ArticlePDF AvailableLiterature Review

Ferrous Iron Efflux Systems in Bacteria

Authors:

Abstract

Bacteria require iron for growth, with only a few reported exceptions. In many environments, iron is a limiting nutrient for growth and high affinity uptake systems play a central role in iron homeostasis. However, iron can also be detrimental to cells when it is present in excess, particularly under aerobic conditions where its participation in Fenton chemistry generates highly reactive hydroxyl radicals. Recent results have revealed a critical role for iron efflux transporters in protecting bacteria from iron intoxication. Systems that efflux iron are widely distributed amongst bacteria and fall into several categories: P1B-type ATPases, cation diffusion facilitator (CDF) proteins, major facilitator superfamily (MFS) proteins, and membrane bound ferritin-like proteins. Here, we review the emerging role of iron export in both iron homeostasis and as part of the adaptive response to oxidative stress.
Ferrous Iron Efflux Systems in Bacteria
Hualiang Pi and John D. Helmann*
Abstract
Bacteria require iron for growth, with only a few reported exceptions. In many environments, iron
is a limiting nutrient for growth and high affinity uptake systems play a central role in iron
homeostasis. However, iron can also be detrimental to cells when it is present in excess,
particularly under aerobic conditions where its participation in Fenton chemistry generates highly
reactive hydroxyl radicals. Recent results have revealed a critical role for iron efflux transporters in
protecting bacteria from iron intoxication. Systems that efflux iron are widely distributed amongst
bacteria and fall into several categories: P1B-type ATPases, cation diffusion facilitator (CDF)
proteins, major facilitator superfamily (MFS) proteins, and membrane bound ferritin-like proteins.
Here, we review the emerging role of iron export in both iron homeostasis and as part of the
adaptive response to oxidative stress.
Graphical abstract
Introduction
Iron is critical for cell growth and survival. However, when present in excess, it is also
detrimental to cells. Under aerobic conditions, iron toxicity is closely related to oxidative
stress through Fenton chemistry1. Hydrogen peroxide (H2O2) reacts with ferrous iron (Fe2+)
to generate highly reactive hydroxyl radicals that damage macromolecules such as DNA,
proteins and fatty acids, resulting in disruption of cell metabolism and ultimately cell death2.
Therefore, the toxicity of reactive oxygen species (ROS) is generally thought to be
exacerbated by conditions that elevate the intracellular iron pool. Conversely, high levels of
intracellular iron may also be toxic independent of ROS, presumably due to the ability of
*Corresponding author: John D. Helmann, Department of Microbiology, Cornell University, Ithaca, NY 14853-8101, USA, Phone:
607-255-1517, Fax: 607-255-3904, jdh9@cornell.edu.
HHS Public Access
Author manuscript
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Published in final edited form as:
Metallomics
. 2017 July 19; 9(7): 840–851. doi:10.1039/c7mt00112f.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
iron to compete with other transition metals, such as manganese, for binding to metal-
dependent enzymes or regulators, resulting in mismetallation and inactivation of these
proteins3, 4. ROS such as H2O2 and superoxide radical can disrupt iron-sulfur clusters and
mononuclear iron centers of iron-enzymes, thereby leading to iron release5, 6. Therefore,
iron intoxication may also be exacerbated by an elevation in ROS. Clearly, the toxicity of
iron and ROS are closely intertwined, with each potentially increasing the toxicity of the
other.
Bacteria adapt to environmental stresses by activation of specific transcriptional programs.
In the case of iron homeostasis, bacteria monitor intracellular iron levels using metal-sensing
(metalloregulatory) proteins7, 8. The ferric uptake regulator (Fur) protein is the most
widespread bacterial iron sensor9, but it can be replaced by functionally analogous proteins
such as IdeR (in actinomycetes)10, 11 and Irr (in alpha-proteobacteria)12–14. Fur helps to
maintain iron homeostasis by regulating genes implicated in iron uptake, storage, and
efflux15. Typically, Fur is considered to function as an Fe2+-activated transcriptional
repressor for most of its targets, but there are increasing examples where Fur functions as a
transcriptional activator or where it binds DNA in the absence of bound iron16–18.
Iron-sensing regulators such as Fur play a central role in the control of iron homeostasis19.
The
Escherichia coli
Fur regulon illustrates the diverse roles that Fur may play.
E. coli
Fur
(FurEC) binds to DNA when associated with Fe2+ and serves to repress the expression of
target operons20. This repression is relieved under iron-limited conditions, and this results in
the derepression of iron uptake systems, including the synthesis of the high-affinity iron-
chelating compound siderophore known as enterobactin and its cognate import system21.
Fur also helps bacteria to remodel their proteomes to prioritize the utilization of iron, in a
process known as "iron-sparing" (Fig. 1)22–24. In
E. coli
, the loss of FurEC DNA-binding
activity (under low iron conditions) results in expression of the RyhB small RNA (sRNA)
that represses translation of non-essential iron-enzymes22–24. Fur also participates in the
regulation of gene expression under conditions of iron excess. For example, FurEC positively
regulates expression of the iron storage protein ferritin by occluding the binding of the H-NS
transcriptional repressor25. In general, adaptation to iron excess often involves expression of
iron storage functions (including heme-containing bacterioferritins, ferritins, and Dps-family
mini-ferritins) but may additionally require iron efflux systems (Fig. 1). In light of the
central role of Fur in coordinating iron homeostasis, it is not surprising that some iron efflux
systems are induced by Fur in response to iron excess26, 27.
Bacteria also adapt to oxidative stress by the induction of specific defensive genes. For
example, H2O2 induces a specific peroxide-stress response that is regulated by the OxyR
repressor in
E. coli
28 and by the PerR repressor in
Bacillus subtilis
29. In both model
organisms, a rise in intracellular H2O2 triggers the induction of defensive enzymes such as
catalase and alkyl hydroperoxide reductase, which can directly detoxify H2O2. In addition,
cells scavenge excess iron from the cytosol by sequestration into mini-ferritin proteins,
including Dps in
E. coli
30 and the Dps ortholog MrgA in
B. subtilis
31. The co-regulation of
H2O2 degradation enzymes and iron-sequestering proteins further highlights the central role
of iron in peroxide intoxication. In addition to scavenging iron, peroxide stress also
frequently modulates metal uptake and efflux systems32. In
E. coli
, H2O2 induces an OxyR-
Pi and Helmann Page 2
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
activated Mn2+ uptake system (MntH)33, 34, and in
B. subtilis
H2O2 induces a PerR-
regulated iron efflux system, PfeT35, 36. PfeT is a member of the P1B4-type ATPases, and
recent results indicate that several close homologs also function as Fe2+ efflux
pumps27, 37–39. Fe2+ efflux pumps have now been documented in a wide variety of bacteria,
and include P1B-type ATPases, cation diffusion facilitator (CDF) proteins, major facilitator
superfamily (MFS) proteins, and membrane bound ferritin-like proteins (Table 1 & Fig. 2).
Here, we summarize the emerging role of these ferrous iron efflux pumps in helping
ameliorate the deleterious effects of excess iron and peroxide.
P-type ATPases
The P-type ATPases are a large group of transmembrane proteins that transport ions and
lipids across cellular membranes, energetically driven by ATP hydrolysis40. Five subgroups
of P-type ATPases have been defined based on sequence homology and substrate
specificity41. These are the P1-type (K+ and transition metal transporters), P2-type (Ca2+,
Na+/K+, and H+/K+ pumps), P3-type (H+ pumps), P4-type (phospholipid transporters), and
P5-type ATPases (unknown substrate). The P2-type ATPases have been well studied and are
more prevalent in eukaryotes than in prokaryotes. The majority of P3-type ATPases are H+
pumps found in plants and fungi. Some of the P4-type ATPases have been revealed to be
phospholipid transporters42, 43. No specific substrate has yet been identified for the P5-type
ATPases that are only found in eukaryotes.
The P1-type ATPases exist predominately in prokaryotes but are omnipresent across all
domains of life44: P1A-ATPases are involved in K+ transport whereas P1B-ATPases are
important for maintaining transition metal homeostasis. P1B-ATPases are known to transport
Cu+ 45, 46, Ag+ 47, Zn2+ 48, Cd2+ 49, Cu2+ 50, Co2+ 51 and Fe2+ 27, 36, 37. The structure of a
typical P1B-ATPase includes a transmembrane domain with 6–8 helices, a soluble actuator
domain, and an ATP-binding domain52 (Fig. 2). The P1B-ATPases can be further divided
into seven subclasses based on sequence similarity and metal substrate specificity52. The
P1B4-type ATPases were originally assigned a role in Co2+ export, based on the properties of
some of the first characterized members53. However, P1B4-type ATPases have recently been
found to function instead, or in addition, as Fe2+ efflux transporters including
Bacillus
subtilis
PfeT36,
Listeria monocytogenes
FrvA27,
Mycobacterium tuberculosis
CtpD37, and
group A
Streptococcus
PmtA38, 39.
PfeT in Bacillus subtilis
B. subtilis
is a Gram-positive soil microorganism and encodes two transcriptional regulators
critical for iron homeostasis, FurBs and PerR. FurBs is a global transcriptional regulator of
iron homeostasis analogous to FurEC54 and PerR mediates the adaptive response to peroxide
stress by regulating genes involved in iron storage and peroxide detoxification29. The
regulons for both FurBs and PerR have been well defined55, 56. FurBs senses intracellular
iron sufficiency and represses genes that are involved in siderophore synthesis and
uptake54, 57. FurBs also regulates an iron sparing response mediated by the small non-coding
RNA FsrA (Fig. 1) and its coregulators FbpA, FbpB and FbpC58–60. This system, analogous
to RyhB in
E. coli
, blocks the translation of non-essential iron-containing enzymes such as
Pi and Helmann Page 3
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
aconitase and succinate dehydrogenase58–60. PerR regulates peroxide detoxification
enzymes (catalase, alkyl hydroperoxide reductase), iron sequestration (MrgA) and the P1B4-
type ATPase (PfeT). Although the Fur and PerR regulons are largely non-overlapping,
pfeT
is the exception and is regulated by both proteins26. The result is that
pfeT
is induced by
either peroxide stress or by iron excess (unpublished data, Pinochet-Barros A & Helmann
JD).
PfeT is one of three P1B ATPases encoded by
B. subtilis
. CopA is a P1B1-ATPase that
functions as a Cu+ efflux transporter and, appropriate to its function, is regulated by the
CsoR Cu+ sensor61. CadA is a P1B2-ATPase that confers resistance to Cd2+, Zn2+, and Co2+
and is regulated by the divalent cation sensor CzrA62. PfeT (formerly named as ZosA) is a
P1B4-type ATPase and was discovered as a transporter induced by H2O2 that plays a role in
protecting cells against oxidative stress35. Initial results indicated that deletion of
pfeT
enhanced Zn2+ tolerance, as monitored in cells lacking the CadA efflux system35. This led to
the proposal that PfeT might function as a Zn2+ importer under oxidative stress conditions,
consistent with the idea that Zn2+ has a role in protecting cells against oxidative damage35.
As a result, PfeT was originally named for this proposed role as ZosA (Zn2+ uptake under
oxidative stress)35.
Contrary to this model, most P1B-type ATPases function in metal export rather than import,
which motivated a reinvestigation of the role of PfeT. Further study revealed that a
pfeT
null
mutant is sensitive to Fe2+ and Fe3+, particularly under acidic media conditions, but not to
Zn2+ or Co2+. Moreover, a
pfeT
null mutant accumulates elevated levels of intracellular
Fe2+, as judged by sensitivity to the Fe2+-activated antibiotic streptonigrin and by direct
chemical measurement36. Biochemical studies confirmed that the ATPase activity of PfeT is
induced the most by Fe2+, with modest induction by Co2+ but not with other metals,
including Zn2+. In addition to H2O2,
pfeT
is strongly and specifically induced by iron, but
not by other metals. Together, these findings indicate that PfeT function as a peroxide- and
iron induced ferrous efflux transporter36. The ability of PfeT to protect against H2O2 is
secondary to that of the detoxification enzymes catalase and alkyl hydroperoxide reductase.
However, PfeT plays a dominant role in protecting cells from iron overload with the MrgA
miniferritin playing a secondary role36. The revelation that PfeT functions in Fe2+ efflux, in
turn, prompted a re-evaluation of the roles of several other P1B4-type ATPases in bacterial
iron homeostasis.
FrvA in Listeria monocytogenes
L. monocytogenes
is the causative agent of the foodborne disease listeriosis, which is
associated with central nervous system infections and bacteraemia. FrvA (Lmo0641) is a
P1B4-ATPase originally described as a Fur-regulated virulence factor63. FrvA was proposed
to function as a heme exporter that was suggested to be induced by iron deficiency and to be
under negative regulation of both Fur and PerR63, 64. However, a different transcriptome
study showed a downregulation of
frvA
in a
fur
null mutant65, indicating a positive
regulatory role of Fur in
frvA
expression.
To resolve these contradictory reports of iron regulation, and to test if FrvA might function
in Fe2+ efflux, the mutant phenotype was reinvestigated and the FrvA protein was purified
Pi and Helmann Page 4
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
for biochemical studies27. As predicted based on studies of
pfeT
, a
frvA
null mutant was
sensitive to iron intoxication, but not to other metals or heme. Like
B. subtilis pfeT, frvA
is
positively regulated by Fur in response to high Fe2+ levels and is repressed by PerR27, 64.
Biochemical studies indicate the FrvA ATPase activity is stimulated most strongly by Fe2+
with weaker stimulation in the presence of Co2+ or Zn2+. Based on the Fe2+ concentration
dependence of ATPase activity, FrvA seems to have a higher affinity for Fe2+ than
B. subtilis
PfeT. Consistent with this, not only does FrvA complement the iron-sensitive phenotype of a
B. subtilis pfeT
null mutant, its expression depletes the cytosol of iron (even under iron-rich
conditions) thereby leading to derepression of the Fur regulon27. These results support the
hypothesis that FrvA functions as a Fe2+ efflux transporter that protects cells from Fe2+
intoxication27.
FrvA is required for virulence in murine and insect (
Galleria mellonella
) infection models63.
The
frvA
null mutant strain shows strong attenuation in virulence, but is still able to invade
and propagate inside antigen-presenting cells66, suggesting an important link between iron
homeostasis and virulence, but it is not clear at which stage(s) of the
L. monocytogenes
life
cycle FrvA is important. The phagocytic vacuole is generally considered to be an iron-
limited environment. One possibility is that the expression of high affinity iron uptake
systems by iron limitation during infection or in the phagocytic vacuole can contribute to
iron overload upon escape of cells into the relatively iron-rich cytosol. Alternatively, the
imposition of oxidative damage from host immune cells may trigger iron release from
listerial iron enzymes and this may lead to iron overload. The points in the infection cycle
where FrvA plays a critical role are not yet clearly defined and provide an interesting avenue
for future research.
CtpD in Mycobacterium tuberculosis
M. tuberculosis
is an obligate pathogen and the causative agent of human tuberculosis.
Nearly one-third of the world's population is infected with
M. tuberculosis
, which can persist
in a latent state for decades and then later emerge (in ~10% of cases) as an active lung
infection.
M. tuberculosis
encodes a total of 11 P-type ATPases, which have been suggested
to be possible targets for therapeutic intervention67. Of these, two encode P1B4-ATPases:
CtpD (Rv1469) and CtpJ (Rv3743)37. CtpD, but not CtpJ, was found to be important for
survival in macrophages and the mouse lung37. Biochemical studies had previously
highlighted the activity of these two P1B4-ATPases with Co2+, but it was not clear why
M.
tuberculosis
would encode two such proteins, nor was it understood why Co2+ efflux would
be important for survival in the host.
In light of the finding that PfeT functions as an Fe2+ efflux transporter, the roles of CtpD and
CtpJ were reinvestigated. Biochemical studies indicated that the ATPase activity of CtpD is
most strongly activated by Fe2+. Although Co2+ also activates ATPase activity, the maximal
activity (Vmax) is 10-fold lower than with ferrous iron37. CtpD also binds Fe2+ with 3-fold
higher affinity than Co2+. In contrast, the CtpJ ATPase activity is activated by both Fe2+ and
Co2+, and has a slightly higher affinity for Co2+ than Fe2+. To better understand their roles
in vivo, metal accumulation and sensitivity was monitored for strains lacking either
ctpD
or
ctpJ
. The
ctpD
mutant strain did not accumulate Co2+ and was impaired in growth in iron-
Pi and Helmann Page 5
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
amended medium, consistent with a primary role in resistance to iron intoxication37.
Mutation of
ctpJ
led to a significant increase in Co2+ accumulation and expression was
induced by Co2+, consistent with a primary role in Co2+ resistance37. However, the
ctpJ
mutant was also growth impaired in the presence of excess iron. Thus, these two paralogous
transporters seem to have overlapping metal selectivity, but largely distinct physiological
roles. Further studies are needed to understand the molecular mechanism of substrate
specificity, but based on X-ray absorption spectroscopy (XAS) analysis, it is likely that
distinct metal coordination geometry plays an important role37.
During infection,
M. tuberculosis
propagates in the host macrophages, which are considered
iron-poor environments. Just as noted for
L. monocytogenes
, it is not yet clear where in the
infection process cells experience iron intoxication. Further studies are needed to better
understand the conditions that lead to induction of
ctpD
. In prior work
ctpD
was not induced
by metals such as Co2+, Zn2+, and Ni2+, but its cognate substrate Fe2+ was not tested68. It
might be induced by Fe2+ and, by analogy with its orthologs, this might involve an iron-
sensing transcription factor. IdeR, a member of DtxR family, is the major iron-dependent
transcriptional regulator in
M. tuberculosis
10, 11. IdeR represses transcription of genes
involved in iron uptake and siderophore biosynthesis and activates expression of genes
encoding iron-storage proteins such as bacterioferritin and a ferritin-like protein10, 11. Since
M. tuberculosis
is primarily a pathogen of the mammalian respiratory system it might
frequently encounter oxidative stress. Thus, it is also possible that
ctpD
might be induced in
response to H2O2 stress. Future work to monitor the expression of
ctpD
in vitro in response
to specific stresses and in vivo during the course of infection will be needed to elucidate the
physiological role of CtpD during the infection process.
PmtA in group A Streptococcus
Group A
Streptococcus
(GAS), a human pathogen, is the causative agent of a wide range of
diseases, from mild skin infection to life-threatening diseases such as necrotizing fasciitis69.
GAS encodes a P1B4-type ATPase under regulation of PerR, and was therefore named a
PerR-regulated metal transporter (PmtA). In a
perR
null mutant, high level expression of
pmtA
is associated with derepression of genes normally responsive to cellular zinc status
due to repression by AdcR70, a Zn2+-dependent repressor. This simplest interpretation of
this result is that PmtA may function as a Zn2+ efflux transporter. Consistent with this
notion, a
perR
null mutant has an increased resistance to Zn2+, and this depends on PmtA70.
However, it is unclear why cells would efflux Zn2+ in response to H2O2 stress, nor is there
any evidence that PmtA is important for Zn2+ resistance in wild-type cells, which
presumably relies on the Zn2+-inducible CzcD efflux pump to ameliorate Zn2+-toxicity.
By analogy with PfeT and its orthologs, an alternative interpretation is that the primary role
of PmtA is as a H2O2-inducible Fe2+-efflux pump and that activity with Zn2+ may only be
revealed when it is constitutively overexpressed in a
perR
null mutant. Two recent studies
have confirmed the primary role of PmtA as an Fe2+-efflux pump38, 39. PmtA is important
for resistance to iron intoxication, and a
pmtA
null mutant accumulates elevated levels of
intracellular iron. As expected, expression of
pmtA
is strongly induced by Fe2+. Although a
pmtA
null mutant shows similar sensitivity to peroxide stress as a wild type strain in the
Pi and Helmann Page 6
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
absence of excess Fe2+, it exhibits significantly increased susceptibility to peroxide stress
when treated with Fe2+. Since GAS is catalase negative, PmtA might be a frontline defense
against peroxide stress. PmtA is also a critical virulence factor and is required for survival
during infection in both intramuscular and subcutaneous mouse models38, which again links
iron efflux and peroxide resistance to pathogen virulence.
Nia in Sinorhizobium meliloti
In addition to the P1B4-ATPases featured above, it is possible that P1B-ATPases of other
groups may also have physiologically relevant activity with iron. One example is Nia, a
P1B5-ATPase with a C-terminal hemerythrin domain. Since hemerythrin domains bind O2
via a diiron active site, this suggests a possible role in O2-sensing71, 72. Nia is encoded by
the symbiotic plasmid A of
Sinorhizobium meliloti
, a nitrogen fixing microbe in the
Rhizobiales
lineage that has a symbiotic relationship with legumes in which it establishes
nodules associated with roots.
Consistent with a possible role in Fe2+ efflux, a
nia
null mutant accumulates Fe2+ under
excess metal conditions73. However, Nia also functions with Ni2+ and a
nia
null mutant
accumulates Ni2+ when in excess. The precise physiological role of Nia is not yet resolved.
Biochemical assays suggest that Nia interacts with both Fe2+ and Ni2+ (but not Co2+).
However, a
nia
null mutant showed moderate sensitivity to Ni2+, but not to Fe2+, under the
conditions tested73. Expression of
nia
was moderately induced by Fe2+ (3-fold), Ni2+ (3-
fold), and Co2+ (2-fold), but not by other metals. Interestingly,
nia
was most strongly
induced (20-fold) in root nodules, thought to be a microaerobic, iron-rich environment74.
These results lead to a model in which Nia is expressed in nitrogen-fixing root nodules, in
response to either iron excess or microaerobic conditions. The C-terminal hemerythrin
domain may also participate in or regulate transport activity, perhaps in response to O273.
More work needs to be done to characterize the details of
nia
gene regulation and to more
clearly define the physiological role of Nia during the
S. meliloti
-plant symbiosis.
Cation diffusion facilitator (CDF) proteins
Cation diffusion facilitators (CDFs) are a family of membrane-bound proteins that export
and thereby confer tolerance to heavy metal ions75, 76. CDF proteins are ubiquitous in
bacteria, archaea, and eukaryotes77. Collectively, bacterial CDF proteins have been
implicated in transport of a wide range of metal ions (Zn2+, Cd2+, Co2+, Ni2+, Fe2+ and
Mn2+) with some transporters able to transport multiple metals78–82. Phylogenetic analysis
of the CDF transporters defines three major groups corresponding to substrate specificity: 1)
manganese efflux, 2) iron/zinc efflux, 3) zinc and other metals (but not manganese or iron)
efflux83.
A typical bacterial CDF contains an N-terminal domain (NTD), 6 transmembrane helices
(TM), a histidine-rich interconnecting loop (IL) between TM4 and TM5, and a C-terminal
cytoplasmic domain (CTD)75 (Fig 2). However, the detailed mechanisms of metal selectivity
are unknown. Some studies suggest the cytoplasmic domain or the IL loop is important for
metal specificity84–86, but other studies highlight the role of residues in the TM3 helix on
metal selectivity87. For the
E. coli
FieF transporter, evidence supports a role for a tetrahedral
Pi and Helmann Page 7
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
metal-binding site formed between TM2 and TM5 in metal selectivity88. So far, there is no
unifying model that can account for metal selectivity of CDF proteins.
FieF in E. coli: Zn2+ vs. Fe2+ efflux
There are two CDF transporters in
E. coli
: ZitB and FieF (also named as YiiP). ZitB is the
secondary zinc efflux system that is critical for maintaining zinc homeostasis only when the
zinc efflux ATPase ZntA is absent89. FieF has been studied for more than a decade, but its
physiological function has been controversial. In 2004, the first two reports of its structural
analysis were built on the assumption that FieF acts as a zinc efflux protein90, 91. In fact,
prior studies had demonstrated that
fieF
is induced by either zinc or iron89. However, ectopic
expression of FieF does not restore zinc tolerance in a zinc-sensitive strain, suggesting it
might not play a role in zinc homeostasis89.
Physiological studies suggest that the major physiological role of FieF may be in iron
tolerance. Indeed, FieF is important for full resistance to iron intoxication in a
fur
null
mutant, where iron homeostasis is disrupted and iron uptake systems are constitutively
expressed92. Ectopic expression of FieF leads to reduced accumulation of iron in a
fieF
null
mutant. Moreover, reconstitution of FieF in proteoliposomes showed that it mediates iron
transport in vitro92. These results all support the assignment of FieF (ferrous iron efflux) as
an iron efflux transporter. However, this notion has been challenged by others. For example,
FieF was shown to selectively bind zinc and cadmium with high affinity, but not iron or
other metals tested93. Based on the site-directed fluorescence resonance energy transfer
(FRET) measurements, Lu
et al
. proposed an autoregulation model of transport activity in
response to intracellular zinc levels94. Currently, FieF (YiiP) is referred to as a Zn2+
transporter in most published papers.
Ever since its structure was solved in 200779, FieF has been considered as a prototype for
bacterial CDF proteins, which makes it more frustrating that its physiological role has
remained controversial. The regulation of
fieF
expression has not been well defined, but it
does not appear to be regulated by Fur92. The physiological studies of FieF are certainly
supportive of a role in Fe(II) efflux. This inference is further supported by the observation
that the FieF homologs MamM and MamB form a heterodimeric CDF protein required for
Fe(II) import into vesicles in support of magnetosome formation in the magnetotatic
bacterium
Magnetospirillum gryphiswaldense
95, 96.
AitP in Pseudomonas aeruginosa
Pseudomonas aeruginosa
is Gram-negative, opportunistic pathogen that is highly antibiotic
resistant.
P. aeruginosa
encodes three paralogous CDF efflux systems: CzcD (PA0397), AitP
(PA1297), and YiiP (PA3963). Of these, the alternative iron transport protein (AitP) most
likely functions physiologically in Fe2+ efflux. Deletion of
aitP
leads to an increased
sensitivity to both Fe2+ and Co2+, increased intracellular accumulation of both ions, and
decreased survival in presence of H2O297. The observed sensitivity to H2O2 is most
consistent with a role in Fe2+ efflux, as noted above for P-type ATPases. In contrast with
AitP, the CzcD and YiiP proteins were inferred to function physiologically in Zn2+
resistance, although this role is largely masked in wild-type cells by the activity of the Zn2+
Pi and Helmann Page 8
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
efflux P-type ATPase, ZntA98. All the three transporters are critical for virulence in a plant
infection model97. However, it remains unclear why this organism requires multiple classes
of Zn2+ efflux proteins or under what conditions the three proteins are physiologically
important during the infection process.
FeoE in Shewanella oneidensis MR-1
Shewanella oneidensis
MR-1 is a facultative anaerobe in the γ-proteobacterium family that
is capable of respiration using metals (e.g. manganese, lead, uranium and ferric iron) as
electron acceptors99.
S. oneidensis
cells are usually pink or red, reflective of a high iron
content in hemoproteins and cytochromes100. When Fe3+ is used as a terminal electron
acceptor, cells generate a large amount of soluble Fe2+ which could potentially lead to iron
intoxication. FeoE, a CDF protein, is required for cell growth during anaerobic iron
respiration, and deletion of
feoE
increased susceptibility to Fe2+ intoxication, consistent
with a physiological role in Fe2+ efflux101. Further work is required to understand how
feoE
expression is regulated. It is unclear, for example, whether
feoE
is induced in response to
excess iron. Fur is the primary regulator that modulates iron acquisition in
S. oneidenis
102,
and is a candidate for an iron-responsive transcription factor that could be involved.
Major facilitator superfamily (MFS)
The major facilitator superfamily (MFS) of membrane transporters function with a wide
scope of small molecules such as ions, nucleosides, amino acids, small peptides, and
lipids103. They can be categorized into three groups: uniporters that transport a single
substrate, symporters that transport a substrate coupled with another ion (generally a
proton), and antiporters that transport two substrates in opposite directions104, 105. All the
MFS transporters share a canonical structural fold composed of two distinct domains [Fig.
2], each consisting of six transmembrane helices. The substrate binding site is located at the
interface between these two domains103.
The mechanism of transport by MFS proteins is not clear, but several related models have
been proposed. The first, an alternate-access model, was proposed more than five decades
ago106. This model speculates that the transporters undergo a conformational change that
alternates between a form where substrate can bind from one side of the membrane to one
where it can only bind from the other side. This has been validated by many structural
studies such as the xylose/H+ symporter XylE and for LacY107–109. The second, a rocker-
switch model, postulates that conformational changes are accomplished through rocker-
switch-type rotation between the N and C domain. This model is supported by some open-
conformation structures110 but not by the structures in occluded states111–114. A third,
clamp-and-switch model, provides a two-step transport mechanism: a clamping step that
mediates occlusion of the binding site and a switching step that mediates the exposure of the
binding site. This model postulates four conformational states: inward open, outward open,
inward-facing occlusion, and outward-facing occlusion105. This model is in a good
agreement with studies of some MFS transporters115, 116, but more structural analyses
combined with biochemical and computational analyses are needed to further understand the
transport mechanism of MFS transporters.
Pi and Helmann Page 9
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
IceT (iron and citrate efflux transporter) in Salmonella Typhimurium
Salmonella
Typhimurium is a Gram-negative pathogen commonly found in the
gastrointestinal tract. IceT (MdtD) is a member of the MFS superfamily in
S.
Typhimurium.
The mdtABCD baeSR operon encodes IceT and two other systems: a RND (resistance-
nodulation-division) drug efflux system MdtABC and a two-component regulatory system
BaeSR that regulates antibiotic resistance and efflux117–119. IceT is proposed to be an iron-
citrate efflux transporter and it can export either iron citrate or citrate alone120. The
iceT
null
mutant shows increased susceptibility to the antibiotic streptonigrin (SN), the activity of
which is modulated by the level of intracellular free iron121. This result suggests that the
mutation of
iceT
leads to an increase in intracellular labile iron pools. Consistent with this
result, induction of IceT expression leads to reduced levels of intracellular iron120.
Although the mdtABCD baeSR operon is not induced directly by high Fe2+ 122, it is induced
by disruption of iron homeostasis in a
fur
null mutant where iron uptake systems are
constitutively expressed, supportive of a physiological role for IceT in iron efflux. Although
IceT confers resistance to peroxide stress in a
fur
null mutant, the mdtABCD baeSR operon
is not induced by H2O2 or superoxide-generating reagents such as paraquat120. However, it
is induced by nitric oxide, which is also known to interact with the labile iron pool120. The
significance of the regulation of IceT, together with its co-transcribed ABC transporter, by
the BaeSR two-component system is not understood, nor is it yet clear whether or not IceT
is important for pathogenesis.
Membrane bound ferritin A (MbfA) in Agrobacterium tumefaciens and
Bradyrhizobium japonicum
Agrobacterium tumefaciens
belongs to the
Rhizobiales
lineage and is the causative agent of
the economically important plant disease, crown gall. MbfA was originally described as
membrane-bound ferritin A, and is a member of the erythrin-vacuolar iron transport (Er-
VIT1) ferritin-like superfamily. MbfA has two major domains: an N-terminal ferritin-like or
Er domain (Er) and a C-terminal membrane-embedded vacuolar iron transporter domain
(VIT1) (Fig. 2). The Er domain has a di-iron binding site and the VIT1 domain shows
sequence homology to
Arabidopsis
VIT1, which is responsible for transferring iron into
vacuoles123. Ferritin is a cytosolic iron storage protein ubiquitous in prokaryotes and
eukaryotes124, however, MbfA is not a
bona fide
ferritin and its physiological function was
not immediately apparent.
Plant hosts often produce reactive oxygen species as a defense mechanism in response to
microbial infection. Initial studies revealed that MbfA confers resistance to H2O2 stress,
suggesting that it may play an important role in plant-pathogen interaction125. Moreover,
mbfA
expression was induced in response to high iron conditions as sensed by the iron
response regulator protein, Irr125. However, these results could not distinguish between a
role for MbfA in sequestration of iron (through its ferritin domain) or iron efflux. A follow
up study revealed that MbfA is important for resistance to iron intoxication under acidic
conditions (pH 5.5), which enhances iron solubility thereby promoting toxicity126.
Compared to wild-type, an
mbfA
null mutant had a modest increase in intracellular total iron
Pi and Helmann Page 10
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
as well as labile iron125. Since its expression is induced by high iron under acidic
conditions125, and leads to reduced intracellular iron levels, MbfA was postulated to
function as an iron efflux transporter125.
Bradyrhizobium japonicum
also encodes an MbfA protein implicated in iron efflux127.
B.
japonicum
is a nitrogen-fixing endosymbiotic microbe that, like
A. tumefaciens
, belongs to
the
Rhizobiales
lineage. As in
A. tumefaciens
, iron homeostasis in
B. japonicum
is also
under control of Irr128, which regulates iron uptake, storage, and utilization129. MbfA in
B.
japonicum
is specifically induced by high iron and confers resistance to iron intoxication
and H2O2 stress. Moreover, an
mbfA
null mutant accumulates significantly high levels of
iron.
Collectively, these data support the idea that MbfA functions physiologically as an iron
efflux transporter127. Interestingly, the N-terminal ferritin-like domain located on the
cytoplasmic side of inner membrane is required for iron transport activity and stress
resistance. The purified ferritin domain forms a dimer in solution, which suggests that MbfA
may dimerize to form a functional channel127. By mediating the efflux of Fe2+, MbfA
functions cooperatively with bacterioferritin (Bfr), which functions in iron sequestration, to
prevent iron intoxication130. Mutation of either
mbfA
or
bfr
increases Fe2+ sensitivity, but a
double
mbfA bfr
mutant is extremely sensitive to iron130.
Conclusions
Efflux systems play a central role in the resistance of bacteria to heavy metals, but their role
in iron homeostasis has been relatively slow to emerge. This is perhaps a reflection of the
fact that iron limitation is a far more prevalent challenge for bacteria than iron
intoxication131, due in part to the very low solubility of iron under aerobic conditions of near
neutral pH. Recent results, however, have greatly expanded our appreciation of the central
importance of iron efflux systems and their contribution to virulence in human
pathogens27, 37, 38. This implies that iron intoxication imposes a selective pressure during
infection, although how this arises is not yet clear. For example, iron intoxication may arise
from an uncontrolled influx of iron into the cell from the outside. Indeed, it is thought that
macrophages impose Zn2+ and Cu+ toxicity on engulfed bacteria by import of metals into
the phagolysosome132. However, iron is not known to be imported into the phagocytic
vacuole. Iron overload may also result when bacteria exposed to an iron limited
environment, and therefore expressing high affinity uptake systems, transition to an iron-rich
environment. The sudden influx of iron may then be best accommodated by storage or
efflux. Alternatively, or in addition, iron intoxication may arise from within the cell. For
example, oxidative stress may lead to the release of iron from abundant iron-sulfur and
mononuclear iron enzymes, thereby leading to an increase in cytosolic iron levels.
Iron intoxication may also be present in specific environments. For example, acidophilic
bacteria grow in low pH environments where iron concentrations may be 1018 times higher
than that found in pH neutral environments133. In the case of iron-respiring bacteria, high
local concentrations of Fe2+ may be produced by reduction of Fe3+-containing minerals101.
Pi and Helmann Page 11
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Further work is needed to better define the prevalence of iron intoxication in natural
environment settings and the role of iron efflux in these environments.
With the identification of the several families of iron efflux systems noted here, the stage is
now set for further structural, biochemical and genetic studies to address their mechanisms
of metal selectivity. It is presently unclear how these efflux transporters discriminate Fe2+
from competing substrates and how, at a structural level, efflux is coupled to substrate
binding and energy consumption. It is also unclear why some cells rely on ATP-dependent
P-type transporters and others utilize CDF proteins, which are coupled to the proton motive
force. It is notable that in several cases efflux pumps were initially assigned a role for
substrates others than Fe2+ (PfeT, FrvA, CtpD), and in other cases (FieF, Nia) the most
relevant physiological substrate is still unclear. This highlights the fact that metal selectivity
cannot be easily predicted from protein sequence alone, and biochemical assays need to be
interpreted in context of the physiology of the organisms. In several of the cases described,
the most compelling evidence to assign function has emerged from a careful analysis of
mutant phenotypes combined with detailed analysis of regulation to infer those conditions
that specifically induce expression.
Supplementary Material
Refer to Web version on PubMed Central for supplementary material.
Acknowledgments
We thank Pete Chandrangsu for helpful comments. This work was supported by a grant from the NIH (GM059323)
to JDH.
References
1. Imlay JA. Pathways of oxidative damage. Annual review of Microbiology. 2003; 57:395–418.
2. Park S, You X, Imlay JA. Substantial DNA damage from submicromolar intracellular hydrogen
peroxide detected in Hpx-mutants of
Escherichia coli
. Proceedings of the National Academy of
Sciences of the United States of America. 2005; 102:9317–9322. [PubMed: 15967999]
3. Imlay JA. The mismetallation of enzymes during oxidative stress. The Journal of biological
chemistry. 2014; 289:28121–28128. [PubMed: 25160623]
4. Barwinska-Sendra, A., Waldron, KJ. Advances in Microbial Physiology. Academic Press; DOI:
http://doi.org/10.1016/bs.ampbs.2017.01.003
5. Anjem A, Imlay JA. Mononuclear iron enzymes are primary targets of hydrogen peroxide stress.
The Journal of biological chemistry. 2012; 287:15544–15556. [PubMed: 22411989]
6. Jang S, Imlay JA. Micromolar intracellular hydrogen peroxide disrupts metabolism by damaging
iron-sulfur enzymes. The Journal of biological chemistry. 2007; 282:929–937. [PubMed: 17102132]
7. Waldron KJ, Rutherford JC, Ford D, Robinson NJ. Metalloproteins and metal sensing. Nature. 2009;
460:823–830. [PubMed: 19675642]
8. Chandrangsu P, Rensing C, Helmann JD. Metal homeostasis and resistance in bacteria,
Nature
reviews
. Microbiology. 2017; doi: 10.1038/nrmicro.2017.15
9. Fleischhacker AS, Kiley PJ. Iron-containing transcription factors and their roles as sensors. Current
opinion in chemical biology. 2011; 15:335–341. [PubMed: 21292540]
10. Rodriguez GM, Voskuil MI, Gold B, Schoolnik GK, Smith I. ideR, An essential gene in
Mycobacterium tuberculosis
: role of IdeR in iron-dependent gene expression, iron metabolism,
and oxidative stress response. Infection and immunity. 2002; 70:3371–3381. [PubMed: 12065475]
Pi and Helmann Page 12
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
11. Sritharan M. Iron Homeostasis in
Mycobacterium tuberculosis
: Mechanistic Insights into
Siderophore-Mediated Iron Uptake. Journal of bacteriology. 2016; 198:2399–2409. [PubMed:
27402628]
12. Rodionov DA, Gelfand MS, Todd JD, Curson AR, Johnston AW. Computational reconstruction of
iron- and manganese-responsive transcriptional networks in alpha-proteobacteria. PLoS
computational biology. 2006; 2:e163. [PubMed: 17173478]
13. Todd JD, Sawers G, Rodionov DA, Johnston AW. The
Rhizobium leguminosarum
regulator IrrA
affects the transcription of a wide range of genes in response to Fe availability. Molecular genetics
and genomics: MGG. 2006; 275:564–577. [PubMed: 16625355]
14. Yang J, Sangwan I, Lindemann A, Hauser F, Hennecke H, Fischer HM, O'Brian MR.
Bradyrhizobium japonicum
senses iron through the status of haem to regulate iron homeostasis
and metabolism. Molecular Microbiology. 2006; 60:427–437. [PubMed: 16573691]
15. Helmann JD. Specificity of metal sensing: iron and manganese homeostasis in
Bacillus subtilis
.
The Journal of biological chemistry. 2014; 289:28112–28120. [PubMed: 25160631]
16. Seo SW, Kim D, Latif H, O'Brien EJ, Szubin R, Palsson BO. Deciphering Fur transcriptional
regulatory network highlights its complex role beyond iron metabolism in
Escherichia coli
. Nature
communications. 2014; 5:4910.
17. Delany I, Rappuoli R, Scarlato V. Fur functions as an activator and as a repressor of putative
virulence genes in
Neisseria meningitidis
. Molecular Microbiology. 2004; 52:1081–1090.
[PubMed: 15130126]
18. Yu C, Genco CA. Fur-mediated activation of gene transcription in the human pathogen
Neisseria
gonorrhoeae
. Journal of bacteriology. 2012; 194:1730–1742. [PubMed: 22287521]
19. Andrews SC, Robinson AK, Rodriguez-Quinones F. Bacterial iron homeostasis. FEMS
Microbiology reviews. 2003; 27:215–237. [PubMed: 12829269]
20. McHugh JP, Rodriguez-Quinones F, Abdul-Tehrani H, Svistunenko DA, Poole RK, Cooper CE,
Andrews SC. Global iron-dependent gene regulation in
Escherichia coli
. A new mechanism for
iron homeostasis. The Journal of biological chemistry. 2003; 278:29478–29486. [PubMed:
12746439]
21. Hunt MD, Pettis GS, McIntosh MA. Promoter and operator determinants for fur-mediated iron
regulation in the bidirectional fepA-fes control region of the
Escherichia coli
enterobactin gene
system. Journal of bacteriology. 1994; 176:3944–3955. [PubMed: 8021177]
22. Masse E, Gottesman S. A small RNA regulates the expression of genes involved in iron
metabolism in
Escherichia coli
. Proceedings of the National Academy of Sciences of the United
States of America. 2002; 99:4620–4625. [PubMed: 11917098]
23. Masse E, Salvail H, Desnoyers G, Arguin M. Small RNAs controlling iron metabolism. Current
opinion in Microbiology. 2007; 10:140–145. [PubMed: 17383226]
24. Masse E, Vanderpool CK, Gottesman S. Effect of RyhB small RNA on global iron use in
Escherichia coli
. Journal of bacteriology. 2005; 187:6962–6971. [PubMed: 16199566]
25. Nandal A, Huggins CC, Woodhall MR, McHugh J, Rodriguez-Quinones F, Quail MA, Guest JR,
Andrews SC. Induction of the ferritin gene (
ftnA
) of
Escherichia coli
by Fe2+-Fur is mediated by
reversal of H-NS silencing and is RyhB independent. Molecular Microbiology. 2010; 75:637–657.
[PubMed: 20015147]
26. Faulkner MJ, Ma Z, Fuangthong M, Helmann JD. Derepression of the
Bacillus subtilis
PerR
peroxide stress response leads to iron deficiency. Journal of bacteriology. 2012; 194:1226–1235.
[PubMed: 22194458]
27. Pi H, Patel SJ, Arguello JM, Helmann JD. The
Listeria monocytogenes
Fur-regulated virulence
protein FrvA is an Fe(II) efflux P1B4 -type ATPase. Molecular Microbiology. 2016; 100:1066–
1079. [PubMed: 26946370]
28. Storz G, Altuvia S. OxyR regulon. Methods in enzymology. 1994; 234:217–223. [PubMed:
7528872]
29. Herbig AF, Helmann JD. Roles of metal ions and hydrogen peroxide in modulating the interaction
of the
Bacillus subtilis
PerR peroxide regulon repressor with operator DNA. Molecular
Microbiology. 2001; 41:849–859. [PubMed: 11532148]
Pi and Helmann Page 13
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
30. Calhoun LN, Kwon YM. Structure, function and regulation of the DNA-binding protein Dps and
its role in acid and oxidative stress resistance in
Escherichia coli
: a review. Journal of applied
Microbiology. 2011; 110:375–386. [PubMed: 21143355]
31. Chen L, Helmann JD.
Bacillus subtilis
MrgA is a Dps(PexB) homologue: evidence for
metalloregulation of an oxidative-stress gene. Molecular Microbiology. 1995; 18:295–300.
[PubMed: 8709848]
32. Faulkner MJ, Helmann JD. Peroxide stress elicits adaptive changes in bacterial metal ion
homeostasis. Antioxidants & redox signaling. 2011; 15:175–189. [PubMed: 20977351]
33. Anjem A, Varghese S, Imlay JA. Manganese import is a key element of the OxyR response to
hydrogen peroxide in
Escherichia coli
. Molecular Microbiology. 2009; 72:844–858. [PubMed:
19400769]
34. Kehres DG, Zaharik ML, Finlay BB, Maguire ME. The NRAMP proteins of
Salmonella
typhimurium and
Escherichia coli
are selective manganese transporters involved in the response to
reactive oxygen. Molecular Microbiology. 2000; 36:1085–1100. [PubMed: 10844693]
35. Gaballa A, Helmann JD. A peroxide-induced zinc uptake system plays an important role in
protection against oxidative stress in
Bacillus subtilis
. Molecular Microbiology. 2002; 45:997–
1005. [PubMed: 12180919]
36. Guan G, Pinochet-Barros A, Gaballa A, Patel SJ, Arguello JM, Helmann JD. PfeT, a P1B4 -type
ATPase, effluxes ferrous iron and protects
Bacillus subtilis
against iron intoxication. Molecular
Microbiology. 2015; 98:787–803. [PubMed: 26261021]
37. Patel SJ, Lewis BE, Long JE, Nambi S, Sassetti CM, Stemmler TL, Arguello JM. Fine-tuning of
Substrate Affinity Leads to Alternative Roles of
Mycobacterium tuberculosis
Fe2+-ATPases. The
Journal of biological chemistry. 2016; 291:11529–11539. [PubMed: 27022029]
38. VanderWal AR, Makthal N, Pinochet-Barros A, Helmann JD, Olsen RJ, Kumaraswami M. Iron
Efflux by PmtA Is Critical for Oxidative Stress Resistance and Contributes Significantly to Group
A
Streptococcus
Virulence. Infection and immunity. 2017; 85
39. Turner AG, Ong CY, Djoko KY, West NP, Davies MR, McEwan AG, Walker MJ. The PerR-
Regulated P1B-4-Type ATPase (PmtA) Acts as a Ferrous Iron Efflux Pump in
Streptococcus
pyogenes
. Infection and immunity. 2017; 85
40. Kuhlbrandt W. Biology, structure and mechanism of P-type ATPases,
Nature reviews
. Molecular
cell biology. 2004; 5:282–295. [PubMed: 15071553]
41. Chan H, Babayan V, Blyumin E, Gandhi C, Hak K, Harake D, Kumar K, Lee P, Li TT, Liu HY, Lo
TC, Meyer CJ, Stanford S, Zamora KS, Saier MH Jr. The P-type ATPase superfamily. J Mol
Microbiol Biotechnol. 2010; 19:5–104. [PubMed: 20962537]
42. Lenoir G, Williamson P, Holthuis JC. On the origin of lipid asymmetry: the flip side of ion
transport. Current opinion in chemical biology. 2007; 11:654–661. [PubMed: 17981493]
43. Lopez-Marques RL, Poulsen LR, Hanisch S, Meffert K, Buch-Pedersen MJ, Jakobsen MK,
Pomorski TG, Palmgren MG. Intracellular targeting signals and lipid specificity determinants of
the ALA/ALIS P4-ATPase complex reside in the catalytic ALA alpha-subunit. Molecular biology
of the cell. 2010; 21:791–801. [PubMed: 20053675]
44. Thever MD, Saier MH Jr. Bioinformatic characterization of P-type ATPases encoded within the
fully sequenced genomes of 26 eukaryotes. The Journal of membrane biology. 2009; 229:115–130.
[PubMed: 19548020]
45. Gonzalez-Guerrero M, Eren E, Rawat S, Stemmler TL, Arguello JM. Structure of the two
transmembrane Cu+ transport sites of the Cu+-ATPases. The Journal of biological chemistry.
2008; 283:29753–29759. [PubMed: 18772137]
46. Fan B, Rosen BP. Biochemical characterization of CopA, the
Escherichia coli
Cu(I)-translocating
P-type ATPase. The Journal of biological chemistry. 2002; 277:46987–46992. [PubMed:
12351646]
47. Mandal AK, Cheung WD, Arguello JM. Characterization of a thermophilic P-type Ag+/Cu+-
ATPase from the extremophile
Archaeoglobus fulgidus
. The Journal of biological chemistry. 2002;
277:7201–7208. [PubMed: 11756450]
48. Liu J, Dutta SJ, Stemmler AJ, Mitra B. Metal-binding affinity of the transmembrane site in ZntA:
implications for metal selectivity. Biochemistry. 2006; 45:763–772. [PubMed: 16411752]
Pi and Helmann Page 14
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
49. Nucifora G, Chu L, Misra TK, Silver S. Cadmium resistance from
Staphylococcus aureus
plasmid
pI258 cadA gene results from a cadmium-efflux ATPase. Proceedings of the National Academy of
Sciences of the United States of America. 1989; 86:3544–3548. [PubMed: 2524829]
50. Mana-Capelli S, Mandal AK, Arguello JM.
Archaeoglobus fulgidus
CopB is a thermophilic Cu2+-
ATPase: functional role of its histidine-rich-N-terminal metal binding domain. The Journal of
biological chemistry. 2003; 278:40534–40541. [PubMed: 12876283]
51. Zielazinski EL, Cutsail GE 3rd, Hoffman BM, Stemmler TL, Rosenzweig AC. Characterization of
a cobalt-specific P(1B)-ATPase. Biochemistry. 2012; 51:7891–7900. [PubMed: 22971227]
52. Smith AT, Smith KP, Rosenzweig AC. Diversity of the metal-transporting P1B-type ATPases.
Journal of biological inorganic chemistry: JBIC. 2014; 19:947–960. a publication of the Society of
Biological Inorganic Chemistry. [PubMed: 24729073]
53. Arguello JM. Identification of ion-selectivity determinants in heavy-metal transport P1B-type
ATPases. The Journal of membrane biology. 2003; 195:93–108. [PubMed: 14692449]
54. Ollinger J, Song KB, Antelmann H, Hecker M, Helmann JD. Role of the Fur regulon in iron
transport in
Bacillus subtilis
. Journal of bacteriology. 2006; 188:3664–3673. [PubMed: 16672620]
55. Baichoo N, Wang T, Ye R, Helmann JD. Global analysis of the
Bacillus subtilis
Fur regulon and
the iron starvation stimulon. Molecular Microbiology. 2002; 45:1613–1629. [PubMed: 12354229]
56. Fuangthong M, Herbig AF, Bsat N, Helmann JD. Regulation of the
Bacillus subtilis fur
and
perR
genes by PerR: not all members of the PerR regulon are peroxide inducible. Journal of
bacteriology. 2002; 184:3276–3286. [PubMed: 12029044]
57. Lee JW, Helmann JD. Functional specialization within the Fur family of metalloregulators,
Biometals
: an international journal on the role of metal ions in biology. Biochemistry, and
medicine. 2007; 20:485–499.
58. Gaballa A, Antelmann H, Aguilar C, Khakh SK, Song KB, Smaldone GT, Helmann JD. The
Bacillus subtilis
iron-sparing response is mediated by a Fur-regulated small RNA and three small,
basic proteins. Proceedings of the National Academy of Sciences of the United States of America.
2008; 105:11927–11932. [PubMed: 18697947]
59. Smaldone GT, Antelmann H, Gaballa A, Helmann JD. The FsrA sRNA and FbpB protein mediate
the iron-dependent induction of the
Bacillus subtilis
lutABC iron-sulfur-containing oxidases.
Journal of bacteriology. 2012; 194:2586–2593. [PubMed: 22427629]
60. Smaldone GT, Revelles O, Gaballa A, Sauer U, Antelmann H, Helmann JD. A global investigation
of the
Bacillus subtilis
iron-sparing response identifies major changes in metabolism. Journal of
bacteriology. 2012; 194:2594–2605. [PubMed: 22389480]
61. Smaldone GT, Helmann JD. CsoR regulates the copper efflux operon
copZA
in
Bacillus subtilis
.
Microbiology (Reading, England). 2007; 153:4123–4128.
62. Moore CM, Gaballa A, Hui M, Ye RW, Helmann JD. Genetic and physiological responses of
Bacillus subtilis
to metal ion stress. Molecular Microbiology. 2005; 57:27–40. [PubMed:
15948947]
63. McLaughlin HP, Xiao Q, Rea RB, Pi H, Casey PG, Darby T, Charbit A, Sleator RD, Joyce SA,
Cowart RE, Hill C, Klebba PE, Gahan CG. A putative P-type ATPase required for virulence and
resistance to haem toxicity in
Listeria monocytogenes
. PloS one. 2012; 7:e30928. [PubMed:
22363518]
64. Rea R, Hill C, Gahan CG.
Listeria monocytogenes
PerR mutants display a small-colony
phenotype, increased sensitivity to hydrogen peroxide, and significantly reduced murine virulence.
Applied and environmental Microbiology. 2005; 71:8314–8322. [PubMed: 16332818]
65. Ledala N, Sengupta M, Muthaiyan A, Wilkinson BJ, Jayaswal RK. Transcriptomic response of
Listeria monocytogenes
to iron limitation and Fur mutation. Applied and environmental
Microbiology. 2010; 76:406–416. [PubMed: 19933349]
66. McLaughlin HP, Bahey-El-Din M, Casey PG, Hill C, Gahan CG. A mutant in the
Listeria
monocytogenes
Fur-regulated virulence locus (
frvA
) induces cellular immunity and confers
protection against listeriosis in mice. Journal of medical Microbiology. 2013; 62:185–190.
[PubMed: 23105022]
Pi and Helmann Page 15
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
67. Novoa-Aponte L, Soto Ospina CY.
Mycobacterium tuberculosis
P-type ATPases: possible targets
for drug or vaccine development. BioMed research international. 2014:296986. [PubMed:
25110669]
68. Raimunda D, Long JE, Padilla-Benavides T, Sassetti CM, Arguello JM. Differential roles for the
Co2+ /Ni2+ transporting ATPases, CtpD and CtpJ, in
Mycobacterium tuberculosis
virulence.
Molecular Microbiology. 2014; 91:185–197. [PubMed: 24255990]
69. Olsen RJ, Shelburne SA, Musser JM. Molecular mechanisms underlying group A
streptococcal
pathogenesis. Cellular Microbiology. 2009; 11:1–12. [PubMed: 18710460]
70. Brenot A, Weston BF, Caparon MG. A PerR-regulated metal transporter (PmtA) is an interface
between oxidative stress and metal homeostasis in
Streptococcus pyogenes
. Molecular
Microbiology. 2007; 63:1185–1196. [PubMed: 17238923]
71. Xiong J, Kurtz DM Jr, Ai J, Sanders-Loehr J. A hemerythrin-like domain in a bacterial chemotaxis
protein. Biochemistry. 2000; 39:5117–5125. [PubMed: 10819979]
72. Karlsen OA, Ramsevik L, Bruseth LJ, Larsen O, Brenner A, Berven FS, Jensen HB, Lillehaug JR.
Characterization of a prokaryotic haemerythrin from the methanotrophic bacterium
Methylococcus
capsulatus
(Bath). The FEBS journal. 2005; 272:2428–2440. [PubMed: 15885093]
73. Zielazinski EL, Gonzalez-Guerrero M, Subramanian P, Stemmler TL, Arguello JM, Rosenzweig
AC.
Sinorhizobium meliloti
Nia is a P(1B-5)-ATPase expressed in the nodule during plant
symbiosis and is involved in Ni and Fe transport. Metallomics: integrated biometal science. 2013;
5:1614–1623. [PubMed: 24056637]
74. Becker A, Berges H, Krol E, Bruand C, Ruberg S, Capela D, Lauber E, Meilhoc E, Ampe F, de
Bruijn FJ, Fourment J, Francez-Charlot A, Kahn D, Kuster H, Liebe C, Puhler A, Weidner S, Batut
J. Global changes in gene expression in
Sinorhizobium meliloti
1021 under microoxic and
symbiotic conditions. Molecular plant-microbe interactions: MPMI. 2004; 17:292–303. [PubMed:
15000396]
75. Haney CJ, Grass G, Franke S, Rensing C. New developments in the understanding of the cation
diffusion facilitator family. Journal of industrial Microbiology & biotechnology. 2005; 32:215–
226. [PubMed: 15889311]
76. Kolaj-Robin O, Russell D, Hayes KA, Pembroke JT, Soulimane T. Cation Diffusion Facilitator
family: Structure and function. FEBS letters. 2015; 589:1283–1295. [PubMed: 25896018]
77. Nies DH. Efflux-mediated heavy metal resistance in prokaryotes. FEMS Microbiology reviews.
2003; 27:313–339. [PubMed: 12829273]
78. Munkelt D, Grass G, Nies DH. The chromosomally encoded cation diffusion facilitator proteins
DmeF and FieF from
Wautersia metallidurans
CH34 are transporters of broad metal specificity.
Journal of bacteriology. 2004; 186:8036–8043. [PubMed: 15547276]
79. Lu M, Fu D. Structure of the zinc transporter YiiP. Science (New York, N.Y.). 2007; 317:1746–
1748.
80. Cubillas C, Vinuesa P, Tabche ML, Davalos A, Vazquez A, Hernandez-Lucas I, Romero D, Garcia-
de los Santos A. The cation diffusion facilitator protein EmfA of
Rhizobium etli
belongs to a novel
subfamily of Mn(2+)/Fe(2+) transporters conserved in alpha-proteobacteria. Metallomics :
integrated biometal science. 2014; 6:1808–1815. [PubMed: 25054342]
81. Raimunda D, Elso-Berberian G. Functional characterization of the CDF transporter SMc02724
(SmYiiP) in
Sinorhizobium meliloti
: Roles in manganese homeostasis and nodulation. Biochimica
et biophysica acta. 2014; 1838:3203–3211. [PubMed: 25242380]
82. Rosch JW, Gao G, Ridout G, Wang YD, Tuomanen EI. Role of the manganese efflux system
mntE
for signalling and pathogenesis in
Streptococcus pneumoniae
. Molecular Microbiology. 2009;
72:12–25. [PubMed: 19226324]
83. Montanini B, Blaudez D, Jeandroz S, Sanders D, Chalot M. Phylogenetic and functional analysis
of the Cation Diffusion Facilitator (CDF) family: improved signature and prediction of substrate
specificity. BMC genomics. 2007; 8:107. [PubMed: 17448255]
84. Blindauer CA, Schmid R. Cytosolic metal handling in plants: determinants for zinc specificity in
metal transporters and metallothioneins. Metallomics: integrated biometal science. 2010; 2:510–
529. [PubMed: 21072336]
Pi and Helmann Page 16
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
85. Podar D, Scherer J, Noordally Z, Herzyk P, Nies D, Sanders D. Metal selectivity determinants in a
family of transition metal transporters. The Journal of biological chemistry. 2012; 287:3185–3196.
[PubMed: 22139846]
86. Kawachi M, Kobae Y, Mimura T, Maeshima M. Deletion of a histidine-rich loop of AtMTP1, a
vacuolar Zn(2+)/H(+) antiporter of
Arabidopsis thaliana
, stimulates the transport activity. The
Journal of biological chemistry. 2008; 283:8374–8383. [PubMed: 18203721]
87. Lin H, Burton D, Li L, Warner DE, Phillips JD, Ward DM, Kaplan J. Gain-of-function mutations
identify amino acids within transmembrane domains of the yeast vacuolar transporter Zrc1 that
determine metal specificity. The Biochemical journal. 2009; 422:273–283. [PubMed: 19538181]
88. Hoch E, Lin W, Chai J, Hershfinkel M, Fu D, Sekler I. Histidine pairing at the metal transport site
of mammalian ZnT transporters controls Zn2+ over Cd2+ selectivity. Proceedings of the National
Academy of Sciences of the United States of America. 2012; 109:7202–7207. [PubMed:
22529353]
89. Grass G, Fan B, Rosen BP, Franke S, Nies DH, Rensing C. ZitB (YbgR), a member of the cation
diffusion facilitator family, is an additional zinc transporter in
Escherichia coli
. Journal of
bacteriology. 2001; 183:4664–4667. [PubMed: 11443104]
90. Chao Y, Fu D. Thermodynamic studies of the mechanism of metal binding to the
Escherichia coli
zinc transporter YiiP. The Journal of biological chemistry. 2004; 279:17173–17180. [PubMed:
14960568]
91. Wei Y, Li H, Fu D. Oligomeric state of the
Escherichia coli
metal transporter YiiP. The Journal of
biological chemistry. 2004; 279:39251–39259. [PubMed: 15258151]
92. Grass G, Otto M, Fricke B, Haney CJ, Rensing C, Nies DH, Munkelt D. FieF (YiiP) from
Escherichia coli
mediates decreased cellular accumulation of iron and relieves iron stress. Archives
of Microbiology. 2005; 183:9–18. [PubMed: 15549269]
93. Wei Y, Fu D. Selective metal binding to a membrane-embedded aspartate in the
Escherichia coli
metal transporter YiiP (FieF). The Journal of biological chemistry. 2005; 280:33716–33724.
[PubMed: 16049012]
94. Lu M, Chai J, Fu D. Structural basis for autoregulation of the zinc transporter YiiP. Nature
structural & molecular biology. 2009; 16:1063–1067.
95. Nies DH. How iron is transported into magnetosomes. Molecular Microbiology. 2011; 82:792–796.
[PubMed: 21999528]
96. Uebe R, Junge K, Henn V, Poxleitner G, Katzmann E, Plitzko JM, Zarivach R, Kasama T, Wanner
G, Posfai M, Bottger L, Matzanke B, Schuler D. The cation diffusion facilitator proteins MamB
and MamM of
Magnetospirillum gryphiswaldense
have distinct and complex functions, and are
involved in magnetite biomineralization and magnetosome membrane assembly. Molecular
Microbiology. 2011; 82:818–835. [PubMed: 22007638]
97. Salusso A, Raimunda D. Defining the Roles of the Cation Diffusion Facilitators in Fe2+/Zn2+
Homeostasis and Establishment of Their Participation in Virulence in
Pseudomonas aeruginosa
.
Frontiers in cellular and infection Microbiology. 2017; 7:84. [PubMed: 28373967]
98. Pederick VG, Eijkelkamp BA, Begg SL, Ween MP, McAllister LJ, Paton JC, McDevitt CA. ZnuA
and zinc homeostasis in
Pseudomonas aeruginosa
. Scientific reports. 2015; 5:13139. [PubMed:
26290475]
99. Hau HH, Gralnick JA. Ecology and biotechnology of the genus
Shewanella
. Annual review of
Microbiology. 2007; 61:237–258.
100. Meyer TE, Tsapin AI, Vandenberghe I, de Smet L, Frishman D, Nealson KH, Cusanovich MA,
van Beeumen JJ. Identification of 42 possible cytochrome C genes in the
Shewanella oneidensis
genome and characterization of six soluble cytochromes. Omics: a journal of integrative biology.
2004; 8:57–77. [PubMed: 15107237]
101. Bennett BD, Brutinel ED, Gralnick JA. A Ferrous Iron Exporter Mediates Iron Resistance in
Shewanella oneidensis
MR-1. Applied and environmental Microbiology. 2015; 81:7938–7944.
[PubMed: 26341213]
102. Yang Y, Harris DP, Luo F, Wu L, Parsons AB, Palumbo AV, Zhou J. Characterization of the
Shewanella oneidensis
Fur gene: roles in iron and acid tolerance response. BMC genomics. 2008;
9(Suppl 1):S11.
Pi and Helmann Page 17
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
103. Yan N. Structural Biology of the Major Facilitator Superfamily Transporters. Annual review of
biophysics. 2015; 44:257–283.
104. Forrest LR, Kramer R, Ziegler C. The structural basis of secondary active transport mechanisms.
Biochimica et biophysica acta. 2011; 1807:167–188. [PubMed: 21029721]
105. Quistgaard EM, Low C, Guettou F, Nordlund P. Understanding transport by the major facilitator
superfamily (MFS): structures pave the way,
Nature reviews
. Molecular cell biology. 2016;
17:123–132. [PubMed: 26758938]
106. Jardetzky O. Simple allosteric model for membrane pumps. Nature. 1966; 211:969–970.
[PubMed: 5968307]
107. Quistgaard EM, Low C, Moberg P, Tresaugues L, Nordlund P. Structural basis for substrate
transport in the GLUT-homology family of monosaccharide transporters. Nature structural &
molecular biology. 2013; 20:766–768.
108. Kumar H, Finer-Moore JS, Kaback HR, Stroud RM. Structure of LacY with an alpha-substituted
galactoside: Connecting the binding site to the protonation site. Proceedings of the National
Academy of Sciences of the United States of America. 2015; 112:9004–9009. [PubMed:
26157133]
109. Kumar H, Kasho V, Smirnova I, Finer-Moore JS, Kaback HR, Stroud RM. Structure of sugar-
bound LacY. Proceedings of the National Academy of Sciences of the United States of America.
2014; 111:1784–1788. [PubMed: 24453216]
110. Dang S, Sun L, Huang Y, Lu F, Liu Y, Gong H, Wang J, Yan N. Structure of a fucose transporter
in an outward-open conformation. Nature. 2010; 467:734–738. [PubMed: 20877283]
111. Yin Y, He X, Szewczyk P, Nguyen T, Chang G. Structure of the multidrug transporter EmrD from
Escherichia coli
. Science (New York, N.Y.). 2006; 312:741–744.
112. Newstead S, Drew D, Cameron AD, Postis VL, Xia X, Fowler PW, Ingram JC, Carpenter EP,
Sansom MS, McPherson MJ, Baldwin SA, Iwata S. Crystal structure of a prokaryotic homologue
of the mammalian oligopeptide-proton symporters, PepT1 and PepT2. The EMBO journal. 2011;
30:417–426. [PubMed: 21131908]
113. Yan H, Huang W, Yan C, Gong X, Jiang S, Zhao Y, Wang J, Shi Y. Structure and mechanism of a
nitrate transporter. Cell reports. 2013; 3:716–723. [PubMed: 23523348]
114. Fukuda M, Takeda H, Kato HE, Doki S, Ito K, Maturana AD, Ishitani R, Nureki O. Structural
basis for dynamic mechanism of nitrate/nitrite antiport by NarK. Nature communications. 2015;
6:7097.
115. Deng D, Sun P, Yan C, Ke M, Jiang X, Xiong L, Ren W, Hirata K, Yamamoto M, Fan S, Yan N.
Molecular basis of ligand recognition and transport by glucose transporters. Nature. 2015;
526:391–396. [PubMed: 26176916]
116. Nomura N, Verdon G, Kang HJ, Shimamura T, Nomura Y, Sonoda Y, Hussien SA, Qureshi AA,
Coincon M, Sato Y, Abe H, Nakada-Nakura Y, Hino T, Arakawa T, Kusano-Arai O, Iwanari H,
Murata T, Kobayashi T, Hamakubo T, Kasahara M, Iwata S, Drew D. Structure and mechanism
of the mammalian fructose transporter GLUT5. Nature. 2015; 526:397–401. [PubMed:
26416735]
117. Baranova N, Nikaido H. The baeSR two-component regulatory system activates transcription of
the yegMNOB (mdtABCD) transporter gene cluster in
Escherichia coli
and increases its
resistance to novobiocin and deoxycholate. Journal of bacteriology. 2002; 184:4168–4176.
[PubMed: 12107134]
118. Leblanc SK, Oates CW, Raivio TL. Characterization of the induction and cellular role of the
BaeSR two-component envelope stress response of
Escherichia coli
. Journal of bacteriology.
2011; 193:3367–3375. [PubMed: 21515766]
119. Nagakubo S, Nishino K, Hirata T, Yamaguchi A. The putative response regulator BaeR stimulates
multidrug resistance of
Escherichia coli
via a novel multidrug exporter system, MdtABC. Journal
of bacteriology. 2002; 184:4161–4167. [PubMed: 12107133]
120. Frawley ER, Crouch ML, Bingham-Ramos LK, Robbins HF, Wang W, Wright GD, Fang FC. Iron
and citrate export by a major facilitator superfamily pump regulates metabolism and stress
resistance in
Salmonella
Typhimurium. Proceedings of the National Academy of Sciences of the
United States of America. 2013; 110:12054–12059. [PubMed: 23821749]
Pi and Helmann Page 18
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
121. Yeowell HN, White JR. Iron requirement in the bactericidal mechanism of streptonigrin.
Antimicrobial agents and chemotherapy. 1982; 22:961–968. [PubMed: 6218780]
122. Bjarnason J, Southward CM, Surette MG. Genomic profiling of iron-responsive genes in
Salmonella enterica
serovar typhimurium by high-throughput screening of a random promoter
library. Journal of bacteriology. 2003; 185:4973–4982. [PubMed: 12897017]
123. Kim SA, Punshon T, Lanzirotti A, Li L, Alonso JM, Ecker JR, Kaplan J, Guerinot ML.
Localization of iron in
Arabidopsis
seed requires the vacuolar membrane transporter VIT1.
Science (New York, N.Y.). 2006; 314:1295–1298.
124. Arosio P, Elia L, Poli M. Ferritin, cellular iron storage and regulation. IUBMB life. 2017; doi:
10.1002/iub.1621
125. Ruangkiattikul N, Bhubhanil S, Chamsing J, Niamyim P, Sukchawalit R, Mongkolsuk S.
Agrobacterium tumefaciens
membrane-bound ferritin plays a role in protection against hydrogen
peroxide toxicity and is negatively regulated by the iron response regulator. FEMS Microbiology
letters. 2012; 329:87–92. [PubMed: 22268462]
126. Johnson DB, Kanao T, Hedrich S. Redox Transformations of Iron at Extremely Low pH:
Fundamental and Applied Aspects. Frontiers in Microbiology. 2012; 3:96. [PubMed: 22438853]
127. Sankari S, O'Brian MR. A bacterial iron exporter for maintenance of iron homeostasis. The
Journal of biological chemistry. 2014; 289:16498–16507. [PubMed: 24782310]
128. Hamza I, Qi Z, King ND, O'Brian MR. Fur-independent regulation of iron metabolism by Irr in
Bradyrhizobium japonicum
. Microbiology (Reading, England). 2000; 146(Pt 3):669–676.
129. Rudolph G, Semini G, Hauser F, Lindemann A, Friberg M, Hennecke H, Fischer HM. The Iron
control element, acting in positive and negative control of iron-regulated
Bradyrhizobium
japonicum
genes, is a target for the Irr protein. Journal of bacteriology. 2006; 188:733–744.
[PubMed: 16385063]
130. Sankari S, O'Brian MR. Synthetic Lethality of the bfr and mbfA Genes Reveals a Functional
Relationship between Iron Storage and Iron Export in Managing Stress Responses in
Bradyrhizobium japonicum
. PloS one. 2016; 11:e0157250. [PubMed: 27285822]
131. Hood MI, Skaar EP. Nutritional immunity: transition metals at the pathogen–host interface. Nat
Rev Micro. 2012; 10:525–537.
132. Flannagan RS, Heit B, Heinrichs DE. Antimicrobial Mechanisms of Macrophages and the
Immune Evasion Strategies of
Staphylococcus aureus
. Pathogens (Basel, Switzerland). 2015;
4:826–868.
133. Osorio H, Martinez V, Nieto PA, Holmes DS, Quatrini R. Microbial iron management
mechanisms in extremely acidic environments: comparative genomics evidence for diversity and
versatility. BMC Microbiology. 2008; 8:203. [PubMed: 19025650]
Pi and Helmann Page 19
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
fig. 1. Iron homeostasis in bacteria
Under iron deficient conditions (left), high affinity iron uptake systems are induced to
scavenge iron from the surroundings to maintain the cell's labile iron pool. when iron is
limiting, it is selectively partitioned to the most essential functions and incorporation into
lower priority iron enzymes is translationally inhibited as part of an iron sparing response. in
many cases, iron-independent enzymes may be derepressed to replace functions that would
otherwise depend on iron. under iron excess conditions, the cell will have a full complement
of iron-requiring enzymes, and iron in excess of immediate needs will be either stored for
future use or exported by fe2+ efflux transporters to prevent iron overload.
Pi and Helmann Page 20
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
fig. 2. Ferrous iron efflux systems in bacteria
Four different groups of transporters can function as fe2+ efflux pumps. i. p1b-atpase; ii.
cation diffusion facilitator (cdf); iii. major facilitator superfamily (mfs); iv. membrane-bound
ferritin. a typical p1b-atpase consists of a transmembrane domain (tmd) that has 6–8 helices,
a soluble actuator domain (not shown), and an atp-binding domain (atp-bd)52. a cdf
transporter contains a n-terminal domain (ntd), a transmembrane domain (tmd) that has 6
helices, a histine-rich interconnecting loop (il) between tm4 and tm5 (not shown), and a c-
terminal cytoplasmic domain (ctd)75. the common structural fold (mfs fold) of a mfs
transporter is composed of two distinct domains, n domain and c domain. each domain has
six consecutive transmembrane helices103. a membrane-bound ferritin transporter has two
major domains, n-terminal ferritin-like or er domain (er) and c-terminal membrane-
embedded vacuolar iron transporter domain (vit1).
Pi and Helmann Page 21
Metallomics
. Author manuscript; available in PMC 2018 July 19.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Pi and Helmann Page 22
Table 1
Fe2+ efflux transporters in bacteria.
Protein Organism Function Category Substrate
specificity*Transcription regulation References
PfeT
Bacillus subtilis
Fe2+ efflux P1B4-type ATPase Fe2+
a
,
b
, Co2+
a
PerR & Fur 36
FrvA
Listeria monocytogenes
Fe2+ efflux P1B4-type ATPase Fe2+
a
,
b
, Co2+
a
, Zn2+
a
PerR & Fur 27
CtpD
Mycobacterium tuberculosis
Fe2+ efflux P1B4-type ATPase Fe2+
a
,
b
, Co2+
a
Unknown 37
PmtA group A
Streptococcus
Fe2+ efflux P1B4-type ATPase Fe2+
b
PerR 38, 39
Nia
Sinorhizobium meliloti
Fe2+ or Ni2+ efflux P1B5-type ATPase Fe2+
a
,
b
, Ni2+
a
,
b
Unknown 73
FieF (YiiP)
Escherichia coli
Fe2+ or Zn2+ efflux CDF family Fe2+
b
, Zn2+
a
, Cd2+
a
Unknown 92–94
AitP
Pseudomonas aeruginosa
Fe2+ or Co2+ efflux CDF family Fe2+
b
Unknown 97
FeoE
Shewanella oneidensis
Fe2+ efflux CDF family Fe2+
b
Unknown 101
IceT
Salmonella typhimurium
Fe2+ citrate or citrate efflux MFS family Fe2+
b
BaeSR 120
MbfA
Agrobacterium tumefaciens
Fe2+ efflux Membrane bound ferritin Fe2+
b
Irr 125
MbfA
Bradyrhizobium japonicum
Fe2+ efflux Membrane bound ferritin Fe2+
b
Irr 127
*
Note: the substrate specificity of the transporters is either based on biochemical measurements (a), inferred from physiology studies (b), or both (a, b).
Metallomics
. Author manuscript; available in PMC 2018 July 19.
... In many environments, including the human gut, iron is a limiting nutrient for growth, and high-affinity uptake systems play a central role in ferrous iron homeostasis 40 . Maintenance of ferrous iron homeostasis is crucial for the production of butyrate 37 because this process requires two iron-binding ferredoxins as illustrated in Fig. 3A. ...
... Interestingly, together with upregulation of feoAABC, we also detected upregulation of a flavodoxin-encoding gene (see Supplementary Table S4 online). This is consistent with previous studies reporting that iron-free flavodoxin replaces iron-sulfur ferredoxin under iron-limited conditions 40,41 . Because flavodoxin and ferredoxin both mediate electron transfer in redox processes, the use of flavodoxin could free up iron for utilization by other iron-dependent enzymes. ...
... Because flavodoxin and ferredoxin both mediate electron transfer in redox processes, the use of flavodoxin could free up iron for utilization by other iron-dependent enzymes. In other word, flavodoxins are key players in maintenance of ferrous iron homeostasis in iron-limited environment 40,41 . For example, in C. difficile, the production of flavodoxin is tied to iron homeostasis in iron-limited conditions 42 . ...
Article
Full-text available
The promising next-generation probiotic Faecalibacterium prausnitzii is one of the most abundant acetate-consuming, butyrate-producing bacteria in the healthy human gut. Yet, little is known about how acetate availability affects this bacterium’s gene expression strategies. Here, we investigated the effect of acetate on temporal changes in the transcriptome of F. duncaniae A2-165 cultures using RNA sequencing. We compared gene expression patterns between two growth phases (early stationary vs. late exponential) and two acetate levels (low: 3 mM vs. high: 23 mM). Only in low-acetate conditions, a general stress response was activated. In high-acetate conditions, there was greater expression of genes related to butyrate synthesis and to the importation of B vitamins and iron. Specifically, expression was strongly activated in the case of the feoAABC operon, which encodes a FeoB ferrous iron transporter, but not in the case of the feoAB gene, which encodes a second putative FeoAB transporter. Moreover, excess ferrous iron repressed feoB expression but not feoAB. Lastly, FeoB but not FeoAB peptides from strain A2-165 were found in abundance in a healthy human fecal metaproteome. In conclusion, we characterized two early-stationary transcriptomes based on acetate consumption and this work highlights the regulation of feoB expression in F. duncaniae A2-165.
... Over the last decade, iron export systems have been described in several bacterial species that mitigate iron intoxication and H 2 O 2 exposure (Reviewed in [Pi & Helmann, 2017]). There are several known families of iron exporters, including P-type ATPases (Guan et al., 2015;Patel et al., 2016;Pi et al., 2016;Turner et al., 2017;VanderWal et al., 2017;Zielazinski et al., 2013), cation diffusion facilitator (CDF) proteins (Bennett et al., 2015;Grass et al., 2005;Salusso & Raimunda, 2017), major facilitator superfamily (MFS) proteins (Frawley et al., 2013) and membrane-bound ferritin-like proteins (Bhubhanil et al., 2014;Sankari & O'Brian, 2014). ...
... 2.6 | mbfA, but not ihpA, is required to confer resistance to hydrogen peroxide exposure Iron generates reactive oxygen species to induce oxidative stress, and iron export has been shown to confer resistance to induced oxidative stress in B. japonicum (Sankari & O'Brian, 2014, 2016b and other bacteria (Pi & Helmann, 2017). Here, we wanted to determine whether IhpABC contributes to H 2 O 2 stress response. ...
... Prior work characterizes iron export as a mechanism to cope with high iron and oxidative stresses (Reviewed in [Pi & Helmann, 2017]). ...
Article
Full-text available
Bacterial iron export mitigates high iron stress, but a role for it under lower iron conditions has not been established. MbfA is the high iron stress exporter in Bradyrhizobium japonicum. Here, we identify the ihpABC genes in a selection for secondary site mutations that suppress the poor growth phenotype of feoAB mutants defective in iron acquisition. IhpABC belongs to the RND tripartite efflux pump family. High iron conditions that derepress the mbfA gene partially rescued the growth of an ihpC mutant but reverted the feoB ihpC mutant to the feoB growth phenotype. The ihpA mutant grown under low iron conditions accumulated higher levels of iron compared to the wild type, and it displayed aberrant iron‐responsive gene expression. The mbfA mutant was more sensitive than the wild type to H2O2, but the ihpA mutant was not sensitive. The ihpA mutant accumulated more Zn, Co and Cd than was found in the wild type, and growth of the mutant was more sensitive to inhibition by ZnCl2, CoCl2 and CdCl2. The findings suggest that IhpABC is a divalent metal ion exporter that helps maintain iron homeostasis under low to moderate environmental iron levels. Thus, iron export is not limited to managing high iron stress.
... Iron is indispensable for almost all forms of life but toxic at elevated levels [1][2][3][4] . To survive within their hosts, bacterial pathogens have evolved iron uptake, storage and detoxification strategies to maintain iron homeostasis 1,5,6 . ...
... Iron is indispensable for almost all forms of life but toxic at elevated levels [1][2][3][4] . To survive within their hosts, bacterial pathogens have evolved iron uptake, storage and detoxification strategies to maintain iron homeostasis 1,5,6 . Recent studies showed that three Gram-negative environmental anaerobes produce iron-containing ferrosome granules 7,8 . ...
Article
Full-text available
Iron is indispensable for almost all forms of life but toxic at elevated levels1–4. To survive within their hosts, bacterial pathogens have evolved iron uptake, storage and detoxification strategies to maintain iron homeostasis1,5,6. Recent studies showed that three Gram-negative environmental anaerobes produce iron-containing ferrosome granules7,8. However, it remains unclear whether ferrosomes are generated exclusively by Gram-negative bacteria. The Gram-positive bacterium Clostridioides difficile is the leading cause of nosocomial and antibiotic-associated infections in the USA⁹. Here we report that C. difficile undergoes an intracellular iron biomineralization process and stores iron in membrane-bound ferrosome organelles containing non-crystalline iron phosphate biominerals. We found that a membrane protein (FezA) and a P1B6-ATPase transporter (FezB), repressed by both iron and the ferric uptake regulator Fur, are required for ferrosome formation and play an important role in iron homeostasis during transition from iron deficiency to excess. Additionally, ferrosomes are often localized adjacent to cellular membranes as shown by cryo-electron tomography. Furthermore, using two mouse models of C. difficile infection, we demonstrated that the ferrosome system is activated in the inflamed gut to combat calprotectin-mediated iron sequestration and is important for bacterial colonization and survival during C. difficile infection.
... To maintain the balance of metal ion concentrations, microbes can use metal transporters to transport metals into various cellular membrane compartments [28]. Efflux mechanisms are critical in the resistance of bacteria to a diverse array of heavy metals [29]. Among these mechanisms, exporting P-type ATPases can efficiently promote the detoxification of heavy metal cations by exporting inorganic substrates from the cytoplasm to the external environment or periplasm [30]. ...
... The vacuolar iron transporter (VIT) proteins are involved in transport of iron and have been studied mainly in plants and yeasts, although homologs have also been identified in other eukaryotes and in bacteria and archaea [49]. For example, the membrane-bound ferritin (MbfA) protein belonging to the erythrin-vacuolar iron transport (Er-VIT1) family may function physiologically as an iron efflux transporter [29,50]. In this study, 40 MAGs possessed the gene encoding VIT. ...
Article
Full-text available
Background Ferromanganese nodule-bearing deep-sea sediments cover vast areas of the ocean floor, representing a distinctive habitat in the abyss. These sediments harbor unique conditions characterized by high iron concentration and low degradable nutrient levels, which pose challenges to the survival and growth of most microorganisms. While the microbial diversity in ferromanganese nodule-associated sediments has been surveyed several times, little is known about the functional capacities of the communities adapted to these unique habitats. Results Seven sediment samples collected adjacent to ferromanganese nodules from the Clarion–Clipperton Fracture Zone (CCFZ) in the eastern Pacific Ocean were subjected to metagenomic analysis. As a result, 179 high-quality metagenome-assembled genomes (MAGs) were reconstructed and assigned to 21 bacterial phyla and 1 archaeal phylum, with 88.8% of the MAGs remaining unclassified at the species level. The main mechanisms of resistance to heavy metals for microorganisms in sediments included oxidation (Mn), reduction (Cr and Hg), efflux (Pb), synergy of reduction and efflux (As), and synergy of oxidation and efflux (Cu). Iron, which had the highest content among all metallic elements, may occur mainly as Fe(III) that potentially functioned as an electron acceptor. We found that microorganisms with a diverse array of CAZymes did not exhibit higher community abundance. Instead, microorganisms mainly obtained energy from oxidation of metal (e.g., Mn(II)) and sulfur compounds using oxygen or nitrate as an electron acceptor. Chemolithoautotrophic organisms (Thaumarchaeota and Nitrospirota phyla) were found to be potential manganese oxidizers. The functional profile analysis of the dominant microorganisms further indicated that utilization of inorganic nutrients by redox reactions (rather than organic nutrient metabolism) is a major adaptive strategy used by microorganisms to support their survival in the ferromanganese nodule sediments. Conclusions This study provides a comprehensive metagenomic analysis of microbes inhabiting metal-rich ferromanganese nodule sediments. Our results reveal extensive redundancy across taxa for pathways of metal resistance and transformation, the highly diverse mechanisms used by microbes to obtain nutrition, and their participation in various element cycles in these unique environments. 7UX-FAbsHcrvGN6PxGni8JVideo Abstract
... Even though Mn and Fe are relatively weak-binding metals (at the lower affinity end of the Irving-Williams series), cells can still experience metal intoxication when their homeostasis systems are disrupted. In B. subtilis, Fe intoxication is apparent in strains lacking the PfeT Fe efflux ATPase (44,45), and Mn intoxication is apparent in efflux-deficient PS strains (9,11) and, as shown here, in the absence of the YS operon. The molecular basis for intoxication by these more weakly interacting metal ions is not well understood, but previous results have suggested that excess Mn may interfere with Mg-dependent processes (14,46). ...
Article
Full-text available
Microbes encounter a myriad of stresses during their life cycle. Dysregulation of metal ion homeostasis is increasingly recognized as a key factor in host–microbe interactions. Bacterial metal ion homeostasis is tightly regulated by dedicated metalloregulators that control uptake, sequestration, trafficking, and efflux. Here, we demonstrate that deletion of the Bacillus subtilis yqgC-sodA (YS) complex operon, but not deletion of the individual genes, causes hypersensitivity to manganese (Mn). YqgC is an integral membrane protein of unknown function, and SodA is a Mn-dependent superoxide dismutase (MnSOD). The YS strain has reduced expression of two Mn efflux proteins, MneP and MneS, consistent with the observed Mn sensitivity. The YS strain accumulated high levels of Mn, had increased reactive radical species (RRS), and had broad metabolic alterations that can be partially explained by the inhibition of Mg-dependent enzymes. Although the YS operon deletion strain and an efflux-deficient mneP mneS double mutant both accumulate Mn and have similar metabolic perturbations, they also display phenotypic differences. Several mutations that suppressed Mn intoxication of the mneP mneS efflux mutant did not benefit the YS mutant. Further, Mn intoxication in the YS mutant, but not the mneP mneS strain, was alleviated by expression of Mg-dependent, chorismate-utilizing enzymes of the menaquinone, siderophore, and tryptophan (MST) family. Therefore, despite their phenotypic similarities, the Mn sensitivity in the mneP mneS and the YS deletion mutants results from distinct enzymatic vulnerabilities. IMPORTANCE Bacteria require multiple trace metal ions for survival. Metal homeostasis relies on the tightly regulated expression of metal uptake, storage, and efflux proteins. Metal intoxication occurs when metal homeostasis is perturbed and often results from enzyme mis-metalation. In Bacillus subtilis, Mn-dependent superoxide dismutase (MnSOD) is the most abundant Mn-containing protein and is important for oxidative stress resistance. Here, we report novel roles for MnSOD and a co-regulated membrane protein, YqgC, in Mn homeostasis. Loss of both MnSOD and YqgC (but not the individual proteins) prevents the efficient expression of Mn efflux proteins and leads to a large-scale perturbation of the metabolome due to inhibition of Mg-dependent enzymes, including key chorismate-utilizing MST (menaquinone, siderophore, and tryptophan) family enzymes.
... Flagellated bacteria have been shown to move through albumen and reach the yolk surface, allowing for rapid reproduction [82]. While no tests have been performed to determine whether L. monocytogenes interacts with the egg albumen, the presence of flagella, iron scavenging proteins (FrvA), and a high pH tolerance (3.0 to 9.5) suggests the bacterium could survive and reproduce within the egg yolk if it were present [94][95][96][97]. ...
Article
Full-text available
Listeria continues to be a persistent foodborne pathogen that is responsible for human cases of listeriosis when contaminated food products are consumed. Human subjects considered to be most susceptible include the elderly, immunocompromised, and pregnant women. Listeria is characterized as a saprophytic organism with the capability of responding and adapting to constantly changing environments because it possesses multiple stress response mechanisms to overcome varying temperatures, salt concentrations, and pH, among others. Primary foods and food products associated with listeriosis include dairy products and ready-to-eat meats such as turkey products. Historically, chicken eggs have not been identified as a primary source of Listeria, but the potential for contamination during egg production and processing does exist. Listeria species have been isolated from egg-processing plant equipment and are presumed to occur in egg-processing plant environments. Whether Listeria is consistently disseminated onto eggs beyond the egg-processing plant is a risk factor that remains to be determined. However, research has been conducted over the years to develop egg wash solutions that generate combinations of pH and other properties that would be considered inhibitory to Listeria. Even less is known regarding the association of Listeria with alternative egg production systems, but Listeria has been isolated from pasture flock broilers, so it is conceivable, given the nature of the outdoor environments, that layer birds under these conditions would also be exposed to Listeria and their eggs become contaminated. This review focuses on the possibility of Listeria occurring in conventional and alternative egg-laying production and processing systems.
Article
Background: Riemerella anatipestifer encodes an iron acquisition system, but whether it encodes the iron efflux pump and its role in antibiotic resistance are largely unknown. Objectives: To screen and identify an iron efflux gene in R. anatipestifer and determine whether and how the iron efflux gene is involved in antibiotic resistance. Methods: In this study, gene knockout, streptonigrin susceptibility assay and inductively coupled plasma mass spectrometry were used to screen for the iron efflux gene ietA. The MIC measurements, scanning electron microscopy and reactive oxygen species (ROS) detection were used to verify the role of IetA in aztreonam resistance and its mechanism. Mortality and colonization assay were used to investigate the role of IetA in virulence. Results: The deletion mutant ΔietA showed heightened susceptibility to streptonigrin, and prominent intracellular iron accumulation was observed in ΔfurΔietA under excess iron conditions. Additionally, ΔietA exhibited increased sensitivity to H2O2-produced oxidative stress. Under aerobic conditions with abundant iron, ΔietA displayed increased susceptibility to the β-lactam antibiotic aztreonam due to heightened ROS production. However, the killing efficacy of aztreonam was diminished in both WT and ΔietA under anaerobic or iron restriction conditions. Further experiments demonstrated that the efficiency of aztreonam against ΔietA was dependent on respiratory complexes Ⅰ and Ⅱ. Finally, in a duckling model, ΔietA had reduced virulence compared with the WT. Conclusion: Iron efflux is critical to alleviate oxidative stress damage and β-lactam aztreonam killing in R. anatipestifer, which is linked by cellular respiration.
Preprint
Full-text available
Microbes encounter a myriad of stresses during their life cycle. Dysregulation of metal ion homeostasis is increasingly recognized as a key factor in host-microbe interactions. Bacterial metal ion homeostasis is tightly regulated by dedicated metalloregulators that control uptake, sequestration, trafficking, and efflux. Here, we demonstrate that deletion of the Bacillus subtilis yqgC-sodA (YS) complex operon, but not deletion of the individual genes, causes hypersensitivity to manganese (Mn). YqgC is an integral membrane protein of unknown function and SodA is a Mn-dependent superoxide dismutase (MnSOD). The YS strain has reduced expression of two Mn efflux proteins, MneP and MneS, consistent with the observed Mn sensitivity. The YS strain accumulated high levels of Mn, had increased reactive radical species (RRS), and had broad metabolic alterations that can be partially explained by the inhibition of Mg-dependent enzymes. Although the YS operon deletion strain and an efflux-deficient mneP mneS double mutant both accumulate Mn and have similar metabolic perturbations they also display phenotypic differences. Several mutations that suppressed Mn intoxication of the mneP mneS efflux mutant did not benefit the YS mutant. Further, Mn intoxication in the YS mutant, but not the mneP mneS strain, was alleviated by expression of Mg-dependent, chorismate-utilizing enzymes of the m enaquinone, s iderophore, and tryptophan (MST) family. Therefore, despite their phenotypic similarities, the Mn sensitivity in the mneP mneS and the yqgC-sodA deletion mutants results from distinct enzymatic vulnerabilities. Importance Bacteria require multiple trace metal ions for survival. Metal homeostasis relies on the tightly regulated expression of metal uptake, storage, and efflux proteins. Metal intoxication occurs when metal homeostasis is perturbed and often results from enzyme mis-metalation. In Bacillus subtilis , MnSOD is the most abundant Mn-containing protein and is important for oxidative stress resistance. Here, we report novel roles for MnSOD and a co-regulated membrane protein, YqgC, in Mn homeostasis. Loss of both MnSOD and YqgC (but not the individual mutations) prevents the efficient expression of Mn efflux proteins and leads to a large-scale perturbation of the metabolome due to inhibition of Mg-dependent enzymes, including key chorismate-utilizing MST (menaquinone, siderophore, and tryptophan) family enzymes.
Article
Environmental compartments polluted with animal charcoal from the skin and hide cottage industries are rich in toxic heavy metals and diverse hydrocarbon classes, some of which are carcinogenic, mutagenic, and genotoxic, and thus require a bio-based eco-benign decommission strategies. A shotgun metagenomic approach was used to decipher the microbiome, hydrocarbon degradation genes, and heavy metal resistome of a microbial consortium (FN8) from an animal-charcoal polluted site enriched with fluorene. Structurally, the FN8 microbial consortium consists of 26 phyla, 53 classes, 119 orders, 245 families, 620 genera, and 1021 species. The dominant phylum, class, order, family, genus, and species in the consortium are Proteobacteria (51.37%), Gammaproteobacteria (39.01%), Bacillales (18.09%), Microbulbiferaceae (11.65%), Microbulbifer (12.21%), and Microbulbifer sp. A4B17 (19.65%), respectively. The microbial consortium degraded 57.56% (28.78 mg/L) and 87.14% (43.57 mg/L) of the initial fluorene concentration in 14 and 21 days. Functional annotation of the protein sequences (ORFs) of the FN8 metagenome using the KEGG GhostKOALA, KofamKOALA, NCBI’s conserved domain database, and BacMet revealed the detection of hydrocarbon degradation genes for benzoate, aminobenzoate, polycyclic aromatic hydrocarbons (PAHs), chlorocyclohexane/chlorobenzene, chloroalkane/chloroalkene, toluene, xylene, styrene, naphthalene, nitrotoluene, and several others. The annotation also revealed putative genes for the transport, uptake, efflux, and regulation of heavy metals such as arsenic, cadmium, chromium, mercury, nickel, copper, zinc, and several others. Findings from this study have established that members of the FN8 consortium are well-adapted and imbued with requisite gene sets and could be a potential bioresource for on-site depuration of animal charcoal polluted sites.
Article
Full-text available
Streptococcus pyogenes (group A Streptococcus; GAS) is an obligate human pathogen responsible for a broad spectrum of human disease. GAS has a requirement for metal homeostasis within the human host and as such, tightly modulates metal uptake and efflux during infection. Metal acquisition systems are required to combat metal sequestration by the host, while metal efflux systems are essential to protect against metal overload poisoning. Here, we investigated the function of PmtA (PerR-regulated metal transporter A), a P1B-4 type ATPase efflux pump, in the invasive GAS M1T1 strain 5448. We reveal that PmtA functions as a ferrous iron [Fe(II)] efflux system. In the presence of high Fe(II) concentrations, the 5448?pmtA deletion mutant exhibited diminished growth and accumulated 5-fold higher intracellular Fe(II) compared to the wild-type and complemented mutant. The 5448?pmtA deletion mutant also showed enhanced susceptibility to killing by the Fe-dependent antibiotic, streptonigrin, as well as increased sensitivity to hydrogen peroxide and superoxide. We suggest that the PerR-mediated control of Fe(II) efflux by PmtA is important for bacterial defense against oxidative stress. PmtA represents an exemplar for a Fe(II) efflux system in a host-adapted Gram-positive bacterial pathogen.
Article
Full-text available
Group A streptococcus (GAS) is a human-only pathogen that causes a spectrum of disease conditions. Given its survival in inflamed lesions, the ability to sense and overcome oxidative stress is critical for GAS pathogenesis. PerR senses oxidative stress and coordinates the regulation of genes involved in GAS antioxidant defenses. In this study, we investigated the role of PerR-controlled metal transporter A (PmtA) in GAS pathogenesis. Previously, PmtA was implicated in GAS antioxidant defenses and suggested to protect against zinc toxicity. Here, we report that PmtA is a P1B4-type ATPase that functions as an Fe(II) exporter and aids GAS defenses against iron intoxication and oxidative stress. Expression of pmtA is specifically induced by iron excess and this induction requires PerR. Furthermore, the pmtA mutant exhibited increased sensitivity to iron toxicity and oxidative stress due to elevated intracellular accumulation of iron. RNA-sequencing analysis revealed that GAS undergoes significant alterations in gene expression to adapt to iron toxicity. Finally, using two mouse models of invasive infection, we demonstrated that iron efflux by PmtA is critical for bacterial survival during infection and GAS virulence. Together, these data demonstrate that PmtA is a key component of GAS antioxidant defenses and contributes significantly to GAS virulence.
Article
Full-text available
Transporters of the cation diffusion facilitator (CDF) family form dimers that export transition metals from the cytosol. The opportunistic pathogen Pseudomonas aeruginosa encodes three homologous CDF genes, czcD (PA0397), aitP (PA1297), and yiiP (PA3963). The three proteins are required for virulence in a plant host model. Disruption of the aitP gene leads to higher Fe²⁺ and Co²⁺ sensitivity together with an intracellular accumulation of these ions and to a decreased survival in presence of H2O2. Strains lacking czcD and yiiP showed low Zn²⁺ sensitivity. However, in iron-rich media and in the presence of Zn²⁺ these strains secreted higher levels of the iron chelator pyoverdine. Disruption of czcD and yiiP in a non-pyoverdine producer strain and lacking the Zn²⁺-transporting ATPase, increased the Zn²⁺ sensitivity and the accumulation of this ion. Most importantly, independent of the pyoverdine production strains lacking CzcD or YiiP, presented lower resistance to imipenem, ciprofloxacin, chloramphenicol, and gentamicin. These observations correlated with a lower survival rate upon EDTA-lysozyme treatment and overexpression of OprN and OprD porins. We hypothesize that while AitP is an Fe²⁺/Co²⁺ efflux transporter required for Fe²⁺ homeostasis, and ultimately redox stress handling, CzcD, and YiiP export Zn²⁺ to the periplasm for proper Zn²⁺-dependent signaling regulating outer membrane stability and therefore antibiotic tolerance.
Article
Full-text available
Mycobacterium tuberculosis requires iron for its normal growth but faces limitation of the metal ion due to its low solubility at biological pH and the withholding of iron by the mammalian host. The pathogen expresses Fe 3+ -specific siderophores mycobactin and carboxymycobactin to chelate the metal ion from insoluble iron and the host proteins transferrin, lactoferrin and ferritin. Siderophore-mediated iron uptake is essential for the survival of M. tuberculosis as knock out mutants, defective in siderophore synthesis or uptake failed to survive in low iron medium and inside macrophages. But as excess iron is toxic due to its catalytic role in the generation of free radicals, regulation of iron uptake is necessary to maintain optimal levels of intracellular iron. The focus of this review is to present a comprehensive overview of iron homeostasis in M. tuberculosis that is discussed in the context of mycobactin biosynthesis, transport of iron across the mycobacterial cell envelope and storage of excess iron. The clinical significance of the serum iron status and the expression of the iron-regulated protein HupB in tuberculosis (TB) patients is presented, highlighting the potential of HupB as a marker, notably in extrapulmonary TB cases.
Article
Full-text available
An mbfA mutant of Bradyrhizobium japonicum defective in iron export is sensitive to short term exposure to high levels iron or H2O2. Here, we found that the mbfA strain grown in elevated iron media (100 μM) became resistant to those treatments, suggesting a stress response adaptation. The bfr gene encodes the iron storage protein bacterioferritin, and its expression is derepressed by iron. An mbfA bfr double mutant showed a loss of stress adaptation, and had a severe growth phenotype in high iron media. Moreover, a bfrup allele in which bfr is constitutively derepressed conferred stress tolerance on an mbfA mutant without elevating the iron content in the growth media. The intracellular iron content of the mbfA bfr double mutant was substantially higher than that found in the wild type, even when grown in relatively low iron media (5 μM). Under that condition, iron-responsive gene expression was aberrant in the mbfA bfr strain. Moreover, the double mutant was sensitive to the iron-activated antibiotic streptonigrin. We conclude that MbfA and Bfr work in concert to manage iron and oxidative stresses. In addition, the need for iron detoxification is not limited to extreme environments, but is also required for normal cellular function.
Article
Full-text available
Little is known about iron efflux transporters within bacterial systems. Recently, the participation of Bacillus subtilis PfeT, a P1B4-ATPase, in cytoplasmic Fe2+ efflux has been proposed. We report here the distinct roles of mycobacterial P1B4-ATPases in the homeostasis of Co2+ and Fe2+. Mutation of Mycobacterium smegmatis ctpJ affects the homeostasis of both ions. Alternatively, a M. tuberculosis ctpJ mutant is more sensitive to Co2+ than Fe2+, while mutation of the homologous M. tuberculosis ctpD leads to Fe2+ sensitivity but no alterations in Co2+ homeostasis. In vitro, the three enzymes are activated by both Fe2+ and Co2+ and bind one equivalent of either ion at their transport site. However, equilibrium binding affinities and activity kinetics show that M. tuberculosis CtpD has higher affinity for Fe2+ and twice the Fe2+ stimulated activity than the CtpJs. These parameters are paralleled by a lower activation by and affinity for Co2+. Analysis of Fe2+ and Co2+ binding to CtpD by X-ray spectroscopy shows both ions are 5-6 coordinate, constrained within O/N environments with similar geometries. Mutagenesis studies suggest the involvement of invariant Ser, His and Glu in metal coordination. Interestingly, replacement of Cys in the conserved CPS at the metal binding pocket leads to a large reduction in Fe2+ but not Co2+ binding affinity. We propose that CtpJ ATPases participate in the control of steady state Fe2+ levels. CtpD, required for M. tuberculosis virulence, is a high affinity Fe2+ transporter involved in the rapid response to iron dyshomeostasis generated upon redox stress.
Article
Group A streptococcus (GAS) is a human-only pathogen that causes a spectrum of disease conditions. Given its survival in inflamed lesions, the ability to sense and overcome oxidative stress is critical for GAS pathogenesis. PerR senses oxidative stress and coordinates the regulation of genes involved in GAS antioxidant defenses. In this study, we investigated the role of PerR-controlled metal transporter A (PmtA) in GAS pathogenesis. Previously, PmtA was implicated in GAS antioxidant defenses and suggested to protect against zinc toxicity. Here, we report that PmtA is a P1B4-type ATPase that functions as an Fe(II) exporter and aids GAS defenses against iron intoxication and oxidative stress. Expression of pmtA is specifically induced by iron excess and this induction requires PerR. Furthermore, the pmtA mutant exhibited increased sensitivity to iron toxicity and oxidative stress due to elevated intracellular accumulation of iron. RNA-sequencing analysis revealed that GAS undergoes significant alterations in gene expression to adapt to iron toxicity. Finally, using two mouse models of invasive infection, we demonstrated that iron efflux by PmtA is critical for bacterial survival during infection and GAS virulence. Together, these data demonstrate that PmtA is a key component of GAS antioxidant defenses and contributes significantly to GAS virulence.
Article
Ferritin is considered the major iron storage protein which maintains a large iron core in its cavity and has ferroxidase activity. There are many types of ferritin particularly in prokaryotes that include the canonical 24-mer FTN molecules, the heme-containing BFR, the smaller 12-mer DPS and the newly recognized EncFtn of encapsulin that forms a very large iron storage compartment. Recent studies show that ferritin function is more dynamic than previous depicted and new mechanisms of ferritin iron recycling are emerging. They participate to the regulation of cellular iron homeostasis as those of ferritin biosynthesis, cooperating also with the iron-dependent mechanism of cellular iron secretion. Some of these basic processes are in common between unicellular and animal cells, and this review aims at discussing the findings on the connections between iron storage, cellular iron regulation and ferritin iron recycling that have been explored in unicellular organisms and in animals. ? 2017 IUBMB Life, 2017.
Article
Metal ions are essential for many reactions, but excess metals can be toxic. In bacteria, metal limitation activates pathways that are involved in the import and mobilization of metals, whereas excess metals induce efflux and storage. In this Review, we highlight recent insights into metal homeostasis, including protein-based and RNA-based sensors that interact directly with metals or metal-containing cofactors. The resulting transcriptional response to metal stress takes place in a stepwise manner and is reinforced by post-transcriptional regulatory systems. Metal limitation and intoxication by the host are evolutionarily ancient strategies for limiting bacterial growth. The details of the resulting growth restriction are beginning to be understood and seem to be organism-specific.
Chapter
The metals manganese, iron, cobalt, nickel, copper and zinc are essential for almost all bacteria, but their precise metal requirements vary by species, by ecological niche and by growth condition. Bacteria thus must acquire each of these essential elements in sufficient quantity to satisfy their cellular demand, but in excess these same elements are toxic. Metal toxicity has been exploited by humanity for centuries, and by the mammalian immune system for far longer, yet the mechanisms by which these elements cause toxicity to bacteria are not fully understood. There has been a resurgence of interest in metal toxicity in recent decades due to the problematic spread of antibiotic resistance amongst bacterial pathogens, which has led to an increased research effort to understand these toxicity mechanisms at the molecular level. A recurring theme from these studies is the role of intermetal competition in bacterial metal toxicity.