You are on page 1of 21

Metallomics

View Article Online


MINIREVIEW View Journal | View Issue

Iron–sulfur cluster biosynthesis and trafficking –


impact on human disease conditions
Cite this: Metallomics, 2018,
10, 9
C. Wachnowsky,†ab I. Fidai†ac and J. A. Cowan *abc

Iron–sulfur clusters (Fe–S) are one of the most ancient, ubiquitous and versatile classes of metal cofactors
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

found in nature. Proteins that contain Fe–S clusters constitute one of the largest families of proteins, with
varied functions that include electron transport, regulation of gene expression, substrate binding and activa-
tion, radical generation, and, more recently discovered, DNA repair. Research during the past two decades
has shown that mitochondria are central to the biogenesis of Fe–S clusters in eukaryotic cells via a con-
served cluster assembly machinery (ISC assembly machinery) that also controls the synthesis of Fe–S clus-
ters of cytosolic and nuclear proteins. Several key steps for synthesis and trafficking have been determined
for mitochondrial Fe–S clusters, as well as the cytosol (CIA – cytosolic iron–sulfur protein assembly), but
detailed mechanisms of cluster biosynthesis, transport, and exchange are not well established. Genetic
Received 12th June 2017, mutations and the instability of certain steps in the biosynthesis and maturation of mitochondrial, cytosolic
Accepted 26th September 2017 and nuclear Fe–S cluster proteins affects overall cellular iron homeostasis and can lead to severe metabolic,
DOI: 10.1039/c7mt00180k systemic, neurological and hematological diseases, often resulting in fatality. In this review we briefly sum-
marize the current molecular understanding of both mitochondrial ISC and CIA assembly machineries, and
rsc.li/metallomics present a comprehensive overview of various associated inborn human disease states.

Significance to metallomics
Herein, we summarize current models for iron–sulfur cluster biogenesis, which reflects the complex and intricate series of cellular exchange pathways required
for proper iron homeostasis and metal cofactor trafficking. Given the complexity of iron–sulfur cluster assembly and delivery, which begins in the mitochondria
and includes a mechanism for nuclear delivery, malfunctions in these trafficking processes can lead to a number of disease conditions. In this review we report
on Fe–S cluster-based diseases that have been attributed to defects in cluster trafficking proteins and pathways and can lead to lethal consequences.

Introduction severe threat to the genome due to cytotoxicity via DNA


damage.2a,3 As such, highly conserved and controlled mecha-
Iron–sulfur (Fe–S) clusters are small inorganic cofactors, com- nisms are in place for cluster production and trafficking to
posed of iron and sulfur. Protein-bound Fe–S clusters are among target proteins to allow for their wide range of function as
the most structurally and functionally versatile cofactors across protein cofactors.
all kingdoms of life.1 These cofactors are evolutionarily conserved Over the last decade, several new roles associated with Fe–S
and ubiquitous in nature,2 with the ability to be formed sponta- clusters have emerged, both as a result of progress in identify-
neously in solution from Fe2+/Fe3+, RS , and inorganic S2 , or ing and purifying proteins with intact clusters, and also due to
from Fe2+, R-SH and sulfur under reducing conditions. However, the higher level of sophistication of the biophysical techniques
Fe–S clusters are susceptible to oxidation, presenting challenges used in their characterization. Yet the presence of these clusters
for cluster synthesis and binding to proteins due to atmospheric remains puzzling, given the challenges present with oxygen and
oxygen. In addition, the iron-mediated Fenton reaction poses a the potential for cellular toxicity. This raises the fundamental
question of why Fe–S clusters have been retained as a vital
component of essential macromolecules ranging from proteins
a
Department of Chemistry and Biochemistry, The Ohio State University, to nucleic acids, and highlights the need for mechanistic and
100 West 18th Avenue, Columbus, Ohio 43210, USA.
biochemical studies to promote understanding of the role of these
E-mail: cowan@chemistry.ohio-state.edu
b
The Ohio State Biochemistry Program, The Ohio State University, USA
versatile cofactors. With recent advances, a variety of disease
c
The Biophysics Graduate Program, The Ohio State University, USA conditions have been associated with Fe–S cluster proteins that
† These authors contributed equally to this work. impact overall cluster trafficking. Herein, we briefly review the

This journal is © The Royal Society of Chemistry 2018 Metallomics, 2018, 10, 9--29 | 9
View Article Online

Minireview Metallomics

cluster biogenesis process and describe the associated inborn proteins in vitro. The overview presented here summarizes a
human disease phenotypes. compilation of this research, with the goal of primarily describ-
ing the current model for the human system.

Mitochondrial Fe–S cluster biogenesis 1. Cluster assembly on the scaffold protein


Elucidation of the molecular and cellular basis of Fe–S cluster The first step in Fe–S cluster biogenesis is the assembly of iron
biogenesis and transfer to putative Fe–S cluster proteins has and sulfur on the scaffold protein IscU. Sulfur is provided by
been an area of intense investigation since it was discovered the eukaryotic cysteine desulfurase complex of NFS1–LYRM4
that biogenesis is not spontaneous, but rather catalyzed.4 Three (human ortholog of the Nfs1–Isd11 complex), which converts
distinct pathways have been identified for bacterial Fe–S cluster cysteine to alanine through a persulfide intermediate (Fig. 1
biosynthesis.5 The ISC system is considered the major Fe–S and Table 1).6 The precise details of the assembly complex of
cluster assembly system in humans,1a,2b required not only for Iscu–Isd11–Nfs are somewhat unclear, as larger complexes have
the maturation of mitochondrial but also cytosolic and nuclear been observed in yeast and bacteria, but the physiological
significance of these oligomers is unknown.7 Redox chemistry
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

Fe/S proteins. This machinery primarily mediates three steps of


Fe–S cluster biosynthesis; namely, (1) cluster assembly on the ensues, although the details are poorly understood, possibly with
scaffold protein, (2) chaperone-assisted release of the Fe–S the assistance of a redox-active ferredoxin.7d,8 Mitochondria
cluster and a currently unclear mechanism of mitochondrial import ferrous iron in a membrane potential-dependent manner
export, if necessary, and (3) subsequent transfer to putative via the mammalian carrier proteins mitoferrin 1 and 2.9 This iron
target apo-proteins.1b,c Much of the research concerning mito- is then transferred to IscU through an iron donor protein called
chondrial Fe–S cluster biosynthesis has been carried out in frataxin (Fxn),10 for complete assembly of a [2Fe–2S] cluster.
yeast cells, human cells, and with human, yeast, or bacterial Frataxin can also function as a regulatory partner in the Nfs1–
Isd11–IscU complex to control not only sulfur production, but
also iron entry during Fe–S cluster assembly, to accelerate the rate
Christine Wachnowsky first studied of Fe–S cluster biogenesis.11,12
biochemistry at Allegheny College
in the lab of Dr Margaret Nelson, 2. Chaperone assisted release of the Fe–S cluster and
focusing on cell differentiation mitochondrial export, if necessary
in Dictyostelium discoideum. Cluster release from IscU to target proteins is mediated by a
She earned her PhD at the Ohio dedicated chaperone system comprising the Hsp70-type pro-
State University as part of the tein HSPA9 (homolog of yeast Ssq1 and bacterial HscA) and the
Ohio State Biochemistry Program. co-chaperone Hsc20 (homolog of yeast Jac1 and bacterial HscB)
Christine conducted research in (Fig. 1 and Table 1).13 The chaperone complex binds and induces
the laboratory of Dr James A. a conformational change in IscU, which enables release of the
Cowan on the topic of iron–sulfur IscU-bound Fe–S cluster either to selected target proteins or to
cluster biosynthesis and trafficking specific Fe–S cluster carrier proteins, with the proposed major
C. Wachnowsky in relation to disease states. target being the monothiol glutaredoxin, Grx5, a homodimeric

Insiya Fidai completed her James A. Cowan graduated with


Bachelor’s and Master’ in Bio- first class honours in Chemistry
technology from the University from the University of Glasgow,
of Mumbai, India. During her before moving to Cambridge
Masters, she worked on biocom- University to pursue research in
patible synthesis of noble metal synthetic organic chemistry and
nanoparticles using various reducing chemical physics. Subsequently
sugars under the guidance of he took up a postdoctoral appoint-
Dr Pankaj Poddar at the National ment at Caltech to pursue studies
Chemical Laboratory, Pune, India. in bioinorganic chemistry before
She then earned her PhD at The taking up a faculty position
Ohio State University as part at The Ohio State University.
I. Fidai of the Ohio State Biophysics J. A. Cowan Dr Cowan’s research interests
Graduate Program. In Dr Cowan’s cover metal catalysis, metals in
laboratory, Insiya worked on ATCUN complexes as potential catalytic medicine, and the cellular chemistry
metallodrugs and role of iron–sulfur clusters in biogenesis, trafficking of iron and metal cofactors.
and storage, and implications for disease states.

10 | Metallomics, 2018, 10, 9--29 This journal is © The Royal Society of Chemistry 2018
View Article Online

Metallomics Minireview
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

Fig. 1 Schematic representation summarizing current models of mammalian Fe–S cluster biogenesis based on disease conditions. Nfs1, the cysteine
desulfurase, with its cofactor protein Isd11 donates sulfur from L-cysteine for nascent Fe–S cluster assembly. Together, they bind to the primary scaffold
protein IscU, the regulator protein frataxin (Fxn), which controls cysteine desulfurase activity and iron delivery, and a source of electrons. The newly
formed IscU-bound [2Fe–2S] is subsequently transferred via a dedicated chaperone-co-chaperone system (Hsc20–HSPA9) to Grx5. From Grx5, the
Fe–S clusters are either directly inserted into mitochondrial [2Fe–2S] proteins or transferred via specific carrier systems such as the one composed of
IscA1, IscA2 and Iba57 to mitochondrial [4Fe–4S] proteins. To date, the presence of another cluster type (such as a glutathione-complexed [2Fe–2S]
cluster) under physiological conditions cannot be excluded. For extra-mitochondrial Fe–S cluster, an uncharacterized compound (X) is exported for
cytosolic and nuclear cluster trafficking via the Fe–S cluster export machinery consisting of ALR, glutathione and ABCB7. Species X has been proposed to
be a glutathione-complexed [2Fe–2S] cluster and is depicted as the [2Fe–2S](GS)4 complex.17 Cytosolic cluster trafficking has been studied primarily in
yeast. Fe–S clusters can be trafficked through Grx3 to anamorsin, which provides electrons to the cytosolic scaffold CFD1–NBP35. From there, [4Fe–4S]
clusters are transferred via IOP1 to the CIA proteins and final target proteins, such as IRP. The CIA proteins can complex with MMS19 for cluster delivery to
nuclear targets, like polymerases, helicases, and glycosylases. The red numbers in the figure correspond to the disease numbers in the review and
in Table 1.

protein, that binds a [2Fe–2S] cluster with two exogenous gluta- mitochondria.18 Hence, glutathione is required for extra-
thione ligands for downstream cluster delivery and trafficking mitochondrial Fe–S cluster biogenesis and therefore impacts
(Fig. 1).14 cellular iron homeostasis.19 Prior reports have provided evidence
From Grx5, the [2Fe–2S] cluster can also be targeted to other for a tetrameric GSH-coordinated [2Fe–2S] cluster as a possible
downstream proteins within the mitochondria, or for export substrate for the exporter protein.17a In fact, mixing GSH-
from the mitochondria to the cytosol via the ABC transporter coordinated [2Fe–2S] complex with purified Atm1 stimulated
ABCB7 (yeast Atm1) (Fig. 1).15 The crystal structures of yeast its ATPase activity up to 1.8-fold.17b Structures were solved with
Atm1 and a related bacterial ABC transporter that confers heavy potential glutathione and oxidized glutathione (GSSG) binding
metal tolerance16 indicate that the transporter exports a sulfur sites about 5 Å apart, which could readily accommodate a
containing molecule from the mitochondria to the cytosol, [2Fe–2S] cluster and provide support for a glutathione com-
which is critical for the CIA machinery.17 A role for glutathione plexed Fe–S cluster.16 In vivo determination of these glutathione
in the ISC export process has been established in yeast cells, complexed [2Fe–2S] clusters can further strengthen the physio-
where a depletion of glutathione results in a phenotype similar logical existence of these small molecules in the cell. Another
to that of Atm1 deficiency that produces iron overload in study has suggested the molecule GSSSG, i.e., the oxidized form

This journal is © The Royal Society of Chemistry 2018 Metallomics, 2018, 10, 9--29 | 11
View Article Online

Minireview Metallomics

Table 1 Iron–sulfur cluster proteins in relation to disease states. The number in the first column corresponds with the number in the review and Fig. 1.
All of the proteins listed are shown in the general schematic of cluster biosynthesis, except Grx2 and FECH

Number Protein Function Disease Most common cause


1 Frataxin Iron donor; allosteric Friedreich’s ataxia (FRDA) GAA-triplet expansion causing transcriptional
effector of NFS1 silencing
2 IscU Fe–S cluster scaffold IscU myopathy Splice mutation in intron 4 resulting in decreased
protein levels
3 Fdx2 Electron transport Fdx2 myopathy c. 1A 4 T mutation disrupting the initiation codon
4 Nfs1 Sulfur donor for cluster Infantile mitochondrial p. R72Q decreasing transcripts and protein levels
assembly complex II/III
4 ISD11 Stability factor Combined oxidative p. R68L resulting in undetectable ISD11 protein levels
phosphorylation deficiency 19
(COXPD19)
5 Nfu Late acting [Fe–S] MMDS1 p. G208C resulting in insufficient downstream holo targets
targeting factor
5 BOLA3 Late acting [Fe–S] MMDS2 Truncation mutation resulting in insufficient downstream
targeting factor holo targets
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

5 Iba57 [4Fe–4S] cluster MMDS3 p. Q314P causing protein misfolding and degradation
assembly factor
5 IscA1/2 [4Fe–4S] cluster assembly MMDS4 p. G77S resulting in insufficient downstream holo targets
6 Ind1 Assembly factor Complex I deficiency p. G56R and a c. 815-27T 4 C branch site mutation
for complex I leading to aberrant splicing
6 SDHB Respiratory complex II Neurodegeneration and Mutations at Fe–S cluster ligating cysteines preventing
cancer cluster binding
7 SDHAF1 Assembly factor leukoencephalopathy Mutations that disrupt binding interface for either
for complex II Hsc20 or SDHB prevent cluster maturation on SDHB
8 MitoNEET Maintenance of Association with cystic Related to decrease in mRNA levels
iron homeostasis fibrosis, Parkinson’s disease,
and cancer
9 MUTYH Base excision repair Colorectal polyposis p. P281L resulting in low protein that cannot bind DNA
of 8-oxoguanine: adenine
mismatches
10 FANCJ Genome maintenance Fanconi anemia and breast p. M299I upregulates helicase activity or p. A349P that
and double-strand cancer impairs DNA–protein interactions
break repair
10 XPD Nucleotide excision repair
Xeroderma pigmentosum (XP), p. R112H that impairs transcriptional function and
and transcription Cockayne’s syndrome (CS), inactivates helicase activity
trichothiodystrophy (TTD),
and Cerebro-oculo-facial-skeletal
(COFS) syndrome
10 ChlR1 Genome maintenance Warsaw breakage syndrome p. R263Q inhibits DNA binding and DNA-dependent
hydrolysis
10 RTEL1 Maintenance of telomeres Cancer, dyskeratosis Protein mutations that prevent telomere maintenance
congenita (DC),
and Hoyeraal-Hreidarsson
syndrome (HHS)
11 POL d DNA polymerases Cancer Unclear connection
and e
12 ABCB7 Fe–S cluster export from X-linked sideroblastic Protein mutations that result in mitochondrial iron overload
the mitochondria anemia (XLSA)
13 Grx5 [2Fe–2S] cluster carrier Sideroblastic-like anemia Aberrant splicing leading to decreased Fe–S cluster transfer
and impaired iron homeostasis
14 IRP Iron homeostasis Parkinson’s disease Erroneous regulation generating reactive oxygen species
14 Grx2 Cluster carrier and redox Parkinson’s disease Erroneous regulation of GSH levels
regulation
15 FECH Heme biosynthesis Erythropoietic protoporphyria Over 130 different mutations that impair function

of GSSH (glutathione persulfide) and GSH, as the Atm1 sub- [4Fe–4S] cluster.1b,c A current model suggests that [4Fe–4S]
strate and potential sulfur carrier.20 Nevertheless, an as yet cluster maturation requires additional ISC proteins: IscA1 and
ill-defined species X must be exported from the mitochondria IscA2, and the folate binding protein Iba57,21 in a process that
in order to support proper Fe–S cluster trafficking in other requires sequential delivery of a [2Fe–2S], postulated to be from
cellular compartments. Grx5, with reductive coupling to form a [4Fe–4S] cluster (Fig. 1).22
However, knockdown of these IscA proteins does not eliminate
3. Transfer to putative target apo-proteins production of [4Fe–4S] clusters, so other proteins and complexes
Recent studies have shown that a number of other proteins are may utilize a similar reductive coupling mechanism on target
further involved in Fe–S cluster biogenesis. Following release Fe–S cluster proteins.21b Other more specialized targeting factors
of the Fe–S cluster from the scaffold protein, it can either be have evolved to assist the maturation of dedicated subsets of
directly delivered to [2Fe–2S] target proteins or converted to a [4Fe–4S] proteins and also for transfer of [2Fe–2S] clusters

12 | Metallomics, 2018, 10, 9--29 This journal is © The Royal Society of Chemistry 2018
View Article Online

Metallomics Minireview

downstream of the scaffold assembly to final target proteins, protein Nar1 and the CIA targeting complex Cia1–Cia2–Mms19
such as Nfu, BolA3, Ind1, and SDHAF (Fig. 1 and Table 1).1a (Fig. 1).1a,4c Nar1 coordinates two [4Fe–4S] cofactors that are
However, the precise mapping of these pathways and all of the similar to those in hydrogenases and bridges the early and late
potential functions of these proteins remain unclear. For example, stage CIA machinery.61,62 Cia1, Cia2 and Mms19 form dimeric
Nfu may act as an alternative scaffold protein to IscU, because Nfu and tetrameric complexes in both yeast and mammalian cells,
can accommodate both a [2Fe–2S] cluster as well as a labile and interact with target apo-proteins and assure specific Fe–S
[4Fe–4S] cluster in vitro and pass these on to apo-proteins.23 cluster insertion,28 including the crucial function of maturation
of nuclear Fe–S proteins (Fig. 1). Several of the proteins
involved in mitochondrial Fe–S cluster biogenesis have also
Cytosolic and nuclear Fe–S cluster been reported to be localized in the cytosol such as IscU, Nfu,
protein biogenesis Nfs1 and Isd11.29 However, their functional relevance is yet to
be established.
The biogenesis of cytosolic and nuclear Fe–S proteins is heavily
dependent on the conserved mitochondrial Fe–S cluster (ISC)
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

assembly machinery in addition to the ABC transporter protein Diseases associated with Fe–S cluster
and cytosolic Fe–S cluster (CIA) assembly machinery. To date,
nine components of the CIA machinery have been identified
biogenesis proteins
that play a role in mobilization of Fe–S cluster and its insertion A number of inborn human diseases have been identified with
into extra-mitochondrial apo-proteins. The assembly process mutations associated either with the gene or the protein involved
has been primarily studied in yeast and comprises two mains in Fe–S cluster biogenesis. In recent years, additional diseases
steps:1a,4c (1) [4Fe–4S] cluster assembly on the Cfd1–Nbp35 have been identified with the increasing availability of informa-
scaffold complex and (2) transfer of Fe–S clusters to target apo- tion concerning the function and properties of Fe–S cluster
proteins from the CIA scaffold complex.1b,c Yeast nomenclature proteins.1b,c Several of these disease states are discussed below
has been used throughout this section and describes the proposed (Table 1). Additional disease states related to Fe–S cluster
models based on the work in yeast. Similar pathways are likely maturation have been discussed in terms of infectious agents,
functional in humans too. but those will only be described briefly in this work.

1. [4Fe–4S] cluster assembly on the CFD1–NBP35 scaffold Diseases associated with mitochondrial Fe S cluster
complex biogenesis
An initial step in cytosolic Fe–S protein biogenesis is the assem- 1. Friedreich’s ataxia (FRDA). Friedreich’s ataxia (FRDA) is
bly and binding of a transient [4Fe–4S] cluster on the cytosolic a neurodegenerative disease that is by far the most common
scaffold protein complex, a heterotetramer comprised of the two disease linked to Fe–S cluster biosynthesis.30 FRDA patients carry
P-loop NTPases Cfd1 and Nbp35, which bind two different kinds a GAA-triplet expansion mutation in the first intron of the frataxin
of [4Fe–4S] clusters (Fig. 1).4c,24 A loosely bound [4Fe–4S] cluster gene (Fxn). This expansion leads to transcriptional silencing of
at the C-terminus of both proteins bridges the two complex frataxin thereby resulting in severely decreased levels of the
subunits together, whereas the other [4Fe–4S] cluster is tightly frataxin protein. Nonsense and missense mutations or deletions
coordinated at the N-terminal domain of the Nbp35 protein. The also result in the same symptoms for patients.30c,31 No therapy for
lability of the C-terminus cluster is considered to be important FRDA exists today, despite much effort to increase frataxin levels
for the second step, involving transfer of the cluster to target apo- or mitochondrial function, or alleviating secondary symptoms by
proteins. Generation of the functionally essential N-terminal antioxidants or iron chelator strategies.32
Fe–S cluster (N) of Nbp35 depends on the flavoprotein Tah18 2. IscU myopathy. IscU myopathy is characterized by severe
(yeast homolog of NDOR1) and the Fe–S protein Dre2 (yeast exercise intolerance, tachycardia, fatigue and pain in active
homolog of CIAPIN1), which serve as an NADPH-dependent muscles.1a,2b Patients have decreased mitochondrial Fe–S cluster
electron (e ) transfer chain (Fig. 1).25 Cytosolic–nuclear Fe–S enzyme activities, as well as iron deposits.33 Most affected patients
protein biogenesis additionally requires the cytosolic multidomain are homozygous for a splice mutation in intron 4 of the IscU gene
glutaredoxin, Grx3, yet its precise function and site of action in the resulting in decreased IscU protein levels.33,34 A therapeutic
pathway remains to be determined,26 although studies from the approach consisting of antisense oligonucleotides, in order to
yeast system suggest roles in iron regulation and maturation of induce skipping of the splice site responsible for the condition
other proteins such as ribonucleotide reductase.26b,27 Accordingly, and restore normal mRNA splicing in fibroblasts, is currently
it is speculated that these cytosolic monothiol glutaredoxins play a being tested,35 as are treatment options that control miRNA levels
general role in iron trafficking. to deal with the vascular symptoms and conditions.36 IscU has
also recently been found as a target for regulation by the tumor
2. Transfer of Fe–S clusters to target apo-proteins from the suppressor protein p53 with implications for modulating iron
CIA scaffold complex homeostasis and iron levels in cancer.37
The bridging Fe–S clusters are released from Cfd1–Nbp35 and 3. Fdx2 myopathy. In addition to IscU myopathy, a second
transferred to apo-proteins in a reaction mediated by the Fe–S novel mitochondrial myopathy has been found in a patient due

This journal is © The Royal Society of Chemistry 2018 Metallomics, 2018, 10, 9--29 | 13
View Article Online

Minireview Metallomics

to a disruption on the FDX1L gene, which encodes the mito- close to a splice site and results in skipping of exon 6 and no
chondrial protein ferredoxin 2 (Fdx2).38 The myopathy is due to detectable protein levels.46 Most other patients were described
a mutation (c. 1A 4 T) that disrupts the ATG initiation codon, with a homozygous mutation (p. Gly208Cys) that affects an
resulting in low levels of Fdx2 in muscle and fibroblasts, and amino acid close to a highly conserved Fe–S cluster binding
decreased activities of respiratory complexes I–III and mito- domain of the Nfu protein. This mutation does not affect protein
chondrial aconitase.38 Symptoms resemble those of IscU myo- levels but does impair Fe–S cluster binding and transfer, most
pathy but are not as severe, possibly indicating a functional likely due to a perturbed monomer–dimer equilibrium that
rescue by the homolog Fdx1 or a bypass mechanism compared limits cellular reconstitution of Nfu.41f,47 Most recently, a study
to the role of IscU in Fe–S biogenesis.38 of patients with the heterozygous gene for Nfu, one with the
4. ISD11–Nfs1. Recently, the importance of ISD11 (human altered splice site or with the Gly208Cys missense mutation and
LYRM4) and Nfs1 in cluster biogenesis as part of the de novo the native allele, has revealed that low levels of the wild type Nfu
cluster assembly complex was highlighted as a result of their mRNA were produced, which yielded sufficient functional LIAS
roles in disease states. For Nfs1, patients were found with a to avoid lipoic acid deficiency, but the wild type transcripts were
missense mutation (p. Arg72Glu) that results in infantile mito- insufficient to support functional respiratory complex II.41d
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

chondrial complex II/III deficiency.39 Levels of Nfs1 transcripts and In the case of MMDS2, 6 different patients have been identi-
protein levels were decreased in patient fibroblasts, as were the fied with mutations on BOLA3.41b,42 The first patient demon-
levels of respiratory complexes II and III, which could be strated a single base pair duplication that produced a frameshift
indicative of an overall disruption in Fe–S cluster biogenesis with a premature stop codon and therefore loss of the full length
and is also consistent with the absence of ISD11 association protein.41b Two other patients have been characterized as
in co-immunoprecipitation assays.39 However, treatment with having a missense mutation at a highly conserved residue
mitochondrial cofactor therapy was successful for one of the (p. Ile67Asn)42b that could affect the overall protein fold based
three patients in preventing lethality during infancy, suggesting on the solution NMR structure of human BOLA3.48 The remain-
a potential therapeutic option.39 ing three patients all demonstrated a truncating mutation of
The requirement for ISD11 in stabilizing Nfs1 has been eight amino acids upstream of the first truncating mutation.42a
confirmed in patients with mutations on ISD11, who demon- To date, the molecular role of BOLA3 remains to be elucidated;
strated a similar phenotype in the form of respiratory complex however the availability of an NMR structure48,49 with additional
deficiency, termed combined oxidative phosphorylation deficiency biochemical characterization will provide increasing understanding
19 (COXPD19).40 Interestingly, these patients also exhibited further of its role in MMDS.
downstream complications with deficiencies in complexes I, Similar to BOLA3 in MMDS2, a wide variety of mutations on
II and III, in addition to other mitochondrial and cytosolic Fe–S Iba57 have been identified for MMDS3. Initial reports described
proteins.40a Both patients were homozygous for a missense muta- different patients with a range of phenotypes. The first case
tion (p. Arg68Leu),40a which caused the levels of ISD11 to be resulted in the most severe condition caused by a missense
undetectable and a decrease in respiratory chain complexes to mutation (p. Gln314Pro) that appeared to result in protein
below 40%. This mutation also impacted the interaction with Nfs1 misfolding with subsequent degradation to concentrations
by promoting aggregation and inhibiting desulfurase activity.40a below critical levels.43a The mildest phenotype was observed
Furthermore, IscU levels were also observed to decrease when the in a patient with an Iba57 mutation that resulted in reduced
R68L mutation was present, suggesting that the complex is also mRNA levels because of aberrant splicing and a frameshift
essential for promoting the stability of IscU for overall Fe–S cluster mutation that yielded a truncated form of Iba57.43c The inter-
biogenesis and trafficking.40b mediate phenotype of MMDS3 was due to a mutation on Iba57
5. Multiple mitochondrial dysfunction syndrome (MMDS). that drastically reduced its ability to function in Fe–S biogenesis
Multiple mitochondrial dysfunction syndrome (MMDS) describes a with diminished levels of [4Fe–4S] clusters.43b Most recently,
class of fatal mitochondrial diseases caused by mutations in four additional mutations on Iba57 have been observed with
proteins involved in Fe–S cluster biogenesis and trafficking. To date, generally the same symptoms and phenotypes.43d The prevalence
four types of MMDS have been identified: namely, MMDS1 due to of such diverse mutations on Iba57 that all result in MMDS3
mutations on Nfu,41 MMDS2 for mutations with BOLA3,41b,42 makes prediction of the disease difficult, based on genetic
MMDS3 with mutations on Iba57,43 and MMDS4, with mutations information, and strengthens the need for further biochemical
on IscA2.44 Recently, an IscA1 variant has been described that characterization of Iba57 to understand why so many possible
results in MMDS-like symptoms, which may expand the class of causes of the disease exist.
disorders.45 All MMDS patients exhibit similar symptoms that MMDS4 was the most recent class to be discovered, with six
include metabolic acidosis with hyperglycemia, and deficiency of patients exhibiting a missense mutation (p. Gly77Ser) on IscA2,
respiratory complexes and lipoic acid bound enzymes, such as which is located in a highly conserved cluster binding domain.
pyruvate dehydrogenase (PDH) and a-ketogluterate dehydro- However, the exact effect of the mutation is unclear, since
genase (KGDH).41f,46 Lipoate biogenesis relies on the mitochondrial mRNA levels were not significantly impacted.44 Interestingly,
[4Fe–4S] enzyme lipoate synthase (LIAS). patient fibroblasts also showed lower levels of IscA1 and Iba57
For MMDS1, five patients have been described with a homo- than control fibroblasts.44 How this mutation results in lower
zygous mutation in Nfu (p. Arg182Gln), where the mutation is protein levels of these partner proteins remains unclear.

14 | Metallomics, 2018, 10, 9--29 This journal is © The Royal Society of Chemistry 2018
View Article Online

Metallomics Minireview

Similarly, an IscA1 mutation (p. Glu87Lys) has been implicated conserved residue (p. Asp48Val) near an iron–sulfur cluster
in causing MMDS-like symptoms, which may result in a new binding site, which is hypothesized to be crucial for efficient
class of the disorder, as one patient has been found with this electron transport through the respiratory chain and resulted
mutation that is expected to lead to protein instability due to in an almost complete loss of SDHB and decreased levels of
loss of a conserved salt-bridge.45 SDHA.56 As with many mitochondrial disorders, there is no
The class of MMDS disorders describes a newly discovered cure for SDH deficiency; however, the patient in this study was
disease condition and further details on how mutations in key supplied with an oral treatment of coenzyme Q10 to yield a
Fe–S biogenesis proteins influence cellular function, and their subjective improvement in strength.56
roles that result in disease, need to be determined in order to The remainder of patients with mutations on SDHB have
fully aid patients. been linked to some form of cancer. A recent analysis of the
6. Respiratory complex 1. Fe–S clusters play essential roles in Leiden Open Variation Database showed that 37% of all the
respiration and metabolism as cofactors in respiratory chain com- reported cancer cases linked to SDHB are due to mutations to
plexes. The manner in which Fe–S clusters are integrated into the the cluster cysteinyl ligands that prevented cluster binding,
respiratory complexes is currently unclear; however, respiratory or in the L(I)YR motif, which blocks chaperone recognition by
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

chain complex maturation factor proteins have recently been Hsc20 and the HSPA9 chaperone to prevent binding interactions
identified that are required for correct formation of the complexes, with either of these proteins.58
either by promoting subunit association or cofactor integration.50 As is the case for respiratory complex I, maturation factors for
In particular, complex I of the respiratory complex, also SDH have also been implicated in causing disease. In the case of
known as NADH-CoQ oxidoreductase, is composed of 44 sub- SDHB, two assembly factors exist: SDHAF1 and SDHAF3.55b,59 Both
units, utilizes at least 10 assembly factors for correct complex factors are believed to help protect SDHB from oxidative damage
formation, and accounts for one-third to one-quarter of mito- during the assembly of the complete SDH complex by shielding
chondrial disease, which occurs in about 1 out of every 5000 the nascent SDHB Fe–S clusters from damage by reactive oxygen
births.51 One such assembly factor, Ind1 or NUBPL, has been species.55b,59 SDHAF1 also functions in concert with Fe–S cluster
implicated as an Fe–S cluster delivery protein that is specifically chaperones in recruiting the chaperone complex to SDHB via the
required for complex I assembly and activity in yeast,51 and L(I)YR motif,60 and was found to co-immunoprecipitate with
mutations or perturbations of the Ind1 gene have resulted in a Hsc20.58b As such, mutations on SDHAF1 that disrupt the
specific deficiency of complex I, while other respiratory com- binding interface for either SDHB or Hsc2060 have also been
plexes remain intact.52 Genetic sequencing has found two muta- implicated in causing leukoencephalopathy,61 where accumu-
tions in patients: namely, a p. Gly56Arg missense mutation and lation of succinate in the white matter of the brain is caused by
a c. 815-27T 4 C branch site mutation. Of the two, pathogenicity a loss of SDH activity. The accumulation of succinate was due
has been more attributed to the branch site mutation, since it to degradation of SDHB, when non-functional SDHAF1 was
results in aberrant splicing, very low levels of the NUBPL protein, present; however, succinate levels could be reduced following
and reduced levels of fully assembled complex I, as little as treatment with riboflavin, which serves to increase the flavinyl-
20%.52,53 However, there remains the possibility that the mis- ation of SDHA and allows it to convert succinate to fumarate to
sense mutation contributes to the pathogenicity of the branch restore the activity of the Krebs cycle.60
site mutation. Furthermore, recent work in Arabidopsis thaliana 8. MitoNEET. Outside of the traditional mitochondrial
suggests that Ind1 may also play a role in translational control,54 Fe–S cluster components and targets, the outer mitochondrial
which may contribute to phenotypes observed in the pathogenic membrane protein, mitoNEET has been implicated in numerous
state. However, no work has been done in human cell lines to diseases.62 MitoNEET was first discovered bound to thiazolidine-
either evaluate this additional functional role or examine the dione, a drug used for the treatment of type 2 diabetes. Since
possible role of Ind1 in Fe–S cluster delivery. then, its functional role has been under active investigation.
7. Respiratory complex II. Respiratory complex II (or Given its association with a diabetes drug, one of the physio-
succinate dehydrogenase, SDH) is composed of four subunits logical roles of mitoNEET is linked to diabetes, via control of
(A–D). Subunits C and D anchor the complex to the membrane, lipid and glucose metabolism,63 most likely by modulation of
while subunits A and B exhibit catalytic activity.55 All of the lipid accumulation,64 which is related to insulin resistance.
subunits have been implicated in disease states, because SDH MitoNEET is also involved in controlling overall mitochondrial
plays vital cellular roles as a result of its involvement in both bioenergetics by regulating homeostasis and the ratio of energy
the Krebs cycle and oxidative phosphorylation. However, only stored to the energy spent,64 which connects with its role in
subunit B (SDHB) contains iron–sulfur clusters.55a SDHB is the causing obesity and potential regulation of mitophagy.63 In
most commonly mutated subunit of the four,55a and mutations conjunction with mitochondrial homeostasis, overexpression of
result primarily in either neurodegeneration, generally in the mitoNEET results in diminished levels of mitochondrial iron,
form of infantile leukodystrophy56 or cancer, particularly renal while knockdown leads to iron overload and reduced activity of
cell carcinoma, paraganglioma, pheochromocytoma, or WT the electron transport chain, resulting in overall lower cellular
gastrointestinal stromal tumors.55,57 energy.64 Accordingly, mitoNEET plays a key role in cellular and
In terms of the neurological impact, one patient has been whole organism iron regulation and metabolism, and in main-
identified with a missense mutation at an evolutionarily tenance of reactive oxygen species,62,65 potentially by binding a

This journal is © The Royal Society of Chemistry 2018 Metallomics, 2018, 10, 9--29 | 15
View Article Online

Minireview Metallomics

[2Fe–2S] cluster for delivery and repair of cytosolic aconitase 9. Glycosylases. The glycosylase family provided the first
(IRP1),66 in a process that is under redox control.67 protein example to be characterized with an Fe–S cluster by way
Given its broad role in metabolism, mitoNEET has also been of endonuclease III in E. coli;2a however, mammalian forms are
associated with cystic fibrosis due to a decrease in mitoNEET now being characterized. Members of this family conduct base
mRNA,68 Parkinson’s disease due to an interaction with excision repair (BER) to detect, remove, and replace damaged
PARKIN,69 and cancer and tumorogenesis,62,70 where mitoNEET DNA bases. In particular, the human protein MUTYH serves to
was found to be upregulated in breast cancer cells.71 Despite the repair mismatches associated with 8-oxoguanine.79 A number
association of mitoNEET with broad cellular processes and of disease variants of MUTYH have been identified that result
diseases, many questions remain unanswered concerning its in cancer in the form of colorectal polyposis, termed MUTYH-
specific cellular mechanism and the connection between its role associated polyposis (MAP).80 To date, one of the mutations
in Fe–S cluster biosynthesis and overall mitochondrial regula- (P281L) was found at a highly conserved proline in the Fe–S
tion. Furthermore, mitoNEET is still under consideration as a cluster binding domain, which impairs the ability to bind
target for drug development for the treatment of cancer and substrate DNA.80–82 A detailed understanding as to why variants,
diabetes based on overall mitochondrial dysfunction,62,63 with such as these, result in MAP will provide assistance to clinicians
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

additional possibilities considering its likely roles in Parkinson’s in patient-screening for cancer risk and subsequent treatment
and cystic fibrosis. As more is learned concerning mitoNEET and options.82
its physiological roles, better therapeutics may be developed to 10. Helicases. Helicases constitute a second family of
compensate for disruptions in its function. nuclear proteins that contain Fe–S clusters and function in
DNA processing. Within the helicase family, the Fe–S cluster
Diseases associated with cytosolic Fe–S cluster proteins subset belongs to the SF2 grouping with shared homology of the
Although a number of proteins have been identified as compo- helicase domain.2a,83 A number of these proteins have disease
nents of the cytosolic Fe–S cluster machinery,4b,15,72 many have implications and are therefore discussed below.
only been discovered or characterized in depth over the last FANCJ, in conjunction with 15 other genes, is implicated in
5 years. For this reason, many of these proteins have not yet been causing Fanconi anemia; a condition characterized by congenital
linked to disease conditions. However, given their required roles defects, progressive bone marrow failure, susceptibility to cancer
in the biogenesis of essential Fe–S clusters and maintenance of with chromosome breaks, and sensitivity to DNA damaging agents
iron homeostasis, there is the possibility of disease association that result in intra-strand cross-links.84 Furthermore, FANCJ plays
as these proteins become better understood and disease states a role in genome stability by unwinding G-quadruplexes85 and by
are examined in greater depth. For example, examination of conducting double-strand break repair.86 Of the numerous muta-
proteins in the NAR family in Arabidopsis thaliana73 and yeast74 tions identified in the gene for FANCJ that are linked to various
have indicated conserved roles in maintaining a response to disease states, two of them (M299I and A349P) occur in the
oxidative stress, which may have downstream implications iron–sulfur cluster binding domain, where the M299I mutation
for genome maintenance,72 due to the connection to DNA has been found in breast cancer patients,83a,87,88 and the A349P
damage repair pathways and corresponding involvement in mutation causes Fanconi anemia.89 These mutational analyses
aging disorders. have demonstrated the importance of the Fe–S cluster in the
Likewise, the cytosolic Fe–S protein MMS19, has not speci- function of FANCJ and its role in downstream DNA processing
fically been associated with any diseases. However, this protein activities in disease.83
has been characterized as required for maturation of many A second Fe–S cluster-containing helicase is XPD, which
nuclear Fe–S cluster containing proteins that are particularly serves key roles in nucleotide excision repair to remove photo-
involved in DNA replication and repair, such as FANCJ, XPD, damaged DNA bases, and in transcription as a member of
RTEL, and POLD (discussed below).75 The essential nature of the TFIIH complex.83 Mutations in XPD have been linked to
MMS19 in maturation of key nuclear proteins may link it to xeroderma pigmentosum (XP), Cockayne’s syndrome (CS), tri-
cancer and neurodegenerative phenotypes in the future.75b chothiodystrophy (TTD), and Cerebro-oculo-facial-skeletal
(COFS) syndrome.83 To date, only one of these disorders has
Diseases associated with nuclear Fe–S cluster containing been linked to a mutation in the Fe–S cluster binding domain.
proteins In patients with TTD, a R112H mutation is present at a highly
With oxygen sensitivity and possible toxicity associated with conserved residue that removes a hydrogen bond to a cluster-
Fe–S clusters, it was assumed that most nucleic acid enzymes ligating cysteine,90 resulting in impaired interactions with
did not contain Fe–S clusters. However, this view changed with proteins in the TFIIH complex and involvement in helicase
the discovery of an Fe–S cluster in the DNA helicase XPD and inactivation.83a,90a
associated protein members that are involved in DNA repair,76 The last two Fe–S cluster helicases that we will describe have
and the possibility that Fe–S cluster containing enzymes find been implicated in disease much more recently. The helicase
DNA damage by probing DNA integrity electronically.77 Today, ChlR1, or DDX11, is less understood than those already discussed.
all DNA polymerases involved in replication and also certain Its specific role in DNA processing and genome integrity is
helicases that participate in Okazaki fragment processing con- unclear; however, it may serve primarily to unwind intermediate
tain some form of Fe–S clusters.2a,78 DNA structures present during replication and recombination,91

16 | Metallomics, 2018, 10, 9--29 This journal is © The Royal Society of Chemistry 2018
View Article Online

Metallomics Minireview

with additional potential roles in sister chromatid cohesion, disease symptoms, new phenotypes are being associated with
heterochromatin organization, viral genome maintenance, ABCB7 mutations that give rise to cerebellar ataxia102 and ringed
DNA repair, and G-quadruplex unwinding, via its helicase and sideroblasts.103
ATPase activities.92 Although its precise function is unknown, 13. Sideroblastic-like anemia and iron overload. A homo-
the first human patient with a defect in ChlR1 was identified in zygous mutation in the exon 1 of the GRX5 gene causes problems
2010 in a condition termed Warsaw breakage syndrome, due to with RNA splicing, resulting in sideroblastic-like anemia and
either a splice site mutation93 or p. Arg263Gln mutation in iron overload condition.104 This condition causes moderate
ChlR1, which perturbs a conserved arginine residue in the Fe–S anemia, hepatosplenomegaly, and iron overload, and leads to
cluster binding domain.94 a decreased level of mRNA for Grx5 but not a complete loss of the
The last Fe–S cluster helicase that we will discuss is the Grx5 protein function.14b,104a The decreased protein levels result
regulator of telomere length 1 (RTEL1), which functions to in impaired cluster transfer to downstream Fe–S proteins, such as
maintain telomere integrity and unwinds various DNA struc- IRP1 and ferrochelatase (FECH).104c An additional sideroblastic
tures that are assembled in the process of replication, recom- anemia patient has been found with a mutation in GRX5,
bination, and repair.95 Thus far, a number of RTEL1 variants which is a compound heterozygous mutation p. Lys101Gln
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

have been found that exhibit single nucleotide polymorphisms and p. Leu148Ser.105 In vitro analysis of these mutations in
(SNPs) and increased susceptibility for brain tumors or hepato- Grx5 revealed that the L148S mutation results in similar effects
cellular carcinomas, suggesting a key role for the protein in as observed in cases of patients with diminished protein levels,
tumorigenesis.95 Mutations of RTEL1 in coding regions have where cluster transfer to downstream targets was impaired.106
instead been implicated in premature aging disorders,95 such The K101Q mutation yielded different effects, in that this
as dyskeratosis congenita (DC)96 and Hoyeraal-Hreidarsson variant was unable to bind a Fe–S cluster,106 and correlates
syndrome (HHS),97 which is considered a more severe form of DC. with the fact that this residue participates in GSH binding,107
Both diseases are characterized by multi-system bone marrow which is required as a cluster ligand. Aside from implications in
failure, due to impaired telomere maintenance.95 One mutation sideroblastic anemia, patients have also been found to display
(p. Glu275Lys) has been found in the Fe–S cluster domain,96b variant non-ketotic hyperglycinemia, with symptoms similar to
but the biochemical characterization and impact of this residue those observed for patients with MMDS2 due to mutations
on the cluster has not yet been examined. in BOLA3.42a In these three patients, Grx5 was found with a
Further biochemical characterization is required to under- p. Lys51del mutation that impaired transfer to downstream
stand the Fe–S cluster helicase family, particularly to determine targets and resulted in low levels of lipoate synthase activity,42a
the role of the cluster in relation to disease phenotypes. Efforts indicating that this mutation has functional consequences
in examining the molecular mechanisms will aid current similar to the mRNA splicing defect and L148S variants.106
potential therapies that use chemical rescue by small molecules Although Grx5 appears to function primarily in the early stage
to restore function to mutated helicases and treat disease of Fe–S cluster trafficking,14a,b,104a,108 additional multifunctional
conditions.2a cellular roles may exist given the variety of phenotypes observed
11. Polymerases. The last class of Fe–S cluster containing in disease conditions related to Grx5.42a,104c–106
DNA processing enzymes that we will briefly discuss are poly- Most recently, a phenotype similar to that observed for sidero-
merases. All replicative DNA polymerases, such as delta (d) and blastic anemia has been linked to mutations in the mitochon-
epsilon (e), contain essential [4Fe–4S] clusters2a,98 that are drial Fe–S cluster biosynthesis chaperone, HSPA9, which has
matured via the CIA machinery.99 These two polymerases have also been implicated in key roles in erythroid differentiation.109
implications in cancer;100 however, the precise functional role Mutations to HSPA9 result in lower mRNA expression levels with
of the Fe–S cluster is unknown,2a and therefore its link to incomplete loss of function leading to the anemia,110 a pheno-
disease is unclear. type which may be under a process of complex regulation.

Conditions resulting in mitochondrial iron overload Fe–S cluster proteins linked to neurological disorders
Although the disorders below are linked primarily to the Similar to the diseases related to iron overload, neurological
misregulation of iron, they occur specifically in Fe–S cluster disorders are often related to iron misregulation; however, it
proteins. Whether this link is due to a simple abundance of remains to be seen if there is a more direct connection between
iron or to a more direct connection between Fe–S clusters and Fe–S clusters and these conditions. Recent work has indicated a
iron regulation, perhaps via the iron regulatory protein (IRP), link between mitochondrial dysfunction and neurodegenerative
remains uncertain and requires further research. disorders, such as Parkinson’s disease, Alzheimer’s disease, and
12. X-linked sideroblastic anemia (XLSA). X-linked side- Huntington’s disease.111 These neurodegenerative conditions are
roblastic anemia is caused by mutations in the mitochondrial thought to exist as a feedback loop, where mitochondrial dys-
transporter protein ABCB7.101 The condition is characterized by function causes increased production of reactive oxygen species,
early-onset ataxia, sideroblastic anemia and iron overload in which decreases Fe–S cluster and heme biosynthesis and leads to
affected tissues, primarily in the mitochondria, which suggests IRP activation and iron overload in mitochondria.111,112 This
that the transporter is unable to function properly for trafficking accumulated iron in turn generates additional reactive oxygen
of clusters in the cytosol and nucleus. Expanding on the previous species via Fenton chemistry. In the case of Alzheimer’s disease,

This journal is © The Royal Society of Chemistry 2018 Metallomics, 2018, 10, 9--29 | 17
View Article Online

Minireview Metallomics

these factors promote oligomerization of amyloid-b to form combinations have been observed, where patients can simply
plaques113 and the formation of Fe3+-paired helical filament exhibit multiple loss of function mutations.124,127 As of 2015, at
aggregates114 to further drive iron accumulation and decrease the least 130 different mutations on FECH have been implicated in
levels of the soluble Tau protein.115 Similarly, in Huntington’s EPP, with new mutations or sites being identified, as sequenc-
disease the Huntington protein (Htt) is believed to play a role in ing is extended and population studies are undertaken.127,128
maintaining iron homeostasis via the transferrin receptor;111b,116 Therefore, specific genotype–phenotype correlations and under-
when mutated in the disease state, Htt likely perturbs iron uptake standing of the specific molecular mechanism are challenging.
and contributes to mitochondrial dysfunction.111b In the class of Furthermore, patients with EPP were also found to display
the neurodegenerative disorders, Parkinson’s disease has most abnormal mRNA levels of mitoferrin-1,129 which has been found
been associated with iron–sulfur cluster proteins. to complex with FECH and ABCB10 for heme biosynthesis130
14. Parkinson’s disease. Parkinson’s disease is a neuro- and may also contribute to the phenotype of EPP. As familial
degenerative disease affecting roughly 1 : 800 individuals with a screening becomes more common, doctors can be more aware of
survival rate of about 15 years after diagnosis. The degenerative carriers to provide better treatment plans for patients,128 which
process affects dopaminergic neurons leading to motor func- typically involve avoiding sunlight and tools to reduce excess
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

tion deficits, such as resting tremor, rigidity, and postural protoporphyrin for the less severe cases, and solutions, such as
instability.111,112 Age is considered to be the main risk factor for bone marrow transplants for long term solutions in the case of
Parkinson’s-however, environmental toxins or genetic suscepti- drastic phenotypes.124
bility are also implicated.117 Data suggests that patients with
Parkinson’s also have a defect in ISC synthesis, partial inhibition Fe–S cluster protein maturation in human infectious agents
of complex I of the respiratory chain, and increased iron levels in Iron–sulfur clusters also play a vital role in prokaryotic meta-
dopaminergic neurons (in the substantia nigra),112,118 again due bolism, as catalysts, redox sensors or electron trafficking agents
to erroneous regulation of iron homeostasis by IRP.111,112 In in a variety of important proteins. For example, across different
addition to this, the pathway which delivers transferrin-bound strains of E. coli there are over 150 proteins that contain Fe–S
iron to mitochondria and to complex I of the respiratory chain is clusters and over 50 different proteins in Mycobacterium tuber-
found in the substantia nigra dopaminergic neurons, but also culosis.131 Fe–S clusters have consequently been exploited for
consists of transferrin receptor 2.119 In Parkinson’s, there is bactericidal toxicity, where antibiotics promote cell respiration,
accumulation of oxidized transferrin inside mitochondria, which forming endogenous superoxide and peroxide species, which
results in the release of ferrous iron from transferrin and the degrade Fe–S clusters that serve important roles in bacterial cell
generation of hydroxyl radicals via Fenton chemistry.119a,120 cycles, and DNA and protein biogenesis.132 Fe–S clusters have
Furthermore, glutaredoxin 2 (Grx2) has been implicated in pro- also been targeted in terms of human pathogens in Pseudomonas
moting the iron accumulation feedback loop in Parkinson’s aeruginosa. Deletion of the transcriptional regulator iscR leads
disease based on its ability to regulate cellular redox status by to increased susceptibility to peroxides and severely decreased
acting on its substrate glutathione (GSH) to impact overall iron virulence in mouse and Drosophila models.133 Much work
homeostasis.112,121 As our understanding of these neurodegene- has been done in studying Fe–S clusters in Mycobacterium
rative disorders is expanded, therapeutic approaches are being tuberculosis, a facultative pathogen that can live inside macro-
explored as well. Thus far, some success has been observed in phages and lead to tuberculosis. These bacteria contain a
patients with Parkinson’s and other diseases in this area through family of WhiB-like 4Fe–4S cluster-containing transcriptional
the use of iron-chelation therapy.111b However, only one of the regulators that could be exploited as an attractive potential
chelators has been successful in clinical trials,122 since many of antibacterial target, since these regulators play a vital role in
them cause adverse side effects due to lack of targeting to specific mycobacterial cell division, lipid metabolism, oxidative stress,
tissues, resulting in systemic iron depletion.111b,112 New com- and antibiotic resistance.134
pounds or approaches that include targeting with fewer broad
effects could provide a successful treatment option.
15. Erythropoietic protoporphyria. Proper Fe–S cluster bio- Summary and perspectives
synthesis is also crucial for the pathway of heme biosynthesis,
since the last step in the process is catalyzed by ferrochelatase Since Fe–S clusters are required in a number of essential
(FECH), which binds a [2Fe–2S] cluster.123 Mutations in FECH proteins or as necessary cofactors for a multitude of enzymatic
account for 98% of erythropoietic protoporphyria (EPP) with reactions and physiological processes, conditions that perturb
patients affected worldwide124 and prevalence as high as 45% the process of cluster biogenesis and trafficking often lead to
in some countries.125 Symptoms of EPP include heightened and disease states. Additionally, a number of these perturbations lead
painful photosensitivity, and in some severe cases, liver damage to lethal consequences, making them challenging to investigate,
occurs;124 all of which are due to the accumulation of the heme which highlights the importance of understanding the molecular
precursor, protoporphyrin IX, from a decrease in FECH activity details of Fe–S cluster biosynthesis. Furthermore, recent work in
to less than 30%.126 The majority of patients present with the bacteria, utilizing various double mutants and media supplemen-
IVS3-48T 4 C low expression allele in conjunction with a missense tation, has demonstrated that these genetically engineered forms
mutation that causes a loss of FECH function, although other can grow, albeit slowly, without Fe–S clusters.135 The implications

18 | Metallomics, 2018, 10, 9--29 This journal is © The Royal Society of Chemistry 2018
View Article Online

Metallomics Minireview

of this work in relation to human Fe–S cluster-based diseases are exquisite balance to escape ferroptosis, Free Radical Biol.
currently unclear, but may provide alternative treatment strategies Med., 2017, 108, 610–626.
in the future. 4 (a) R. Lill, Function and biogenesis of iron–sulphur pro-
To date, the majority of inborn human diseases related to teins, Nature, 2009, 460, 831–838; (b) R. Lill, B. Hoffmann,
biogenesis or Fe–S cluster binding proteins are caused by mito- S. Molik, A. J. Pierik, N. Rietzschel, O. Stehling, M. A.
chondrial proteins.1a,2b No specific diseases have been associated Uzarska, H. Webert, C. Wilbrecht and U. Muhlenhoff, The
with the cytosolic system, even though they have demonstrated role of mitochondria in cellular iron–sulfur protein bio-
roles in yeast viability (Table 1).1a,4c,15,75,98,136 Furthermore, dis- genesis and iron metabolism, Biochim. Biophys. Acta, 2012,
eases have been linked to nuclear proteins that bind Fe–S clusters 1823, 1491–1508; (c) D. J. Netz, J. Mascarenhas, O. Stehling,
to function in DNA replication and repair;2a,80,82–84,86,90a,93,95,100c,d A. J. Pierik and R. Lill, Maturation of cytosolic and nuclear
however, the presence of the cluster and some of the specific iron–sulfur proteins, Trends Cell Biol., 2014, 24, 303–312.
cellular functions remain unclear, presenting challenges when 5 (a) D. C. Johnson, D. R. Dean, A. D. Smith and M. K.
trying to understand the role of the cluster in disease conditions. Johnson, Structure, function, and formation of biological
Aside from diseases associated with specific Fe–S cluster pro- iron–sulfur clusters, Annu. Rev. Biochem., 2005, 74,
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

teins, a subset of neurological disorders, such as Alzheimer’s and 247–281; (b) R. Lill and U. Mühlenhoff, Iron–sulfur protein
Parkinson’s, have been linked to the Fe–S cluster biogenesis biogenesis in eukaryotes: components and mechanisms,
pathway based on the presence of free iron and overall iron Annu. Rev. Cell Dev. Biol., 2006, 22, 457–486; (c) C. Ayala-
dysregulation in mitochondria.111 In addition, Fe–S cluster pro- Castro, A. Saini and F. W. Outten, Microbiol. Mol. Biol. Rev.,
teins from all of the cellular compartments have been implicated 2008, 72, 110; (d) M. Fontecave and S. Ollagnier-de-
in a variety of cancers.3 The essential nature and necessary Choudens, Arch. Biochem. Biophys., 2008, 474; (e) W. Qi
function of these metal cofactors dictates the need for a better and J. A. Cowan, Structural, mechanistic and coordination
understanding of their biogenesis and trafficking mechanisms to chemistry of relevance to the biosynthesis of iron–sulfur
clarify the molecular basis of disease states and lead to potential and related iron cofactors, Coord. Chem. Rev., 2011, 255,
therapeutic approaches. 688–699.
6 (a) A. Pandey, H. Yoon, E. R. Lyver, A. Dancis and
D. Pain, Isd11p protein activates the mitochondrial
Conflicts of interest cysteine desulfurase Nfs1p protein, J. Biol. Chem., 2011,
The authors declare no conflicts of interest. 286, 38242–38252; (b) A. Adam, C. Bornhovd, H. Prokisch,
W. Neupert and K. Hell, The Nfs1 interacting protein Isd11
has an essential role in Fe/S cluster biogenesis in mito-
Acknowledgements chondria, EMBO J., 2006, 25, 174–183.
7 (a) O. Gakh, W. Ranatunga, D. Y. Smith IV, E.-C. Ahlgren,
This work was supported by a grant from the National Institutes S. Al-Karadaghi, J. R. Thompson and G. Isaya, Architecture
of Health [AI072443]. Christine Wachnowsky was supported by an of the human mitochondrial iron–sulfur cluster assembly
NIH Chemistry/Biology Interface training grant (T32 GM095450) machinery, J. Biol. Chem., 2016, 291, 21296–29321;
and an Ohio State University Presidential Fellowship. (b) W. Ranatunga, O. Gakh, B. K. Galeano, D. Y. Smith,
C. A. Söderberg, S. Al-Karadaghi, J. R. Thompson and
References G. Isaya, Architecture of the yeast mitochondrial iron–sulfur
cluster assembly machinery: the sub-complex formed by the
1 (a) N. Maio and T. A. Rouault, Iron–sulfur cluster biogen- iron donor, Yfh1 protein, and the scaffold, Isu1 protein,
esis in mammalian cells: new insights into the molecular J. Biol. Chem., 2016, 291, 10378–10398; (c) S. Schmucker,
mechanisms of cluster delivery, Biochim. Biophys. Acta, A. Martelli, F. Colin, A. Page, M. Wattenhofer-Donzé,
2015, 1853, 1493–1512; (b) J. J. Braymer and R. Lill, Iron– L. Reutenauer and H. Puccio, Mammalian frataxin: an essen-
sulfur cluster biogenesis and trafficking in mitochondria, tial function for cellular viability through an interaction with
J. Biol. Chem., 2017, 292, 12754–12763; (c) T. A. Rouault and a preformed ISCU/NFS1/ISD11 iron–sulfur assembly com-
N. Maio, Biogenesis and functions of mammalian iron–sulfur plex, PLoS One, 2011, 6, e16199; (d) H. Webert, S.-A.
proteins in the regulation of iron homeostasis and pivotal Freibert, A. Gallo, T. Heidenreich, U. Linne, S. Amlacher,
metabolic pathways, J. Biol. Chem., 2017, 292, 12744–12753. E. Hurt, U. Mühlenhoff, L. Banci and R. Lill, Functional
2 (a) J. O. Fuss, C.-L. Tsai, J. P. Ishida and J. A. Tainer, reconstitution of mitochondrial Fe/S cluster synthesis on
Emerging critical roles of Fe–S clusters in DNA replication Isu1 reveals the involvement of ferredoxin, Nat. Commun.,
and repair, Biochim. Biophys. Acta, 2015, 1853, 1253–1271; 2014, 5, 5013.
(b) O. Stehling, C. Wilbrecht and R. Lill, Mitochondrial 8 (a) Y. Shi, M. Ghosh, G. Kovtunovych, D. R. Crooks and
iron–sulfur protein biogenesis and human disease, T. A. Rouault, Both human ferredoxins 1 and 2 and
Biochimie, 2014, 100, 61–77. ferredoxin reductase are important for iron–sulfur cluster
3 S. Toyokuni, F. Ito, K. Yamashita, Y. Okazaki and biogenesis, Biochim. Biophys. Acta, 2012, 1823, 484–492;
S. Akatsuka, Iron and thiol redox signaling in cancer: an (b) Y. Liu and J. A. Cowan, Iron sulfur cluster biosynthesis.

This journal is © The Royal Society of Chemistry 2018 Metallomics, 2018, 10, 9--29 | 19
View Article Online

Minireview Metallomics

Human NFU mediates sulfide delivery to ISU in the final studies, Biochim. Biophys. Acta, 2015, 1853, 1416–1428;
step of [2Fe–2S] cluster assembly, Chem. Commun., 2007, (e) N. Maio and T. A. Rouault, Mammalian Fe–S proteins:
3192–3194; (c) S.-P. Wu, M. Bellei, S. S. Mansy, definition of a consensus motif recognized by the co-
G. Battistuzzi, M. Sola and J. A. Cowan, Redox chemistry chaperone HSC20, Metallomics, 2016, 8, 1032–1046.
of the Schizosaccharomyces pombe ferredoxin electron- 14 (a) M. Uzarska, R. Dutkiewicz, S. Freibert, R. Lill and
transfer domain and influence of Cys to Ser substitutions, U. Muhlenhoff, The mitochondrial Hsp70 chaperone
J. Inorg. Biochem., 2011, 105, 806–811; (d) K. Cai, M. Tonelli, Ssq1 facilitates Fe/S cluster transfer from Isu1 to Grx5 by
R. O. Frederick and J. L. Markley, Human mitochondrial complex formation, Mol. Biol. Cell, 2013, 24, 1830–1841;
ferredoxin 1 (FDX1) and ferredoxin 2 (FDX2) both bind (b) H. Ye, S. Jeong, M. Ghosh, G. Kovtunovych, L. Silvestri,
cysteine desulfurase and donate electrons for iron–sulfur D. Ortillo, N. Uchida, J. Tisdale, C. Camaschella and
cluster biosynthesis, Biochemistry, 2017, 56, 487–499. T. Rouault, Glutaredoxin 5 deficiency causes sideroblastic
9 (a) G. Shaw, J. Cope, L. Li, K. Corson, C. Hersey, anemia by specifically impairing heme biosynthesis and
G. Ackermann, B. Gwynn, A. Lambert, R. Wingert, depleting cytosolic iron in human erythroblasts, J. Clin.
D. Traver, N. Trede, B. Barut, Y. Zhou, E. Minet, Invest., 2010, 120, 1749–1761; (c) C. Lillig, C. Berndt and
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

A. Donovan, A. Brownlie, R. Balzan, M. Weiss, L. Peters, A. Holmgren, Glutaredoxin systems, Biochim. Biophys. Acta,
J. Kaplan, L. Zon and B. Paw, Mitoferrin is essential for 2008, 1780, 1304–1317; (d) J. N. Vranish, D. Das and
erythroid iron assimilation, Nature, 2006, 440, 96–100; D. P. Barondeau, Real-time kinetic probes support mono-
(b) P. Paradkar, K. Zumbrennen, B. Paw, D. Ward and thiol glutaredoxins as intermediate carriers in Fe–S cluster
J. Kaplan, Regulation of mitochondrial iron import through biosynthetic pathways, ACS Chem. Biol., 2016, 11,
differential turnover of mitoferrin 1 and mitoferrin 2, Mol. 3114–3121.
Cell. Biol., 2009, 29, 1007–1016. 15 R. Lill, R. Dutkiewicz, S. A. Freibert, T. Heidenreich,
10 (a) G. Isaya, H. O’Neill, O. Gakh, S. Park, R. Mantcheva and J. Mascarenhas, D. J. Netz, V. D. Paul, A. J. Pierik,
S. Mooney, Functional studies of frataxin, Acta Paediatr., N. Richter, M. Stümpfig, V. Srinivasan, O. Stehling and
2004, 93, 68–72; (b) M. Nair, S. Adinolfi, C. Pastore, U. Mühlenhoff, The role of mitochondria and the CIA
G. Kelly, P. Temussi and A. Pastore, Solution structure of machinery in the maturation of cytosolic and nuclear
the bacterial frataxin ortholog, CyaY: mapping the iron iron–sulfur proteins, Eur. J. Cell Biol., 2015, 94, 280–291.
binding sites, Structure, 2004, 12, 2037–2048. 16 (a) J. Y. Lee, J. G. Yang, D. Zhitnitsky, O. Lewinson and
11 F. Colin, A. Martelli, M. Clemancey, J. Latour, D. C. Rees, Structural basis for heavy metal detoxification
S. Gambarelli, L. Zeppieri, C. Birck, A. Page, H. Puccio by an Atm1-type ABC exporter, Science, 2014, 343,
and S. O. S. Choudens, Mammalian frataxin controls sulfur 1133–1136; (b) V. Srinivasan, A. J. Pierik and R. Lill, Crystal
production and iron entry during de novo Fe4S4 cluster structures of nucleotide-free and glutathione-bound mito-
assembly, J. Am. Chem. Soc., 2013, 135, 733–740. chondrial ABC transporter Atm1, Science, 2014, 343,
12 (a) C. Tsai and D. Barondeau, Human frataxin is an 1137–1140.
allosteric switch that activates the Fe–S cluster biosynthetic 17 (a) W. Qi, J. Li, C. Y. Chain, G. A. Pasquevich,
complex, Biochemistry, 2010, 49, 9132–9139; (b) B. K. Galeano, A. F. Pasquevich and J. A. Cowan, Glutathione complexed
W. Ranatunga, O. Gakh, D. Y. Smith, J. R. Thompson and Fe–S centers, J. Am. Chem. Soc., 2012, 134, 10745–10748;
G. Isaya, Zinc and the iron donor frataxin regulate oligo- (b) J. Li and J. A. Cowan, Glutathione-coordinated [2Fe–2S]
merization of the scaffold protein to form new Fe–S cluster cluster: a viable physiological substrate for mitochondrial
assembly centers, Metallomics, 2017, 9, 773–801. ABCB7 transport, Chem. Commun., 2015, 51, 2253–2255;
13 (a) A. D. Sheftel, O. Stehling, A. J. Pierik, H.-P. Elsässer, (c) W. Qi, J. Li and J. A. Cowan, A structural model for
U. Mühlenhoff, H. Webert, A. Hobler, F. Hannemann, glutathione-complexed iron–sulfur cluster as a substrate
R. Bernhardt and R. Lill, Humans possess two mitochon- for ABCB7-type transporters, Chem. Commun., 2014, 50,
drial ferredoxins, Fdx1 and Fdx2, with distinct roles in 3795.
steroidogenesis, heme, and Fe/S cluster biosynthesis, 18 (a) P. Cavadini, G. Biasiotto, M. Poli, S. Levi, R. Verardi,
Proc. Natl. Acad. Sci. U. S. A., 2010, 107, 11775–11780; I. Zanella, M. Derosas, R. Ingrassia, M. Corrado and
(b) L. Vickery and J. Cupp-Vickery, Molecular chaperones P. Arosio, RNA silencing of the mitochondrial ABCB7
HscA/Ssq1 and HscB/Jac1 and their roles in ironesulfur transporter in HeLa cells causes an iron-deficient pheno-
protein maturation, Crit. Rev. Biochem. Mol. Biol., 2007, 42, type with mitochondrial iron overload, Blood, 2007, 109,
95–111; (c) H. Uhrigshardt, A. Singh, G. Kovtunovych, 3552–3559; (b) R. Miao, H. Kim, U. M. K. Koppolu,
M. Ghosh and T. Rouault, Characterization of the human E. A. Ellis, R. A. Scott and P. A. Lindahl, Biophysical
HSC20, an unusual DnaJ type III protein, involved in characterization of the iron in mitochondria from
ironesulfur cluster biogenesis, Hum. Mol. Genet., 2010, Atm1p-depleted Saccharomyces cerevisiae, Biochemistry,
19, 3816–3834; (d) J. H. Kim, J. R. Bothe, T. R. Alderson 2009, 48, 9556–9568.
and J. L. Markley, Tangled web of interactions among 19 C. Kumar, A. Igbaria, B. D’Autreaux, A. G. Planson,
proteins involved in iron–sulfur cluster assembly as unrav- C. Junot, E. Godat, A. K. Bachhawat, A. Delaunay-Moisan
eled by NMR, SAXS, chemical crosslinking, and functional and M. B. Toledano, Glutathione revisited: a vitalfunction

20 | Metallomics, 2018, 10, 9--29 This journal is © The Royal Society of Chemistry 2018
View Article Online

Metallomics Minireview

in iron metabolism and ancillary role in thiol-redox con- mitochondrial iron–sulfur cluster biogenesis, Structure,
trol, EMBO J., 2011, 30, 2044–2056. 2016, 24, 2080–2091.
20 T. A. Schaedler, J. D. Thornton, I. Kruse, M. Schwarzlander, 24 (a) A. Hausmann, D. J. A. Netz, J. Balk, A. J. Pierik,
A. J. Meyer, H. W. van Veen and J. Balk, A conserved U. Muhlenhoff and R. Lill, The eukaryotic P loopNTPa-
mitochondrial ATP-binding cassette transporter exports seNbp35: an essential component of the cytosolic and
glutathione polysulfide for cytosolic metal cofactor assem- nuclear iron–sulfur protein assembly machinery, Proc.
bly, J. Biol. Chem., 2014, 289, 23264–23274. Natl. Acad. Sci. U. S. A., 2005, 102, 3266–3271; (b) D. J.
21 (a) C. Gelling, I. W. Dawes, N. Richhardt, R. Lill and Netz, A. J. Pierik, M. Stumpfig, U. Muhlenhoff and R. Lill,
U. Muhlenhoff, Mitochondrial Iba57p is required for Fe/S The Cfd1–Nbp35 complex acts as a scaffold for iron–
cluster formation on aconitase and activation of radical sulfur protein assembly in the yeast cytosol, Nat. Chem.
SAM enzymes, Mol. Cell. Biol., 2008, 28, 1851–1861; Biol., 2007, 3, 278–286; (c) O. Stehling, D. J. A. Netz,
(b) U. Muhlenhoff, N. Richter, O. Pines, A. J. Pierik and B. Niggemeyer, R. Rosser, R. S. Eisenstein, H. Puccio,
R. Lill, Specialized function of yeast Isa1 and Isa2 proteins A. J. Pierik and R. Lill, Human Nbp35 is essential for both
in the maturation of mitochondrial [4Fe–4S] proteins, cytosolic iron–sulfur protein assembly and iron homeo-
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

J. Biol. Chem., 2011, 286, 41205–41216; (c) A. D. Sheftel, stasis, Mol. Cell. Biol., 2008, 28, 5517–5528; (d) D. J.
C. Wilbrecht, O. Stehling, B. Niggemeyer, H. P. Elsasser, Netz, A. J. Pierik, M. Stumpfig, E. Bill, A. K. Sharma,
U. Muhlenhoff and R. Lill, The human mitochondrial L. J. Pallesen, W. E. Walden and R. Lill, A bridging
ISCA1, ISCA2, and IBA57 proteins are required for [4Fee4S] [4Fe–4S] cluster and nucleotide binding are essential for
protein maturation, Mol. Biol. Cell, 2012, 23, 1157–1166; function of the Cfd1–Nbp35 complex as a scaffold in
(d) L. K. Beilschmidt, S. Ollagnier de Choudens, M. Fournier, iron–sulfur protein maturation, J. Biol. Chem., 2012, 287,
I. Sanakis, M.-A. Hograindleur, M. Clémancey, G. Blondin, 12365–12378.
S. Schmucker, A. Eisenmann, A. Weiss, P. Koebel, 25 (a) L. Banci, I. Bertini, V. Calderone, S. Ciofi-Baffoni,
N. Messaddeq, H. Puccio and A. Martelli, ISCA1 is essential A. Giachetti, D. Jaiswal, M. Mikolajczyk, M. Piccioli and
for mitochondrial Fe4S4 biogenesis in vivo, Nat. Commun., J. Winkelmann, Molecular view of an electron transfer
2017, 8, 15124. process essential for iron–sulfur protein biogenesis,
22 (a) K. D. Kim, W. H. Chung, H. J. Kim, K. C. Lee and Proc. Natl. Acad. Sci. U. S. A., 2013, 110, 7136–7141;
J. H. Roe, Monothiol glutaredoxin Grx5 interacts with Fe–S (b) D. J. A. Netz, M. Stümpfig, C. Doré, U. Mühlenhoff,
scaffold proteins Isa1 and Isa2 and supports Fe–S assem- A. J. Pierik and R. Lill, Tah18 transfers electrons to dre2 in
bly and DNA integrity in mitochondria of fission yeast, cytosolic iron–sulfur protein biogenesis, Nat. Chem. Biol.,
Biochem. Biophys. Res. Commun., 2010, 392, 467–472; 2010, 6, 758–765.
(b) L. Banci, D. Brancaccio, S. Ciofi-Baffoni, R. Del Conte, 26 (a) P. Haunhorst, E. M. Hanschmann, L. Brautigam,
R. Gadepalli, M. Mikolajczyk, S. Neri, M. Piccioli and O. Stehling, B. Hoffmann, U. Muhlenhoff, R. Lill,
J. Winkelmann, [2Fe–2S] cluster transfer in iron–sulfur C. Berndt and C. H. Lillig, Crucial function of vertebrate
protein biogenesis, Proc. Natl. Acad. Sci. U. S. A., 2014, glutaredoxin 3 (PICOT) in iron homeostasis and hemo-
111, 6203–6208; (c) D. Brancaccio, A. Gallo, M. Mikolajczyk, globin maturation, Mol. Biol. Cell, 2013, 24, 1895–1903;
K. Zovo, P. Palumaa, E. Novellino, M. Piccioli, S. Ciofi- (b) U. Mühlenhoff, S. Molik, J. R. Godoy, M. A. Uzarska,
Baffoni and L. Banci, Formation of [4Fe–4S] clusters in the N. Richter, A. Seubert, Y. Zhang, J. Stubbe, F. Pierrel,
mitochondrial iron–sulfur cluster assembly machinery, E. Herrero, C. H. Lillig and R. Lill, Cytosolic Monothiol
J. Am. Chem. Soc., 2014, 136, 16240–16250. Glutaredoxins Function in Intracellular Iron Sensing and
23 (a) W. H. Tong, G. N. L. Jameson, B. H. Huynh and Trafficking via Their Bound Iron–Sulfur Cluster, Cell
T. A. Rouault, Subcellular compartmentalization of human Metab., 2010, 12, 373–385.
Nfu, an iron–sulfur cluster scaffold protein, and its ability 27 (a) C. E. Outten and A. N. Albetel, Iron sensing and
to assemble a [4Fe–4S] cluster, Proc. Natl. Acad. Sci. U. S. A., regulation in Saccharomyces cerevisiae: ironing out the
2003, 100, 9762–9767; (b) H. Gao, S. Subramanian, mechanistic details, Curr. Opin. Microbiol., 2013, 16,
J. Couturier, S. G. Naik, S.-K. Kim, T. Leustek, D. B. 662–668; (b) Y. Zhang, L. Liu, X. Wu, X. An, J. Stubbe and
Knaff, H.-C. Wu, F. Vignols, B. H. Huynh, N. Rouhier and M. Huang, Investigation of in vivo diferric tyrosyl radical
M. K. Johnson, Arabidopsis thaliana Nfu2 accommodates formation in Saccharomyces cerevisiae Rnr2 protein:
[2Fe–2S] or [4Fe–4S] clusters and is competent for in vitro requirement of Rnr4 and contribution of Grx3/4 AND
maturation of chloroplast [2Fe–2S] and [4Fe–4S] cluster- Dre2 proteins, J. Biol. Chem., 2011, 286, 41499–41509.
containing proteins, Biochemistry, 2013, 52, 6633–6645; 28 (a) M. Seki, Y. Takeda, K. Iwai and K. Tanaka, IOP1 protein
(c) C. Wachnowsky, I. Fidai and J. A. Cowan, Iron–sulfur is an external component of the human cytosolic iron–
cluster exchange reactions mediated by the human Nfu sulfur cluster assembly (CIA) machinery and functions in
protein, J. Biol. Inorg. Chem., 2016, 21, 825–836; (d) K. Cai, the MMS19 protein-dependent CIA pathway, J. Biol. Chem.,
G. Liu, R. O. Frederick, R. Xiao, G. T. Montelione and 2013, 288, 16680–16689; (b) V. Srinivasan, D. J. Netz,
John L. Markley, Structural/functional properties of H. Webert, J. Mascarenhas, A. J. Pierik, H. Michel and
human NFU1, an intermediate [4Fe–4S] carrier in human R. Lill, Structure of the yeast WD40 domain protein Cia1,

This journal is © The Royal Society of Chemistry 2018 Metallomics, 2018, 10, 9--29 | 21
View Article Online

Minireview Metallomics

a component acting late in iron–sulfur protein bio- 34 A. Olsson, L. Lind, L. E. Thornell and M. Holmberg,
genesis, Structure, 2007, 15, 1246–1257; (c) O. Stehling, Myopathy with lactic acidosis is linked to chromosome
J. Mascarenhas, A. A. Vashisht, A. D. Sheftel, 12q23.3-24.11 and caused by an intron mutation in the
B. Niggemeyer, R. Rosser, A. J. Pierik, J. A. Wohlschlegel ISCU gene resulting in a splicing defect, Hum. Mol. Genet.,
and R. Lill, Human CIA2A–FAM96A and CIA2B–FAM96B 2008, 17, 1666–1672.
integrate iron homeostasis and maturation of different 35 (a) P. S. Sanaker, M. Toompuu, V. E. Hogan, L. He,
subsets of cytosolic–nuclear iron–sulfur proteins, Cell C. Tzoulis, Z. M. C. Lightowlers, R. W. Taylor and
Metab., 2013, 18, 187–198; (d) N. V. Wietmarschen, L. A. Bindoff, Differences in RNA processing underlie the
A. Moradian, G. B. Morin, P. M. Lansdorp and tissue specific phenotype of ISCU myopathy, Biochim.
E. J. Uringa, The mammalian proteins MMS19, MIP18, Biophys. Acta, 2010, 1802, 539–544; (b) G. Kollberg and
and ANT2 are involved in cytoplasmic iron–sulfur cluster E. Holme, Antisense oligonucleotide therapeutics for iron-
protein assembly, J. Biol. Chem., 2012, 287, 43351–43358; sulphur cluster deficiency myopathy, Neuromuscular Dis-
(e) E. Weerapana, C. Wang, G. M. Simon, F. Richter, ord., 2009, 19, 833–836.
S. Khare, M. B. Dillon, D. A. Bachovchin, K. Mowen, 36 K. White, Y. Lu, S. Annis, A. E. Hale, B. N. Chau, J. E.
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

D. Baker and B. F. Cravatt, Quantitative reactivity profiling Dahlman, C. Hemann, A. R. Opotowsky, S. O. Vargas,
predicts functional cysteines in proteomes, Nature, 2010, I. Rosas, M. A. Perrella, J. C. Osorio, K. J. Haley, B. B.
468, 790–795. Graham, R. Kumar, R. Saggar, W. D. Wallace, D. J. Ross,
29 (a) W. H. Tong, G. N. Jameson, B. H. Huynh and O. F. Khan, A. Bader, B. R. Gochuico, M. Matar, K. Polach,
T. A. Rouault, Subcellular compartmentalization of human N. M. Johannessen, H. M. Prosser, D. G. Anderson,
Nfu, an iron–sulfur cluster scaffold protein, and its ability R. Langer, J. L. Zweier, L. A. Bindoff, D. Systrom, A. B.
to assemble a [4Fe–4S] cluster, Proc. Natl. Acad. Sci. U. S. A., Waxman, R. C. Jin and S. Y. Chan, Genetic and hypoxic
2003, 100, 9762–9767; (b) W. H. Tong and T. A. Rouault, alterations of the microRNA-210-ISCU1/2 axis promote
Functions of mitochondrial ISCU and cytosolic ISCU in iron–sulfur deficiency and pulmonary hypertension, EMBO
mammalian iron–sulfur cluster biogenesis and iron Mol. Med., 2015, 7, 695–713.
homeostasis, Cell Metab., 2006, 3, 199–210; (c) Y. Shi, 37 Y. Funauchi, C. Tanikawa, P. H. Yi Lo, J. Mori, Y. Daigo,
M. C. Ghosh, W. H. Tong and T. A. Rouault, Human A. Takano, Y. Miyagi, A. Okawa, Y. Nakamura and
ISD11 is essential for both ironesulfur cluster assembly K. Matsuda, Regulation of iron homeostasis by the p53-
and maintenance of normal cellular iron homeostasis, ISCU pathway, Sci. Rep., 2015, 5, 16497.
Hum. Mol. Genet., 2009, 18, 3014–3025; (d) T. Land and 38 R. Spiegel, A. Saada, J. Halvardson, D. Soiferman, A. Shaag,
T. A. Rouault, Targeting of a human ironesulfur cluster S. Edvardson, Y. Horovitz, M. Khayat, S. A. Shalev, L. Feuk
assembly enzyme, nifs, to different subcellular compart- and O. Elpeleg, Deleterious mutation in FDX1L gene is
ments is regulated through alternative AUG utilization, associated with a novel mitochondrial muscle myopathy,
Mol. Cell, 1998, 2, 807–815. Eur. J. Hum. Genet., 2013, 22, 902–906.
30 (a) A. H. Koeppen, Friedreich’s ataxia: pathology, patho- 39 S. M. K. Farhan, J. Wang, J. F. Robinson, P. Lahiry,
genesis, and molecular genetics, J. Neurol. Sci., 2011, 303, V. M. Siu, C. Prasad, J. B. Kronick, D. A. Ramsay,
1–12; (b) R. Santos, S. Lefevre, D. Sliwa, A. Seguin, J.-M. C. A. Rupar and R. A. Hegele, Exome sequencing identifies
Camadro and E. Lesuisse, Friedreich ataxia: molecular NFS1 deficiency in a novel Fe–S cluster disease, infantile
mechanisms, redox considerations, and therapeutic mitochondrial complex II/III deficiency, Mol. Genet.
opportunities, Antioxid. Redox Signaling, 2010, 13, Genomic Med., 2014, 2, 73–80.
651–690; (c) M. Stamelou, Late-onset cerebellar ataxia: do 40 (a) S. C. Lim, M. Friemel, J. E. Marum, E. J. Tucker, D. L.
not forget Friedreich’s, Mov. Disord, 2016, 31, 7–8. Bruno, L. G. Riley, J. Christodoulou, E. P. Kirk, A. Boneh,
31 S. Schmucker and H. Puccio, Understanding the molecular C. M. DeGennaro, M. Springer, V. K. Mootha, T. A. Rouault,
mechanisms of Friedreich’s ataxia to develop therapeutic S. Leimkuhler, D. R. Thorburn and A. G. Compton, Muta-
approaches, Hum. Mol. Genet., 2010, 19, 103–110. tions in LYRM4, encoding iron–sulfur cluster biogenesis
32 M. G. Cotticelli, L. Rasmussen, N. L. Kushner, S. McKellip, factor ISD11, cause deficiency of multiple respiratory chain
M. I. Sosa, A. Manouvakhova, S. Feng, E. L. White, J. A. complexes, Hum. Mol. Genet., 2013, 22, 4460–4473;
Maddry, J. Heemskerk, R. J. Oldt, L. F. Surrey, R. Ochs and (b) P. P. Saha, S. Srivastava, S. K. P. Kumar, D. Sinha and
R. B. Wilson, Primary and secondary drug screening assays P. D’Silva, Mapping key residues of ISD11 critical for
for Friedreich ataxia, J. Biomol. Screening, 2012, 17, NFS1–ISD11 subcomplex stability, J. Biol. Chem., 2015,
303–313. 290, 25876–25890.
33 F. Mochel, M. A. Knight, W. H. Tong, D. Hernandez, 41 (a) U. Ahting, J. A. Mayr, A. V. Vanlander, S. A. Hardy,
K. Ayyad, T. Taivassalo, P. M. Andersen, A. Singleton, S. Santra, C. Makowski, C. L. Alston, F. A. Zimmermann,
T. A. Rouault, K. H. Fischbeck and R. G. Haller, Splice L. Abela, B. Plecko, M. Rohrbach, S. Spranger, S. Seneca,
mutation in the ironesulfur cluster scaffold protein ISCU B. Rolinski, A. Hagendorff, M. Hempel, W. Sperl,
causes myopathy with exercise intolerance, Am. J. Hum. T. Meitinger, J. Smet, R. W. Taylor, R. Van Coster,
Genet., 2008, 82, 652–660. P. Freisinger, H. Prokisch and T. B. Haack, Clinical,

22 | Metallomics, 2018, 10, 9--29 This journal is © The Royal Society of Chemistry 2018
View Article Online

Metallomics Minireview

biochemical, and genetic spectrum of seven patients 43 (a) N. Ajit Bolar, A. V. Vanlander, C. Wilbrecht,
with NFU1 deficiency, Front. Genet., 2015, 6, 123; N. Van der Aa, J. Smet, B. De Paepe, G. Vandeweyer,
(b) J. M. Cameron, A. Janer, V. Levandovskiy, N. Mackay, F. Kooy, F. Eyskens, E. De Latter, G. Delanghe,
T. A. Rouault, W. H. Tong, I. Ogilvie, E. A. Shoubridge and P. Govaert, J. G. Leroy, B. Loeys, R. Lill, L. Van Laer and
B. H. Robinson, Mutations in iron–sulfur cluster scaffold R. Van Coster, Mutation of the iron–sulfur cluster assembly
genes NFU1 and BOLA3 cause a fatal deficiency of multiple gene IBA57 causes severe myopathy and encephalopathy,
respiratory chain and 2-oxoacid dehydrogenase enzymes, Hum. Mol. Genet., 2013, 22, 2590–2602; (b) F.-G. Debray,
Am. J. Hum. Genet., 2011, 89, 486–495; (c) X. Ferrer-Cortès, C. Stümpfig, A. V. Vanlander, V. Dideberg, C. Josse, J.-H.
A. Font, N. Bujan, A. Navarro-Sastre, L. Matalonga, Caberg, F. Boemer, V. Bours, R. Stevens, S. Seneca, J. Smet,
J. A. Arranz, E. Riudor, M. del Toro, A. Garcia-Cazorla, R. Lill and R. van Coster, Mutation of the iron–sulfur
J. Campistol, P. Briones, A. Ribes and F. Tort, Protein cluster assembly gene IBA57 causes fatal infantile leuko-
expression profiles in patients carrying NFU1 mutations. dystrophy, J. Inherited Metab. Dis., 2015, 38, 1147–1153;
Contribution to the pathophysiology of the disease, (c) A. Lossos, C. Stümpfig, G. Stevanin, M. Gaussen, B.-E.
J. Inherited Metab. Dis., 2012, 36, 841–847; (d) X. Ferrer- Zimmerman, E. Mundwiller, M. Asulin, L. Chamma,
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

Cortès, J. Narbona, N. Bujan, L. Matalonga, M. Del Toro, R. Sheffer, A. Misk, S. Dotan, J. M. Gomori, P. Ponger,
J. A. Arranz, E. Riudor, A. Garcia-Cazorla, C. Jou, A. Brice, I. Lerer, V. Meiner and R. Lill, Fe/S protein
M. O’Callaghan, M. Pineda, R. Montero, A. Arias, assembly gene IBA57 mutation causes hereditary spastic
J. Garcı́a-Villoria, C. L. Alston, R. W. Taylor, P. Briones, paraplegia, Neurology, 2015, 84, 659–667; (d) A. Torraco,
A. Ribes and F. Tort, A leaky splicing mutation in NFU1 is A. Ardissone, F. Invernizzi, T. Rizza, G. Fiermonte,
associated with a particular biochemical phenotype. M. Niceta, N. Zanetti, D. Martinelli, A. Vozza, D. Verrigni,
Consequences for the diagnosis, Mitochondrion, 2016, 26, M. Di Nottia, E. Lamantea, D. Diodato, M. Tartaglia,
72–80; (e) F. Invernizzi, A. Ardissone, E. Lamantea, C. Dionisi-Vici, I. Moroni, L. Farina, E. Bertini, D. Ghezzi
B. Garavaglia, M. Zeviani, L. Farina, D. Ghezzi and and R. Carrozzo, Novel mutations in IBA57 are associated
I. Moroni, Cavitating leukoencephalopathy with multiple with leukodystrophy and variable clinical phenotypes,
mitochondrial dysfunction syndrome and NFU1 muta- J. Neurol., 2016, 102–111.
tions, Front. Genet., 2014, 5, 412; ( f ) A. Navarro-Sastre, 44 Z. N. Al-Hassnan, M. Al-Dosary, M. Alfadhel, E. A. Faqeih,
F. Tort, O. Stehling, M. A. Uzarska, J. A. Arranz, M. del M. Alsagob, R. Kenana, R. Almass, O. S. Al-Harazi,
Toro, M. T. Labayru, J. Landa, A. Font, J. Garcia-Villoria, H. Al-Hindi, O. I. Malibari, F. B. Almutari, S. Tulbah,
B. Merinero, M. Ugarte, L. G. Gutierrez-Solana, F. Alhadeq, T. Al-Sheddi, R. Alamro, A. AlAsmari,
J. Campistol, A. Garcia-Cazorla, J. Vaquerizo, E. Riudor, M. Almuntashri, H. Alshaalan, F. A. Al-Mohanna,
P. Briones, O. Elpeleg, A. Ribes and R. Lill, A fatal mito- D. Colak and N. Kaya, ISCA2 mutation causes infantile
chondrial disease is associated with defective NFU1 func- neurodegenerative mitochondrial disorder, J. Med. Genet.,
tion in the maturation of a subset of mitochondrial 2015, 52, 186–194.
Fe–S proteins, Am. J. Hum. Genet., 2011, 89, 656–667; 45 A. Shukla, M. Hebbar, A. Srivastava, R. Kadavigere,
(g) D. Tonduti, I. Dorboz, A. Imbard, A. Slama, P. Upadhyai, A. Kanthi, O. Brandau, S. Bielas and
A. Boutron, S. Pichard, M. Elmaleh, L. Vallée, J. Benoist, K. M. Girisha, Homozygous p.(Glu87Lys) variant in ISCA1
H. Ogier and O. Boespflug-Tanguy, New spastic paraplegia is associated with a multiple mitochondrial dysfunctions
phenotype associated to mutation of NFU1, Orphanet syndrome, J. Hum. Genet., 2017, 62, 723–727.
J. Rare Dis., 2015, 10, 13. 46 J. M. Cameron, A. Janer, V. Levandovskiy, N. MacKay,
42 (a) P. R. Baker, M. W. Friederich, M. A. Swanson, T. Shaikh, T. A. Rouault, W.-H. Tong, I. Ogilvie, E. A. Shoubridge
K. Bhattacharya, G. H. Scharer, J. Aicher, G. Creadon- and B. H. Robinson, Mutations in iron–sulfur cluster
Swindell, E. Geiger, K. N. MacLean, W. T. Lee, C. Deshpande, scaffold genes NFU1 and BOLA3 cause a fatal deficiency
M. L. Freckmann, L. Y. Shih, M. Wasserstein, M. B. of multiple respiratory chain and 2-oxoacid dehydrogenase
Rasmussen, A. M. Lund, P. Procopis, J. M. Cameron, B. H. enzymes, Am. J. Hum. Genet., 2011, 89, 486–495.
Robinson, G. K. Brown, R. M. Brown, A. G. Compton, 47 C. Wachnowsky, N. A. Wesley, I. Fidai and J. A. Cowan,
C. L. Dieckmann, R. Collard, C. R. Coughlin, E. Spector, Understanding the molecular basis for Multiple Mitochon-
M. F. Wempe and J. L. K. Van Hove, Variant non drial Dysfunctions Syndrome 1 (MMDS1) – Impact of a
ketotic hyperglycinemia is caused by mutations in LIAS, disease-causing Gly208Cys substitution on structure and
BOLA3 and the novel gene GLRX5, Brain, 2013, 137, activity of NFU1 in the Fe/S cluster biosynthetic pathway,
366–379; (b) T. B. Haack, B. Rolinski, B. Haberberger, J. Mol. Biol., 2017, 429, 790–807.
F. Zimmermann, J. Schum, V. Strecker, E. Graf, 48 M. A. Uzarska, V. Nasta, B. D. Weiler, F. Spantgar, S. Ciofi-
U. Athing, T. Hoppen, I. Wittig, W. Sperl, P. Freisinger, Baffoni, M. R. Saviello, L. Gonnelli, U. Muehlenhoff, L. Banci
J. A. Mayr, T. M. Strom, T. Meitinger and H. Prokisch, and R. Lill, Mitochondrial Bol1 and Bol3 function as assembly
Homozygous missense mutation in BOLA3 causes multiple factors for specific iron–sulfur proteins, eLife, 2016, 5, e15991.
mitochondrial dysfunctions syndrome in two siblings, 49 A. Melber, U. Na, A. Vashisht, B. D. Weiler, R. Lill,
J. Inherited Metab. Dis., 2012, 36, 55–62. J. A. Wohlschlegel and D. R. Winge, Role of Nfu1 and

This journal is © The Royal Society of Chemistry 2018 Metallomics, 2018, 10, 9--29 | 23
View Article Online

Minireview Metallomics

Bol3 in Iiron–sulfur cluster transfer to mitochondrial Inst., 2016, 108, djv287; (b) N. Maio, A. Singh,
clients, eLife, 2016, 5, e15991. H. Uhrigshardt, N. Saxena and W.-H. Tong, Tracey A.
50 N. Maio, K. S. Kim, A. Singh and T. A. Rouault, A single Rouault, Cochaperone binding to LYR motifs confers
adaptable cochaperone-scaffold complex delivers nascent specificity of iron sulfur cluster delivery, Cell Metab.,
iron–sulfur clusters to mammalian respiratory chain com- 2014, 19, 445–457.
plexes I–III, Cell Metab., 2017, 25, 945–953. 59 U. Na, W. Yu, J. Cox, D. K. Bricker, K. Brockmann, J. Rutter,
51 I. E. Scheffler, Mitochondrial disease associated with C. S. Thummel and D. R. Winge, The LYR factors SDHAF1
complex I (NADH-CoQ oxidoreductase) deficiency, and SDHAF3 mediate maturation of the iron–sulfur sub-
J. Inherited Metab. Dis., 2014, 38, 405–415. unit of succinate dehydrogenase, Cell Metab., 2014, 20,
52 S. H. Kevelam, R. J. Rodenburg, N. I. Wolf, P. Ferreira, 253–266.
R. J. Lunsing, L. G. Nijtmans, A. Mitchell, H. A. Arroyo, 60 N. Maio, D. Ghezzi, D. Verrigni, T. Rizza, E. Bertini,
D. Rating, A. Vanderver, C. G. M. van Berkel, D. Martinelli, M. Zeviani, A. Singh and R. Carrozzo, Tracey
T. E. M. Abbink, P. Heutink and M. S. van der Knapp, A. Rouault, Disease-causing SDHAF1 mutations impair
NUBPL mutations in patients with complex I deficiency transfer of Fe–S clusters to SDHB, Cell Metab., 2016, 23,
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

and a distinct MRI pattern, Neurology, 2013, 80, 292–302.


1577–15783. 61 A. Ohlenbusch, S. Edvardson, J. Skorpen, A. Bjornstad,
53 (a) E. J. Tucker, M. Mimaki, A. G. Compton, M. McKenzie, A. Saada, O. Elpeleg, J. Gärtner and K. Brockmann, Leu-
M. T. Ryan and D. R. Thorburn, Next-generation sequenc- koencephalopathy with accumulated succinate is indica-
ing in molecular diagnosis: NUBPL mutations highlight tive of SDHAF1 related complex II deficiency, Orphanet
the challenges of variant detection and interpretation, J. Rare Dis., 2012, 7, 69.
Hum. Mutat., 2012, 33, 411–418; (b) M. M. Wydro and 62 S. Tamir, M. L. Paddock, M. Darash-Yahana-Baram, S. H.
J. Balk, Insights into the pathogenic character of a com- Holt, Y. S. Sohn, L. Agranat, D. Michaeli, J. T. Stofleth,
mon NUBPL branch-site mutation associated with mito- C. H. Lipper, F. Morcos, I. Z. Cabantchik, J. N. Onuchic,
chondrial disease and complex I deficiency using a yeast P. A. Jennings, R. Mittler and R. Nechushtai, Structure–
model, Dis. Models & Mech., 2013, 6, 1279–1284. function analysis of NEET proteins uncovers their role as
54 M. M. Wydro, P. Sharma, J. M. Foster, K. Bych, E. H. Meyer key regulators of iron and ROS homeostasis in health and
and J. Balk, The evolutionarily conserved iron–sulfur pro- disease, Biochim. Biophys. Acta, 2015, 1853, 1294–1315.
tein INDH is required for complex I assembly and mito- 63 W. J. Geldenhuys, T. C. Leeper and R. T. Carroll, mitoNEET
chondrial translation in Arabidopsis, Plant Cell, 2013, 25, as a novel drug target for mitochondrial dysfunction, Drug
4014–4027. Discovery Today, 2014, 19, 1601–1606.
55 (a) A. S. Hoekstra and J.-P. Bayley, The role of complex II in 64 C. M. Kusminski, W. L. Holland, K. Sun, J. Park, S. B.
disease, Biochim. Biophys. Acta, 2013, 1827, 543–551; Spurgin, Y. Lin, G. R. Askew, J. A. Simcox, D. A. McClain,
(b) J. G. Van Vranken, U. Na, D. R. Winge and J. Rutter, C. Li and P. E. Scherer, MitoNEET-driven alterations in
Protein-mediated assembly of succinate dehydrogenase adipocyte mitochondrial activity reveal a crucial adaptive
and its cofactors, Crit. Rev. Biochem. Mol. Biol., 2014, 50, process that preserves insulin sensitivity in obesity, Nat.
168–180. Med., 2012, 18, 1539–1549.
56 C. L. Alston, J. E. Davison, F. Meloni, F. H. van der 65 R. Nechushtai, A. R. Conlan, Y. Harir, L. Song, O. Yogev,
Westhuizen, L. He, H.-T. Hornig-Do, A. C. Peet, Y. Eisenberg-Domovich, O. Livnah, D. Michaeli, R. Rosen,
P. Gissen, P. Goffrini, I. Ferrero, E. Wassmer, V. Ma, Y. Luo, J. A. Zuris, M. L. Paddock, Z. I. Cabantchik,
R. McFarland and R. W. Taylor, Recessive germline SDHA P. A. Jennings and R. Mittler, Characterization of Arabi-
and SDHB mutations causing leukodystrophy and isolated dopsis NEET reveals an ancient role for NEET proteins in
mitochondrial complex II deficiency, J. Med. Genet., 2012, iron metabolism, Plant Cell, 2012, 24, 2139–2154.
49, 569–577. 66 I. Ferecatu, S. Goncalves, M. P. Golinelli-Cohen,
57 M. A. Pantaleo, A. Astolfi, M. Urbini, M. Nannini, M. Clemancey, A. Martelli, S. Riquier, E. Guittet, J. M.
P. Paterini, V. Indio, M. Saponara, S. Formica, Latour, H. Puccio, J. C. Drapier, E. Lescop and C. Bouton,
C. Ceccarelli, R. Casadio, G. Rossi, F. Bertolini, The diabetes drug target mitoNEET governs a novel traf-
D. Santini, M. G. Pirini, M. Fiorentino, U. Basso and ficking pathway to rebuild an Fe–S cluster into cytosolic
G. Biasco, Analysis of all subunits, SDHA, SDHB, SDHC, aconitase/iron regulatory protein 1, J. Biol. Chem., 2014,
SDHD, of the succinate dehydrogenase complex in KIT/ 289, 28070–28086.
PDGFRA wild-type GIST, Eur. J. Hum. Genet., 2013, 22, 67 M.-P. Golinelli-Cohen, E. Lescop, C. Mons, S. Gonçalves,
32–39. M. Clémancey, J. Santolini, E. Guittet, G. Blondin, J.-M.
58 (a) N. Saxena, N. Maio, D. R. Crooks, C. J. Ricketts, Y. Yang, Latour and C. Bouton, Redox control of the human iron–
M.-H. Wei, T. W. M. Fan, A. N. Lane, C. Sourbier, A. Singh, sulfur repair protein mitoNEET activity via its iron–sulfur
J. K. Killian, P. S. Meltzer, C. D. Vocke, T. A. Rouault and cluster, J. Biol. Chem., 2016, 291, 7583–7593.
W. M. Linehan, SDHB-deficient cancers: the role of muta- 68 G. L. Taminelli, V. Sotomayor, A. G. Valdivieso, M. L.
tions that impair iron sulfur cluster delivery, J. Natl. Cancer Teiber, M. A. C. Marın and T. A. Santa-Coloma, CISD1

24 | Metallomics, 2018, 10, 9--29 This journal is © The Royal Society of Chemistry 2018
View Article Online

Metallomics Minireview

codifies a mitochondrial protein upregulated by the essential iron–sulfur cluster domain, Nucleic Acids Res.,
CFTR channel, Biochem. Biophys. Res. Commun., 2008, 2012, 40, 7821–7830; (b) D. Netz, C. Stith, M. Stümpfig,
365, 856–862. G. Köpf, D. Vogel and H. e. a. Genau, Eukaryotic DNA
69 K. Okatsu, S.-i. Iemura, F. Koyano, E. Go, M. Kimura, polymerases require an iron–sulfur cluster for the for-
T. Natsume, K. Tanaka and N. Matsuda, Mitochondrial mation of active complexes, Nat. Chem. Biol., 2011, 8,
hexokinase HKI is a novel substrate of the Parkin ubiquitin 125–132.
ligase, Biochem. Biophys. Res. Commun., 2012, 428, 79 A. H. S. de Oliveira, A. E. da Silva, I. M. de Oliveira, J. A. P.
197–202. Henriques and L. F. Agnez-Lima, MutY-glycosylase: an
70 (a) A. F. Salem, D. Whitaker-Menezes, A. Howell, F. Sotgia overview on mutagenesis and activities beyond the GO
and M. P. Lisanti, Mitochondrial biogenesis in epithelial system, Mutat. Res., 2014, 769, 119–131.
cancer cells promotes breast cancer tumor growth and 80 F. Mazzei, A. Viel and M. Bignami, Role of MUTYH in
confers autophagy resistance, Cell Cycle, 2012, 11, human cancer, Mutat. Res., 2013, 743-744, 33–43.
4174–4180; (b) F. Bai, F. Morcos, Y.-S. Sohn, M. Darash- 81 A. A. Out, C. M. J. Tops, M. Nielsen, M. M. Weiss,
Yahana, C. O. Rezende, C. H. Lipper, M. L. Paddock, I. J. H. M. van Minderhout, I. F. A. C. Fokkema, M.-P.
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

L. Song, Y. Luo, S. H. Holt, S. Tamir, E. A. Theodorakis, Buisine, K. Claes, C. Colas, R. Fodde, F. Fostira, P. F.
P. A. Jennings, J. N. Onuchica, R. Mittler and Franken, M. Gaustadnes, K. Heinimann, S. V. Hodgson,
R. Nechushtai, The Fe–S cluster-containing NEET proteins F. B. L. Hogervorst, E. Holinski-Feder, K. Lagerstedt-
mitoNEET and NAF-1 as chemotherapeutic targets in Robinson, S. Olschwang, A. M. W. van den Ouweland,
breast cancer, Proc. Natl. Acad. Sci. U. S. A., 2015, 112, E. J. W. Redeker, R. J. Scott, B. Vankeirsbilck, R. V.
3698–3703. Grønlund, J. T. Wijnen, F. P. Wikman, S. Aretz, J. R.
71 Y.-S. Sohn, S. Tamir, L. Song, D. Michaeli, I. Matouk, Sampson, P. Devilee, J. T. den Dunnen and F. J. Hes,
A. R. Conlan, Y. Harir, S. H. Holt, V. Shulaev, M. L. Leiden open variation database of the MUTYH gene,
Paddock, A. Hochberg, I. Z. Cabanchick, J. N. Onuchic, Hum. Mutat., 2010, 31, 1205–1215.
P. A. Jennings, R. Nechushtaia and R. Mittler, NAF-1 and 82 M. K. Brinkmeyer and S. S. David, Distinct functional
mitoNEET are central to human breast cancer proliferation consequences of MUTYH variants associated with color-
by maintaining mitochondrial homeostasis and promot- ectal cancer: damaged DNA affinity, glycosylase activity
ing tumor growth, Proc. Natl. Acad. Sci. U. S. A., 2013, 110, and interaction with PCNA and Hus1, DNA Repair, 2015,
14676–14681. 34, 39–51.
72 V. D. Paul and R. Lill, Biogenesis of cytosolic and nuclear 83 (a) A. N. Suhasini and R. M. Brosh, Disease-causing
iron–sulfur proteins and their role in genome stability, missense mutations in human DNA helicase disorders,
Biochim. Biophys. Acta, 2015, 1853, 1528–1539. Mutat. Res., 2013, 752, 138–152; (b) A. N. Suhasini and
73 M. Nakamura, D. M. Buzas, A. Kato, M. Fujita, N. Kurata R. M. Brosh, DNA helicases asociated with genetic
and T. Kinoshita, The role of Arabidopsis thaliana NAR1, instability, cancer, and aging, Adv. Exp. Med. Biol., 2013,
a cytosolic iron–sulfur cluster assembly component, in 973, 123–144.
gametophytic gene expression and oxidative stress responses 84 H. Walden and A. J. Deans, The Fanconi anemia DNA
in vegetative tissue, New Phytol., 2013, 199, 925–935. repair pathway: structural and functional insights into a
74 M. V. Corbin, D. A. P. Rockx, A. B. Oostra, H. Joenje and complex disorder, Annu. Rev. Biophys., 2014, 43, 257–278.
J. C. Dorsman, The iron–sulfur cluster assembly network 85 S. K. Bharti, J. A. Sommers, F. George, J. Kuper, F. Hamon,
component NARFL is a key element in the cellular defense K. Shin-ya, M. P. Teulade-Fichou, C. Kisker and
against oxidative stress, Free Radical Biol. Med., 2015, 89, R. M. Brosh, Specialization among iron–sulfur cluster
863–872. helicases to resolve G-quadruplex DNA structures that
75 (a) K. Gari, A. M. L. Ortiz, V. Borel, H. Flynn, J. M. Skehel threaten genomic stability, J. Biol. Chem., 2013, 288,
and S. J. Boulton, MMS19 links cytoplasmic iron–sulfur 28217–28229.
cluster assembly to DNA metabolism, Science, 2012, 337, 86 (a) S. B. Cantor and S. Guillemette, Hereditary breast
243–245; (b) O. Stehling, A. A. Vashisht, J. Mascarenhas, cancer and the BRCA1-associated FANCJ/BACH1/BRIP1,
Z. O. Jonsson, T. Sharma, D. J. A. Netz, A. J. Pierik, Future Oncol., 2011, 7, 253–261; (b) T. Rafnar, D. F.
J. A. Wohlschlegel and R. Lill, MMS19 assembles iron– Gudbjartsson, P. Sulem, A. Jonasdottir, A. Sigurdsson,
sulfur proteins required for DNA metabolism and genomic A. Jonasdottir, S. Besenbacher, P. Lundin, S. N. Stacey,
integrity, Science, 2012, 337, 195–199. J. Gudmundsson, O. T. Magnusson, L. le Roux,
76 J. Rudolf, V. Makrantoni, W. Ingledew, M. Stark and G. Orlygsdottir, H. T. Helgadottir, H. Johannsdottir,
M. White, The DNA repair helicases XPD and FancJ A. Gylfason, L. Tryggvadottir, J. G. Jonasson, A. de Juan,
have essential iron–sulfur domains, Mol. Cell, 2006, 23, E. Ortega, J. M. Ramon-Cajal, M. D. Garcı́a-Prats,
801–808. C. Mayordomo, A. Panadero, F. Rivera, K. K. H. Aben,
77 R. Service, Science, 2014, 346, 1284–1287. A. M. van Altena, L. F. A. G. Massuger, M. Aavikko, P. M.
78 (a) S. Pokharel and J. Campbell, Cross talk between the Kujala, S. Staff, L. A. Aaltonen, K. Olafsdottir, J. Bjornsson,
nuclease and helicase activities of Dna2: role of an A. Kong, A. Salvarsdottir, H. Saemundsson, K. Olafsson,

This journal is © The Royal Society of Chemistry 2018 Metallomics, 2018, 10, 9--29 | 25
View Article Online

Minireview Metallomics

K. R. Benediktsdottir, J. Gulcher, G. Masson, 96 (a) B. J. Ballew, M. Yeager, K. Jacobs, N. Giri, J. Boland,


L. A. Kiemeney, J. I. Mayordomo, U. Thorsteinsdottir and L. Burdett, B. P. Alter and S. A. Savage, Germline mutations
K. Stefansson, Mutations in BRIP1 confer high risk of of regulator of telomere elongation helicase 1, RTEL1, in
ovarian cancer, Nat. Genet., 2011, 43, 1104–1107. Dyskeratosis congenita, Hum. Genet., 2013, 132, 473–480;
87 R. Gupta, S. Sharma, K. M. Doherty, J. A. Sommers, (b) Amanda J. Walne, T. Vulliamy, M. Kirwan, V. Plagnol
S. B. Cantor and R. M. Brosh, Inhibition of BACH1 (FANCJ) and I. Dokal, Constitutional mutations in RTEL1 cause
helicase by backbone discontinuity is overcome by severe dyskeratosis congenita, Am. J. Hum. Genet., 2013, 92,
increased motor ATPase or length of loading strand, 448–453.
Nucleic Acids Res., 2006, 34, 6673–6683. 97 (a) Z. Deng, G. Glousker, A. Molczan, A. J. Fox, N. Lamm,
88 J. A. Sommers, N. Rawtani, R. Gupta, D. V. Bugreev, J. Dheekollu, O.-E. Weizman, M. Schertzer, Z. Wang,
A. V. Mazin, S. B. Cantor and R. M. Brosh, FANCJ uses O. Vladimirova, J. Schug, M. Aker, A. Londoño-Vallejo,
its motor ATPase to destabilize protein–DNA complexes, K. H. Kaestner, P. M. Lieberman and Y. Tzfati, Inherited
unwind triplexes, and inhibit RAD51 strand exchange, mutations in the helicase RTEL1 cause telomere dysfunc-
J. Biol. Chem., 2009, 284, 7505–7517. tion and Hoyeraal–Hreidarsson syndrome, Proc. Natl. Acad.
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

89 (a) O. Levran, C. Attwooll, R. T. Henry, K. L. Milton, Sci. U. S. A., 2013, 110, E3408–E3416; (b) T. Le Guen,
K. Neveling, P. Rio, S. D. Batish, R. Kalb, E. Velleuer, L. Jullien, F. Touzot, M. Schertzer, L. Gaillard,
S. Barral, J. Ott, J. Petrini, D. Schindler, H. Hanenberg M. Perderiset, W. Carpentier, P. Nitschke, C. Picard,
and A. D. Auerbach, The BRCA1-interacting helicase BRIP1 G. Couillault, J. Soulier, A. Fischer, I. Callebaut, N. Jabado,
is deficient in Fanconi anemia, Nat. Genet., 2005, 37, A. Londono-Vallejo, J. P. de Villartay and P. Revy, Human
931–933; (b) Y. Wu, J. A. Sommers, A. N. Suhasini, RTEL1 deficiency causes Hoyeraal-Hreidarsson syndrome
T. Leonard, J. S. Deakyne, A. V. Mazin, K. Shin-ya, with short telomeres and genome instability, Hum. Mol.
H. Kitao and R. M. Brosh, Fanconi anemia group J muta- Genet., 2013, 22, 3239–3249.
tion abolishes its DNA repair function by uncoupling DNA 98 D. J. A. Netz, C. M. Stith, M. Stümpfig, G. Köpf, D. Vogel,
translocation from helicase activity or disruption of pro- H. M. Genau, J. L. Stodola, R. Lill, P. M. J. Burgers and
tein–DNA complexes, Blood, 2010, 116, 3780–3791. A. J. Pierik, Eukaryotic DNA polymerases require an iron–
90 (a) L. Fan, J. O. Fuss, Q. J. Cheng, A. S. Arvai, M. Hammel, sulfur cluster for the formation of active complexes, Nat.
V. A. Roberts, P. K. Cooper and J. A. Tainer, XPD helicase Chem. Biol., 2011, 8, 125–132.
structures and activities: insights into the cancer and aging 99 T. N. Moiseeva, A. M. Gamper, B. L. Hood, T. P. Conrads
phenotypes from XPD mutations, Cell, 2008, 133, 789–800; and C. J. Bakkenist, Human DNA polymerase e is phos-
(b) J. J. DiGiovanna and K. H. Kraemer, Shining a light on phorylated at serine-1940 after DNA damage and interacts
xeroderma pigmentosum, J. Invest. Dermatol., 2012, 132, with the iron–sulfur complex chaperones CIAO1 and
785–796. MMS19, DNA Repair, 2016, 43, 9–17.
91 Y. Wu, J. A. Sommers, I. Khan, J. P. de Winter, J. Brosh and 100 (a) E. Heitzer and I. Tomlinson, Replicative DNA polymerase
M. Robert, Biochemical characterization of Warsaw break- mutations in cancer, Curr. Opin. Genet. Dev., 2014, 24,
age syndrome helicase, J. Biol. Chem., 2011, 287, 1007–1021. 107–113; (b) D. P. Kane and P. V. Shcherbakova, A common
92 S. K. Bharti, I. Khan, T. Banerjee, J. A. Sommers, Y. Wu and cancer-associated DNA polymerase e mutation causes an
R. M. Brosh, Molecular functions and cellular roles of the exceptionally strong mutator phenotype, indicating fidelity
ChlR1 (DDX11) helicase defective in the rare cohesinopa- defects distinct from loss of proofreading, Cancer Res.,
thy Warsaw breakage syndrome, Cell. Mol. Life Sci., 2014, 2014, 74, 1895–1901; (c) E. Nicolas, E. A. Golemis and
71, 2625–2639. S. Arora, POLD1: central mediator of DNA replication
93 P. van der Lelij, K. H. Chrzanowska, B. C. Godthelp, M. A. and repair, and implication in cancer and other patholo-
Rooimans, A. B. Oostra, M. Stumm, M. Z. Zdzienicka, gies, Gene, 2016, 590, 128–141; (d) D. N. Church,
H. Joenje and J. P. de Winter, Warsaw breakage syndrome, S. E. W. Briggs, C. Palles, E. Domingo, S. J. Kearsey,
a cohesinopathy associated with mutations in the XPD J. M. Grimes, M. Gorman, L. Martin, K. M. Howarth,
helicase family member DDX11/ChlR1, Am J. Hum. Genet., S. V. Hodgson, K. Kaur, J. Taylor and I. P. M. Tomlinson,
2010, 86, 262–266. DNA polymerase e and d exonuclease domain mutations
94 J.-M. Capo-Chichi, S. K. Bharti, J. A. Sommers, in endometrial cancer, Hum. Mol. Genet., 2013, 22,
T. Yammine, E. Chouery, L. Patry, G. A. Rouleau, 2820–2828.
M. E. Samuels, F. F. Hamdan, J. L. Michaud, R. M. Brosh 101 T. Fujiwara and H. Harigae, Pathophysiology and genetic
Jr, A. Mégarbane and Z. Kibar, Identification and bio- mutations in congenital sideroblastic anemia, Pediatr. Int.,
chemical characterization of a novel mutation in DDX11 2013, 55, 675–679.
causing Warsaw breakage syndrome, Hum. Mutat., 2013, 102 M. S. Protasova, A. P. Grigorenko, T. V. Tyazhelova, T. V.
34, 103–107. Andreeva, D. A. Reshetov, F. E. Gusev, A. E. Laptenko,
95 J.-B. Vannier, G. Sarek and S. J. Boulton, RTEL1: functions I. L. Kuznetsova, A. Y. Goltsov, S. A. Klyushnikov, S. N.
of a disease-associated helicase, Trends Cell Biol., 2014, 24, Illarioshkin and E. I. Rogaev, Whole-genome sequencing
416–425. identifies a novel ABCB7 gene mutation for X-linked

26 | Metallomics, 2018, 10, 9--29 This journal is © The Royal Society of Chemistry 2018
View Article Online

Metallomics Minireview

congenital cerebellar ataxia in a large family of Mongolian A. May, S. S. Bottomley, D. W. Swinkels, K. Markianos,
ancestry, Eur. J. Hum. Genet., 2015, 24, 550–555. E. A. Craig and M. D. Fleming, Congenital sideroblastic
103 K. Sato, Y. Torimoto, T. Hosoki, K. Ikuta, H. Takahashi, anemia due to mutations in the mitochondrial HSP70
M. Yamamoto, S. Ito, N. Okamura, K. Ichiki, H. Tanaka, homologue HSPA9, Blood, 2015, 126, 2734–2738.
M. Shindo, K. Hirai, Y. Mizukami, T. Otake, M. Fujiya, 111 (a) P. J. Urrutia, N. P. Mena and M. T. Nunez, The interplay
K. Sasaki and Y. Kohgo, Loss of ABCB7 gene: pathogenesis between iron accumulation, mitochondrial dysfunction,
of mitochondrial iron accumulation in erythroblasts in and inflammation during the execution step of neuro-
refractory anemia with ringed sideroblast with isodicentric degenerative disorders, Front. Pharmacol., 2014, 5, 38;
(X)(q13), Int. J. Hematol., 2011, 93, 311–318. (b) N. P. Mena, P. J. Urrutia, F. Lourido, C. M. Carrasco
104 (a) C. Camaschella, A. Campanella, L. De Falco, and M. T. Núñez, Mitochondrial iron homeostasis and its
L. Boschetto, R. Merlini, L. Silvestri, S. Levi and dysfunctions in neurodegenerative disorders, Mitochondrion,
A. Iolascon, The human counterpart of zebrafish shiraz 2015, 21, 92–105.
shows sideroblastic-like microcytic anemia and iron over- 112 Y. Muñoz, C. M. Carrasco, J. D. Campos, P. Aguirre and
load, Blood, 2007, 110, 1353–1358; (b) R. A. Wingert, M. T. Núñez, Parkinson’s disease: the mitochondria-iron
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

J. L. Galloway, B. Barut, H. Foott, P. Fraenkel, J. L. Axe, link, Parkinson’s Dis., 2016, 1–21.
G. J. Weber, K. Dooley, A. J. Davidson, B. Schmid, 113 (a) F. Hefti, W. F. Goure, J. Jerecic, K. S. Iverson,
B. H. Paw, G. C. Shaw, P. Kingsley, J. Palis, H. Schubert, P. A. Walicke and G. A. Krafft, The case for soluble Ab
O. Chen, J. Kaplan and L. I. Zon, Deficiency of glutaredoxin oligomers as a drug target in Alzheimer’s disease, Trends
5 reveals Fe–S clusters are required for vertebrate haem Pharmacol. Sci., 2013, 34, 261–266; (b) X.-M. Gu, H.-C.
synthesis, Nature, 2005, 436, 1035–1039; (c) H. Ye, Huang and Z.-F. Jiang, Mitochondrial dysfunction and
S. Y. Jeong, M. C. Ghosh, G. Kovtunovych, L. Silvestri, cellular metabolic deficiency in Alzheimer’s disease,
D. Ortillo, N. Uchida, J. Tisdale, C. Camaschella and Neurosci. Bull., 2012, 28, 631–640.
T. A. Rouault, Glutaredoxin 5 deficiency causes sideroblas- 114 C. Guo, P. Wang, M.-L. Zhong, T. Wang, X.-S. Huang,
tic anemia by specifically impairing heme biosynthesis J.-Y. Li and Z.-Y. Wang, Deferoxamine inhibits iron
and depleting cytosolic iron in human erythroblasts, induced hippocampal tau phosphorylation in the
J. Clin. Invest., 2010, 120, 1749–1761. Alzheimer transgenic mouse brain, Neurochem. Int., 2013,
105 G. Liu, S. Guo, G. J. Anderson, C. Camaschella, B. Han and 62, 165–172.
G. Nie, Heterozygous missense mutations in the GLRX5 115 P. Lei, S. Ayton, D. I. Finkelstein, L. Spoerri, G. D.
gene cause sideroblastic anemia in a Chinese patient, Ciccotosto, D. K. Wright, B. X. W. Wong, P. A. Adlard,
Blood, 2014, 124, 2750–2751. R. A. Cherny, L. Q. Lam, B. R. Roberts, I. Volitakis,
106 G. Liu, Y. Wang, G. J. Anderson, C. Camaschella, Y. Chang G. F. Egan, C. A. McLean, R. Cappai, J. A. Duce and
and G. Nie, Functional analysis of GLRX5 mutants reveals A. I. Bush, Tau deficiency induces parkinsonism with
distinct functionalities of GLRX5 protein, J. Cell. Biochem., dementia by impairing APP-mediated iron export, Nat.
2016, 117, 207–217. Med., 2012, 18, 291–295.
107 C. Johansson, A. K. Roos, S. J. Montano, R. Sengupta, 116 (a) A. L. Lumsden, T. L. Henshall, S. Dayan, M. T. Lardelli
P. Filippakopoulos, K. Guo, F. von Delft, A. Holmgren, and R. I. Richards, Huntingtin-deficient zebrafish exhibit
U. Oppermann and K. L. Kavanagh, The crystal structure of defects in iron utilization and development, Hum. Mol.
human GLRX5: iron–sulfur cluster co-ordination, tetra- Genet., 2007, 16, 1905–1920; (b) P. Hilditch-Maguire,
meric assembly and monomer activity, Biochem. J., 2011, F. Trettel, L. A. Passani, A. Auerbach, F. Persichetti and
433, 303–311. M. E. MacDonald, Huntingtin: an iron-regulated protein
108 R. A. Wingert, J. L. Galloway, B. Barut, H. Foott, P. Fraenkel, essential for normal nuclear and perinuclear organelles,
J. L. Axe, G. J. Weber, K. Dooley, A. J. Davidson, B. Schmidt, Hum. Mol. Genet., 2000, 9, 2789–2797.
B. H. Paw, G. C. Shaw, P. Kingsley, J. Palis, H. Schubert, 117 A. J. Lees, J. Hardy and T. Revesz, Parkinson’s disease,
O. Chen, J. Kaplan, C. The Tübingen Screen and L. I. Zon, Lancet, 2009, 373, 2055–2066.
Deficiency of glutaredoxin 5 reveals Fe–S clusters are 118 (a) D. Berg and H. Hochstrasser, Iron metabolism in
required for vertebrate haem synthesis, Nature, 2005, 436, Parkinsonian syndromes, Mov. Disord., 2006, 21, 1299–1310;
1035–1039. (b) A. E. Oakley, J. F. Collingwood, J. Dobson, G. Love,
109 Y. Shan and G. Cortopassi, Mitochondrial Hspa9/Mortalin H. R. Perrott and J. A. Edwardson, et al., Individual dopami-
regulates erythroid differentiation via iron–sulfur cluster nergic neurons show raised iron levels in Parkinson disease,
assembly, Mitochondrion, 2016, 26, 94–103. Neurology, 2007, 68, 1820–1825.
110 K. Schmitz-Abe, S. J. Ciesielski, P. J. Schmidt, D. R. 119 (a) P. G. Mastroberardino, E. K. Hoffman, M. P. Horowitz,
Campagna, F. Rahimov, B. A. Schilke, M. Cuijpers, R. Betarbet, G. Taylor and D. Cheng, et al., A novel
K. Rieneck, B. Lausen, M. L. Linenberger, A. K. Sendamarai, transferrin/TfR2-mediated mitochondrial iron transport
C. Guo, I. Hofmann, P. E. Newburger, D. Matthews, system is disrupted in Parkinson’s disease, Neurobiol.
A. Shimamura, P. J. L. M. Snijders, M. C. Towne, C. M. Dis., 2009, 34, 417–431; (b) S. L. Rhodes, D. D. Buchanan,
Niemeyer, H. G. Watson, M. H. Dziegiel, M. M. Heeney, I. Ahmed, K. D. Taylor, M.-A. Loriot, J. S. Sinsheimer,

This journal is © The Royal Society of Chemistry 2018 Metallomics, 2018, 10, 9--29 | 27
View Article Online

Minireview Metallomics

J. M. Bronstein, A. Elbaz, G. D. Mellick, J. I. Rotter and 129 Y. Wang, N. B. Langer, G. C. Shaw, G. Yang, L. Li, J. Kaplan,
B. Ritz, Pooled analysis of iron-related genes in Parkinson’s B. H. Paw and J. R. Bloomer, Abnormal mitoferrin-1
disease: association with transferrin, Neurobiol. Dis., 2014, expression in patients with erythropoietic protoporphyria,
62, 172–178. Exp. Hematol., 2011, 39, 784–794.
120 M. P. Horowitz and J. T. Greenamyre, Mitochondrial iron 130 (a) W. Chen, H. A. Dailey and B. H. Paw, Ferrochelatase
metabolism and its role in neurodegeneration, forms an oligomeric complex with mitoferrin-1 and
J. Alzheimer’s Dis., 2010, 20, 551–568. Abcb10 for erythroid heme biosynthesis, Blood, 2010, 116,
121 D. W. Lee, D. Kaur, S. J. Chinta, S. Rajagopalan and 628–630; (b) M. Yamamoto, H. Arimura, T. Fukushige,
J. K. Andersen, A disruption in iron–sulfur center biogen- K. Minami, Y. Nishizawa, A. Tanimoto, T. Kanekura,
esis via inhibition of mitochondrial dithiol glutaredoxin 2 M. Nakagawa, S. I. Akiyama and T. Furukawa, Abcb10 Role
may contribute to mitochondrial and cellular iron dysre- in Heme Biosynthesis In Vivo: Abcb10 Knockout in Mice
gulation in mammalian glutathione-depleted dopaminer- Causes Anemia with Protoporphyrin IX and Iron Accumu-
gic cells: implications for Parkinson’s disease, Antioxid. lation, Mol. Cell. Biol., 2014, 34, 1077–1084; (c) M. B.
Redox Signaling, 2009, 11, 2083–2094. Troadec, D. Warner, J. Wallace, K. Thomas, G. J. Spangrude,
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

122 (a) M. Katsuyama, K. Iwata, M. Ibi, K. Matsuno, J. Phillips, O. Khalimonchuk, B. H. Paw, D. M. Ward and
M. Matsumoto and C. Yabe-Nishimura, Clioquinol induces J. Kaplan, Targeted deletion of the mouse Mitoferrin1
DNA double-strand breaks, activation of ATM, and subse- gene: from anemia to protoporphyria, Blood, 2011, 117,
quent activation of p53 signaling, Toxicology, 2012, 299, 5494–5502.
55–59; (b) D. Devos, C. Moreau, J. C. Devedjian, J. Kluza, 131 H. Beinert, Iron–sulfur proteins: ancient structures, still
M. Petrault, C. Laloux, A. Jonneaux, G. Ryckewaert, full of surprises, J. Biol. Inorg. Chem., 2000, 5, 2–15.
G. Garçon, N. Rouaix, A. Duhamel, P. Jissendi, 132 (a) M. A. Kohanski, D. J. Dwyer and J. J. Collins, How
K. Dujardin, F. Auger, L. Ravasi, L. Hopes, G. Grolez, antibiotics kill bacteria: from targets to networks, Nat. Rev.
W. Firdaus, B. Sablonnière, I. Strubi-Vuillaume, N. Zahr, Microbiol., 2010, 423–435; (b) M. A. Kohanski, D. J. Dwyer,
A. Destée, J.-C. Corvol, D. Pöltl, M. Leist, C. Rose, B. Hayete, C. A. Lawrence and J. J. Collins, A common
L. Defebvre, P. Marchetti, Z. I. Cabantchik and R. Bordet, mechanism of cellular death induced by bactericidal anti-
Targeting chelatable iron as a therapeutic modality in biotics, Cell, 2007, 130, 797–810; (c) M. A. Kohanski,
Parkinson’s disease, Antioxid. Redox Signaling, 2014, 21, D. J. Dwyer, J. Wierzbowski, G. Cottarel and J. J. Collins,
195–210. Mistranslation of membrane proteins and two-component
123 D. P. Barupala, S. P. Dzul, P. J. Riggs-Gelasco and T. L. system activation trigger antibiotic-mediated cell death,
Stemmler, Synthesis, delivery and regulation of eukaryotic Cell, 2008, 135, 679–690.
heme and Fe–S cluster cofactors, Arch. Biochem. Biophys., 133 (a) Y. S. Choi, D. H. Shin, I. Y. Chung, S. H. Kim,
2016, 592, 60–75. Y. J. Heo and Y. H. Cho, Identification of Pseudomonas
124 M. Lecha, H. Puy and J.-C. Deybach, Erythropoietic proto- aeruginosa genes crucial for hydrogen peroxide resistance,
porphyria, Orphanet J. Rare Dis., 2009, 4, 19. J. Microbiol. Biotechnol., 2007, 17, 1344–1352; (b) S. H. Kim,
125 H. Nakano, A. Nakano, Y. Toyomaki, S. Ohashi, K. Harada, B. Y. Lee, G. W. Lau and Y. H. Cho, IscR modulates catalase
R. Moritsugu, H. Takeda, A. Kawada, Y. Mitsuhashi and A (KatA) activity, peroxide resistance, and full virulence of
K. Hanada, Novel ferrochelatase mutations in Japanese Pseudomonas aeruginosa PA14, J. Microbiol. Biotechnol.,
patients with erythropoietic protoporphyria: high fre- 2009, 19, 1520–1526.
quency of the splice site modulator IVS3-48C polymorph- 134 (a) R. P. Morris, L. Nguyen, J. Gatfield, K. Visconti,
ism in the Japanese population, J. Invest. Dermatol., 2006, K. Nguyen, D. Schnappinger, S. Ehrt, Y. Liu, L. Heifets,
126, 2717–2719. J. Pieters, G. Schoolnik and C. J. Thompson, Ancestral
126 X.-F. Kong, J. Ye, D.-Y. Gao, Q.-M. Gong, D.-H. Zhang, antibiotic resistance in Mycobacterium tuberculosis,
Z.-M. Lu, Y.-M. Lu and X.-X. Zhang, Identification of a Proc. Natl. Acad. Sci. U. S. A., 2005, 102, 12200–12205;
ferrochelatase mutation in a Chinese family with erythro- (b) J. Rybniker, A. Nowag, E. Van Gumpel, N. Nissen,
poietic protoporphyria, J. Hepatol., 2008, 48, 375–379. N. Robinson, G. Plum and P. Hartmann, Insights into
127 M. Balwani and D. Doheny, Loss-of-function ferrochelatase the function of the WhiB-like protein of mycobacterioph-
and gain-of-function erythroid-specific 5-aminolevulinate age TM4, a transcriptional inhibitor of WhiB2, Mol. Micro-
synthase mutations causing erythropoietic protoporphyria biol., 2010, 77, 642–657; (c) V. Saini, A. Farhana and
and X-linked protoporphyria in North American patients A. J. Steyn, Mycobacterium tuberculosis WhiB3: a novel
reveal novel mutations and a high prevalence of X-linked iron–sulfur cluster protein that regulates redox homeo-
protoporphyria, Mol. Med., 2013, 19, 1. stasis and virulence, Antioxid. Redox Signaling, 2012, 16,
128 M. S. Farrag, J. Kucerova, L. Šlachtova, O. Šeda, J. Šperl and 687–697.
P. Martasek, A novel mutation in the FECH Gene in a 135 A. G. Rocha and A. Dancis, Life without Fe–S clusters, Mol.
Czech family with erythropoietic protoporphyria and a Microbiol., 2016, 99, 821–826.
population study of IVS3-48C variant contributing to the 136 (a) D. J. A. Netz, A. J. Pierik, M. Stumpfig, E. Bill,
disease, Folia Biol., 2015, 61, 227–232. A. K. Sharma, L. J. Pallesen, W. E. Walden and R. Lill,

28 | Metallomics, 2018, 10, 9--29 This journal is © The Royal Society of Chemistry 2018
View Article Online

Metallomics Minireview

A bridging [4Fe–4S] cluster and nucleotide binding are highly conserved Fe–S containing Dre2 C-terminus is
essential for function of the Cfd1–Nbp35 complex as a essential for yeast viability, Mol. Microbiol., 2011, 82,
scaffold in iron–sulfur protein maturation, J. Biol. Chem., 54–67; (c) L. Vernis, C. Facca, E. Delagoutte, N. Soler,
2012, 287, 12365–12378; (b) N. Soler, E. Delagoutte, R. Chanet, B. Guiard, G. Faye and G. Baldacci, A newly
S. Miron, C. Facca, D. Baı̈lle, B. d’Autreaux, G. Craescu, identified essential complex, Dre2-Tah18, controls mito-
Y.-M. Frapart, D. Mansuy, G. Baldacci, M.-E. Huang and chondria integrity and cell death after oxidative stress in
L. Vernis, Interaction between the reductase Tah18 and yeast, PLoS One, 2009, 4, e4376.
Published on 26 September 2017. Downloaded on 9/19/2020 2:50:26 AM.

This journal is © The Royal Society of Chemistry 2018 Metallomics, 2018, 10, 9--29 | 29

You might also like