a) J. P. Ruparelia, A. K. Chatterjee, S. P. Duttagupta and Meeting on Copper and Related Metals in Biology, EMBO Rep.,
S. Mukherji, Strain specificity in antimicrobial activity of silver 2007, 8, 224–7.

Metallomics This journal is c The Royal Society of Chemistry 2011

View publication stats

You might also like