biochimica et biophysica acta - connecting repositories · 2017. 1. 1. · quences or similar...

12
Review Assembly of Fe/S proteins in bacterial systems Biochemistry of the bacterial ISC system B. Blanc, C. Gerez, S. Ollagnier de Choudens Université Grenoble Alpes, LCBM, 38054 Grenoble, France CEA, DSV, iRTSV, LCBM, Biocatalyse, 38054 Grenoble, France CNRS UMR5249, LCBM, 38054 Grenoble, France abstract article info Article history: Received 20 August 2014 Received in revised form 20 November 2014 Accepted 8 December 2014 Available online 13 December 2014 Keywords: Ironsulfur Fe/S assembly Fe/S transfer Frataxin Protein complex Proteinprotein interaction Iron/sulfur clusters are key cofactors in proteins involved in a large number of conserved cellular processes, in- cluding gene expression, DNA replication and repair, ribosome biogenesis, tRNA modication, central metabo- lism and respiration. Fe/S proteins can perform a wide range of functions, from electron transfer to redox and non-redox catalysis. In all living organisms, Fe/S proteins are rst synthesized in an apo-form. However, as the Fe/S prosthetic group is required for correct folding and/or protein stability, Fe/S clusters are inserted co- translationally or immediately after translation by specic assembly machineries. These systems have been extensively studied over the last decade, both in prokaryotes and eukaryotes. The present review covers the basic principles of the bacterial housekeeping Fe/S biogenesis ISC system, and related recent molecular advances. Some of the most exciting recent highlights relating to this system include structural and functional characteri- zation of binary and ternary complexes involved in Fe/S cluster formation on the scaffold protein IscU. These ad- vances enhance our understanding of the Fe/S cluster assembly mechanism by revealing essential interactions that could never be determined with isolated proteins and likely are closer to an in vivo situation. Much less is currently known about the molecular mechanism of the Fe/S transfer step, but a brief account of the proteinpro- tein interactions involved is given. This article is part of a Special Issue entitled: Fe/S proteins: Analysis, structure, function, biogenesis and diseases. © 2014 Elsevier B.V. All rights reserved. 1. Introduction The function of many proteins depends on the presence of cofactors. Ironsulfur clusters (Fe/S) are one of the most ancient and versatile in- organic cofactors in biology. These clusters confer a number of functions to proteins ranging from electron transfer to catalysis and regulatory processes (14). Ironsulfur clusters are present in mostly all living or- ganisms where they are formed from iron ions (either ferrous (Fe 2+ ) or ferric (Fe 3+ ) depending on cluster type) and inorganic sulde (S 2). The clusters mainly assemble under rhombic [2Fe2S] and cubic [4Fe4S] forms (1). In most cases, the cluster is bound to the protein by sulfur atoms provided by cysteine residues, although there are increasing ex- amples of nitrogen coordinationprovided by histidine or arginine res- iduesand oxygen coordinationfrom aspartate or tyrosine. Examples of coordination by exogenous ligands, such as water molecules, enzyme substrates or cofactors have also been observed (2). Genetic, biochemi- cal and cell-biology studies in the 1990s provided strong evidence, that the maturation of Fe/S proteins in living cells is a controlled process in- volving several complex protein machineries. These studies identied three different systems involved in the biogenesis of bacterial Fe/S pro- teins: the NIF systemwhich is responsible for the specic maturation of nitrogenase in azototrophic bacteria (5,6)and the ISC and SUF sys- tems, which generate housekeeping Fe/S proteins under normal and stress conditions, respectively (7,8) (Fig. 1). Genomic analyses revealed that the number and type of operons coding for these systems vary from one microorganism to another. Some contain all systems, others two or only one, and some only contain a part of one system (810). All these systems share the same basic principles and similar key molecular ac- tors which work together to produce Fe/S clusters. The basic premise of cluster assembly is beautifully simple: First, a cysteine desulfurase (NifS, IscS or SufSE) makes it possible to use L-cysteine as a stable and safe source of sulfur atoms. This sulfur can then be combined with iron (whose origin is unclear) to assemble clusters within a scaffold protein (NifU, IscU, SufB), or cluster factory. This corresponds to the de novo Fe/S cluster assembly step. Finally, the transfer step occurs, when the Fe/S cluster is transferred to recipient proteins. This involves energy-dependent proteins (HscAB, SufCD) and Fe/S carrier proteins (IscA, SufA, etc.). As there are many more Fe/S proteins than it would be possible to have scaffold proteins and Fe/S carriers in a cell, these elements must have evolved to enable efcient cluster delivery to Biochimica et Biophysica Acta 1853 (2015) 14361447 Abbreviations: Ferredoxin:, Fdx; glutaredoxin:, Grx This article is part of a Special Issue entitled: Fe/S proteins: Analysis, structure, func- tion, biogenesis and diseases. Corresponding author. Tel.: +33 4 38 78 91 22; fax: +33 4 38 78 91 24. E-mail address: [email protected] (S. Ollagnier de Choudens). http://dx.doi.org/10.1016/j.bbamcr.2014.12.009 0167-4889/© 2014 Elsevier B.V. All rights reserved. Contents lists available at ScienceDirect Biochimica et Biophysica Acta journal homepage: www.elsevier.com/locate/bbamcr

Upload: others

Post on 08-Feb-2021

0 views

Category:

Documents


0 download

TRANSCRIPT

  • Biochimica et Biophysica Acta 1853 (2015) 1436–1447

    Contents lists available at ScienceDirect

    Biochimica et Biophysica Acta

    j ourna l homepage: www.e lsev ie r .com/ locate /bbamcr

    Review

    Assembly of Fe/S proteins in bacterial systems☆

    Biochemistry of the bacterial ISC system

    B. Blanc, C. Gerez, S. Ollagnier de Choudens ⁎Université Grenoble Alpes, LCBM, 38054 Grenoble, FranceCEA, DSV, iRTSV, LCBM, Biocatalyse, 38054 Grenoble, FranceCNRS UMR5249, LCBM, 38054 Grenoble, France

    Abbreviations: Ferredoxin:, Fdx; glutaredoxin:, Grx☆ This article is part of a Special Issue entitled: Fe/S protion, biogenesis and diseases.⁎ Corresponding author. Tel.: +33 4 38 78 91 22; fax: +

    E-mail address: [email protected] (S. Ollagnier de Chou

    http://dx.doi.org/10.1016/j.bbamcr.2014.12.0090167-4889/© 2014 Elsevier B.V. All rights reserved.

    a b s t r a c t

    a r t i c l e i n f o

    Article history:Received 20 August 2014Received in revised form 20 November 2014Accepted 8 December 2014Available online 13 December 2014

    Keywords:Iron–sulfurFe/S assemblyFe/S transferFrataxinProtein complexProtein–protein interaction

    Iron/sulfur clusters are key cofactors in proteins involved in a large number of conserved cellular processes, in-cluding gene expression, DNA replication and repair, ribosome biogenesis, tRNA modification, central metabo-lism and respiration. Fe/S proteins can perform a wide range of functions, from electron transfer to redox andnon-redox catalysis. In all living organisms, Fe/S proteins are first synthesized in an apo-form. However, as theFe/S prosthetic group is required for correct folding and/or protein stability, Fe/S clusters are inserted co-translationally or immediately after translation by specific assembly machineries. These systems have beenextensively studied over the last decade, both in prokaryotes and eukaryotes. The present review covers thebasic principles of the bacterial housekeeping Fe/S biogenesis ISC system, and related recentmolecular advances.Some of the most exciting recent highlights relating to this system include structural and functional characteri-zation of binary and ternary complexes involved in Fe/S cluster formation on the scaffold protein IscU. These ad-vances enhance our understanding of the Fe/S cluster assembly mechanism by revealing essential interactionsthat could never be determined with isolated proteins and likely are closer to an in vivo situation. Much less iscurrently known about themolecularmechanismof the Fe/S transfer step, but a brief account of the protein–pro-tein interactions involved is given. This article is part of a Special Issue entitled: Fe/S proteins: Analysis, structure,function, biogenesis and diseases.

    © 2014 Elsevier B.V. All rights reserved.

    1. Introduction

    The function of many proteins depends on the presence of cofactors.Iron–sulfur clusters (Fe/S) are one of the most ancient and versatile in-organic cofactors in biology. These clusters confer a number of functionsto proteins ranging from electron transfer to catalysis and regulatoryprocesses (1–4). Iron–sulfur clusters are present in mostly all living or-ganisms where they are formed from iron ions (either ferrous (Fe2+) orferric (Fe3+) depending on cluster type) and inorganic sulfide (S2−).The clusters mainly assemble under rhombic [2Fe–2S] and cubic [4Fe–4S] forms (1). Inmost cases, the cluster is bound to the protein by sulfuratoms provided by cysteine residues, although there are increasing ex-amples of nitrogen coordination—provided by histidine or arginine res-idues—and oxygen coordination—from aspartate or tyrosine. Examplesof coordination by exogenous ligands, such aswatermolecules, enzymesubstrates or cofactors have also been observed (2). Genetic, biochemi-cal and cell-biology studies in the 1990s provided strong evidence, that

    teins: Analysis, structure, func-

    33 4 38 78 91 24.dens).

    the maturation of Fe/S proteins in living cells is a controlled process in-volving several complex protein machineries. These studies identifiedthree different systems involved in the biogenesis of bacterial Fe/S pro-teins: the NIF system—which is responsible for the specific maturationof nitrogenase in azototrophic bacteria (5,6)—and the ISC and SUF sys-tems, which generate housekeeping Fe/S proteins under normal andstress conditions, respectively (7,8) (Fig. 1). Genomic analyses revealedthat the number and type of operons coding for these systems vary fromonemicroorganism to another. Some contain all systems, others two oronly one, and some only contain a part of one system (8–10). All thesesystems share the same basic principles and similar key molecular ac-tors which work together to produce Fe/S clusters. The basic premiseof cluster assembly is beautifully simple: First, a cysteine desulfurase(NifS, IscS or SufSE) makes it possible to use L-cysteine as a stable andsafe source of sulfur atoms. This sulfur can then be combined withiron (whose origin is unclear) to assemble clusters within a scaffoldprotein (NifU, IscU, SufB), or ‘cluster factory’. This corresponds to thede novo Fe/S cluster assembly step. Finally, the transfer step occurs,when the Fe/S cluster is transferred to recipient proteins. This involvesenergy-dependent proteins (HscAB, SufCD) and Fe/S carrier proteins(IscA, SufA, etc.). As there are many more Fe/S proteins than it wouldbe possible to have scaffold proteins and Fe/S carriers in a cell, theseelements must have evolved to enable efficient cluster delivery to

    http://crossmark.crossref.org/dialog/?doi=10.1016/j.bbamcr.2014.12.009&domain=pdfhttp://dx.doi.org/10.1016/j.bbamcr.2014.12.009mailto:[email protected]://dx.doi.org/10.1016/j.bbamcr.2014.12.009http://www.sciencedirect.com/science/journal/01674889www.elsevier.com/locate/bbamcr

  • iscR iscS iscU iscA hscB hscA fdx iscX

    sufA sufB sufC sufD sufEsufS

    ISC

    SUF

    iscAnif nifUNIF

    nifS cysE1

    Fig. 1. Various systems involved in Fe/S assembly in bacteria and comparison of their genetic organization in operons: NIF, ISC and SUF (5,7,8). Genes or regions having homologous se-quences or similar functions between the three systems are color-coded. Different colors within nifU indicate different domains within this modular protein.

    1437B. Blanc et al. / Biochimica et Biophysica Acta 1853 (2015) 1436–1447

    numerous different apoproteins. In this review, we focus on the bacteri-al ISC machinery, with particular emphasis on its biochemical proper-ties. We first provide a brief schematic view of the ISC proteins andthe roles they play in the Fe/S assembly process. We go into detail onwhat is currently known about each protein in the ISC system, whichhas been greatly extended by moving beyond the characterization ofsingle isolated proteins. Thus, particular attention is paid to the contri-bution of studies performedonmore integrated systems,where ISCpro-teins are present in a complex, a situation which likely resembles anin vivo situation. For readers interested in the bacterial SUF machinery,excellent and comprehensive reviews are available in the same series.

    2. Basic principles of Fe/S assembly by the ISC system

    The main steps leading to Fe/S formation and the ISC proteins re-sponsible for each of them are summarized in Fig. 2 (the steps are re-ferred to by bold-face numbers). (1) Sulfur source. The IscS enzymemakes it possible to use L-cysteine as a stable and safe source of sulfuratoms. The sulfur is transiently bound, in the form of a persulfide, toan active-site cysteine in IscS. It is then transferred to a cysteine residueon the scaffold IscU. (2) Iron source. Due to the cellular toxicity of free

    Fig. 2. Schematic view of Fe/S biogenesis in the ISC system. Ovals represent proteins (IscS, IscU(1)–(4) correspond to the assembly step; step (5) is the transfer step in which Fe/S cluster is tIscA Fe/S transporter. Balls in green represent sulfur atoms; balls in violet represent iron, with

    iron, a specific iron donor or carrier is required. This is thought to beCyaY and/or IscX, and/or IscA. (3) Electron source. The rules of chemis-try make it impossible to produce an Fe/S cluster from a persulfide spe-cies (S0) and ferrous iron (Fe2+)without providing additional electrons.Ferredoxin (Fdx) is thought to fulfill this function. (4) Scaffold proteinIscU. This is the platform where the Fe/S cluster is transiently formed.(5) Cluster transfer. Fe/S transfer from the scaffold IscU to apo-targetsinvolves HscA and HscB chaperone proteins, which, through ATP-dependent activation, promote a rapid and controlled transfer. Forsome specific apo-targets, the Fe/S carrier IscA protein is also requiredfor protein maturation.

    While the overall mechanism of Fe/S biogenesis by the ISC systemhas been known for a number of years, the detailed molecular mecha-nisms for each step ((1)–(5)) (e.g. persulfide reduction, iron entry,intermediates involved in Fe/S cluster assembly on IscU, Fe/S clusternuclearity, Fe/S coordination, protein–protein interactions, conforma-tional changes, electron transfer, transfer mechanism, etc.) remain tobe identified. In the next paragraphs we review the most recentlyacquired molecular information relating to ISC proteins, both free andin complex, and discuss how this contributes to the elucidation of Fe/Sassembly and transfer mechanisms.

    , IscA and targets) and do not take into account oligomerization states of proteins. Stepsransferred directly from IscU to targets with the help of chaperones or indirectly throughboth [2Fe–2S] and [4Fe–4S] represented on IscU and on targets.

  • 1438 B. Blanc et al. / Biochimica et Biophysica Acta 1853 (2015) 1436–1447

    3. Function and biochemistry of isolated ISC proteins

    3.1. The cysteine desulfurase, IscS

    IscS is a widely conserved 45-kDa protein which exists in solution asa homodimer containing one pyridoxal-phosphate (PLP) molecule permonomer (11). Through a well-characterized PLP-dependent mecha-nism, it transforms L-cysteine into alanine and sulfur which is storedas a persulfide ion on a conserved cysteine residue within the proteinitself (Cys328 in Escherichia coli) (12,13). The crystal structure of theIscS dimerwas solved. This structure revealed that the catalytic cysteineresidue is in a partially ordered loop (14,15) extending away from theactive site where the PLP cofactor and bound cysteine substrate arefound. This “open conformation” is likely to play a crucial role in sulfurtransfer to the IscU scaffold protein during Fe/S assembly. Interestingly,NifS and SufS cysteine desulfurases have shorter analogous loops whichprefer a “closed conformation” (15). Thus, in these proteins the catalyticcysteine residue is located in close proximity to PLP. This structure iscompatible with direct sulfur loading from the bound cysteine sub-strate. A possible explanation for this significant difference betweenclosely related systems is that IscS provides sulfur atoms which notonly are used in Fe/S clusters, but also for the synthesis of a large num-ber of organic sulfurated cofactors such as thiamine, tRNA,molybdenumcofactor, etc. (15). The longer Cys328-containing loop found in IscS maythus be essential for transfer of the sulfur produced to its multipleacceptors.

    3.2. The scaffold protein, IscU

    A scaffold function for IscU was provided from both in vivo andin vitro experiments (16–18). The scaffold is thought to receive sulfurand iron and to be able to transiently assemble an Fe/S cluster. As-purified, IscU from E. coli and Azotobacter vinelandii exist in a dimericapo-state which makes it impossible to determine the nature of theirFe/S cluster in vivo. However, under in vitro anaerobic conditions theIscU apo-form can be loaded with iron and sulfur to generate a holo-form. This form exists mainly as a dimer in solution (16). ReconstitutedIscU binds and forms Fe/S clusters within hours in a sequential manner:one [2Fe–2S]/dimer, two [2Fe–2S]/dimer, and finally one [4Fe–4S]/dimer. The [4Fe–4S] is converted back to [2Fe–2S] upon exposure tooxygen, thus there is a dynamic equilibrium between the [2Fe–2S]and [4Fe–4S] cluster-bound forms of IscU (16). Various pieces of evi-dence have led authors to consider the 1x [2Fe–2S]/dimer as the physi-ological form. First, the X-ray structure of Fe/S-bound IscU from Aquifexaeolicus reveals the presence of an [2Fe–2S] cluster (19). Second, therate of in vitro Fe/S transfer from the 1x [2Fe–2S] form of IscU to apo-proteins is dramatically enhanced when chaperones are present,whereas the rate of transfer from the 2x [2Fe–2S] form or from the[4Fe–4S] form is chaperone-independent and slower (20,21). Third,one of the clusters in the 2x [2Fe–2S] form is unstable in the presenceof the iron chelator EDTA (16). Finally, when the invariant aspartate res-idue (D39 in A. vinelandii, D40 in Thermotoga maritima)—which hasbeen shown to perform an important function in cluster delivery—ischanged to alanine, the corresponding protein contains an [2Fe–2S]cluster (21,22). For Fe/S cluster coordination, sequence comparisoncombined with spectroscopic and structural analyses suggests that, inaddition to the conserved cysteine residues (Cys37, Cys63 and Cys106in E. coli), a histidine residue (His105 in E. coli) is necessary. However,the structure solved for IscU in which the histidine plays the part of ametal ligand corresponds to a zinc-bound monomeric form of IscU(23) rather than a dimeric Fe/S-bound one. In addition, the histidineresidue is only semi-conserved; thus there remain some doubts as towhether this residue is truly a ligand for the cluster. Further informationon IscU was gleaned from recent studies, where this protein was foundto populate two interconvertible conformational states in solution, adisordered state (D) under its apo-form, and a structured state

    (S) under its Fe/S-form (24). This suggests that conformational changestake place during Fe/S building and transfer.

    As described in Section 4.1, some of the information relating to IscUobtained on the isolated protein in solution should not be taken at face-value. Indeed, in complex with IscS, IscU exists as a monomer ratherthan a dimer, and this could considerably change its biochemical, struc-tural and functional properties.

    3.3. The bacterial frataxin homologue, CyaY

    Genetic and bioinformatic studies performed on eukaryotes re-vealed a direct role for frataxin in the Fe/S metabolism and Fe/S assem-bly process involving ISC proteins (25–28). In particular, phylogeneticstudies showed that the frataxin gene and its orthologs, includingcyaY, co-occur with the chaperone hscAB genes (25). However, the bac-teria frataxin gene, cyaY, does not belong to the isc operon, and neitherE. coli nor Salmonella enterica single cyaYmutants showed a drastic lossin Fe/S enzyme activities (29,30). Nevertheless, recently direct evidenceof the involvement of bacterial frataxin in Fe/Smetabolismwas found inBacillus subtilis. In this microorganism, inactivation of fra, the frataxinhomologue gene, leads to a decrease in Fe/S protein activity (31). Thisis supported also by studies in Salmonella typhimurium where frataxinis proposed to play a role as an iron chaperone in Fe/S cluster repair(32).

    CyaY was initially suggested to be an iron donor due to its in vitrobehavior toward iron. Thus, CyaY was shown to have a medium tolow affinity for ferrous iron (Kd = 3–55 μM range) and a high affinityfor ferric iron (Kass = 1.1017 M−1). This affinity is mediated by interac-tionwith a conserved acidic ridge on the protein (33–35).Whereas apo-frataxin is a monomer (36), frataxin has been shown to form oligomersin the presence of iron in vitro, strengthening the hypothesis that it per-forms an irondonor function (37–39). As further evidence of its involve-ment in Fe/S cluster biogenesis, other in vitro experiments showed thatoligomeric iron-loaded CyaY could provide iron for Fe/S cluster forma-tion on IscU (33,37). The link between iron and frataxin in the Fe/S as-sembly was also observed in eukaryotes, for example yeast frataxininteracts in an iron-dependent manner with the cysteine desulfuraseNfs1 and the scaffold Isu1 (40). In addition, iron-loaded human andyeast frataxin can both provide iron for Fe/S cluster formation onhuman ISCU (33). However, the hypothesis that an iron-loaded oligo-meric form of frataxin plays a physiological role has been questionedin the aftermath of some in vivo experiments in eukaryotes. In theseexperiments, deletion of yeast frataxin was shown to be complementedby amutant frataxin with defects in iron-induced oligomerization (41);in addition, themature form of human frataxin (FXN81–210), which doesnot oligomerize (42), restored cellular viability in frataxin-deleted mu-rine fibroblasts (43). Obviously, frataxin might be functional withoutoligomerization, and as described in Section 4.1, this was recently con-firmed by experiments on complexes containing frataxin.

    The possibility that CyaY might act as a negative regulator of Fe/Sformation, rather than as an iron donor, has recently emerged both inprokaryotes and eukaryotes. This opens up new possibilities as to howfrataxin might be involved in Fe/S cluster formation (44,45). Thispoint is discussed in more detail in Section 4.1.

    3.4. IscX

    IscX, also known as ORF3 or YfhJ, is a small acidicmonomeric proteincomposed of 66 amino acids. Its function is unclear since deletion of theiscX gene, like cyaY, does not lead to any specific phenotype related toFe/S metabolism, except a small decrease in the IscR activity (11,46).The structure of IscX in solution revealed an affinity for iron cationsand the presence of a modified winged helix motif. This type of motifis commonly present in DNA-binding proteins (47,48). Biochemical as-sessment showed that IscX binds Fe2+ much more avidly than Fe3+,suggesting that it could act as an iron donor during Fe/S cluster

  • 1439B. Blanc et al. / Biochimica et Biophysica Acta 1853 (2015) 1436–1447

    assembly (49). However, the most recent results available indicate thatIscX inhibits Fe/S formation on IscU without affecting the cysteinedesulfurase activity of free IscS (49). These observations strongly sug-gested that IscX, rather than being a transcriptional regulator as mightbe inferred from its structure, is a molecular adaptor, and that likeCyaY it can modulate iron–sulfur cluster formation. Recent protein in-teraction studies involving IscX are discussed in Section 4.1.

    3.5. [2Fe–2S] ferredoxin

    Ferredoxins are small globular Fe–S cluster-containing proteinsknown to be involved in various electron transfer processes in bacteria,mammals and plants (50–53). The ferredoxin (Fdx) coded by the isc op-eron is a monomeric 12-kDa protein which contains a stable [2Fe–2S]cluster. The structure of Fdx has been solved (54). Fdx is crucial in Fe/S assembly, since deletion of the fdx gene in an E. coli strain affects theactivities of Fe/S enzymes to a similar extent as iscS or iscU mutations(17). From genetic studies on yeast using labeled 55Fe, it has been sug-gested that ferredoxin (Yah1p in yeast) is involved in the first step ofFe/S biogenesis, namely Fe/S cluster building on IscU (55). It has beenspeculated that Fdx could perform the persulfide reduction step onNfs1 during the initial stages of Fe/S cluster assembly (55). In vitro,this function is generally performed by various exogenously-added re-ducing agents, such as dithiothreitol (DTT) or Tris-2-carboxyethyl-phosphine (TCEP). An alternative, but not mutually exclusive, role forFdx could be an involvement in reductive coupling of two [2Fe–2S]2+

    clusters to form [4Fe–4S]2+ clusters on the IscU scaffold or on anotherprotein such as IscA (see Section 3.7). This is supported by in vitro stud-ies showing that ferredoxin donates the electrons necessary for partialreductive coupling of two [2Fe–2S]2+ clusters to form a single [4Fe–4S]2+ cluster on the homodimeric IscU scaffold protein fromA. vinelandii (56). A dual role for Fdx both in the Fe/S assembly stepand in the Fe/S transfer step is also suggested by pull-down experi-ments. These revealed an interaction between Fdx and IscS (Fe/S assem-bly step), as well as between Fdx and HscA and Fdx and IscA (Fe/Stransfer step) (46,57). As described in Section 4.1 a) andb), a recentmo-lecular investigation of partner interaction studies has shed further lighton Fdx's function.

    3.6. The chaperone system, HscA–HscB

    Numerous genetic and biochemical studies have established an es-sential contribution of HscA and HscB to Fe/S maturation (17,58,59).The substrates for the HscA–HscB system are ATP and the scaffoldIscU. HscA, which has an ATPase activity (58), is known to recognize aspecific LPPVK sequence motif on IscU, and its interaction is regulatedby the co-chaperoneHscBwhich interactswith IscU through hydropho-bic residues (60–63). However, the specific role played byHscA–HscB inthe Fe/S assembly process has been controversial, with conflictingreports from in vitro and in vivo studies. Thus, a role in stabilizing clus-ters assembled on IscU was inferred from in vitro studies (64), whilein vivo studies based on 55Fe immuno-precipitation (65) indicated thatHscA–HscB facilitates cluster transfer from IscU to apo-targets. Luckily,this confusionwas overcome by in vitro studies in E. coliwhich providedevidence for a role in facilitating cluster transfer from IscU to acceptorproteins through an ATP-dependent mechanism (66). Interestingly, inthese studies the HscA–HscB system was found to enhance the rate of[2Fe–2S] cluster transfer from the IscU dimer, but to have no effect onthe rate of [4Fe–4S] cluster transfer from IscU dimer to aconitase (thisis also mentioned in Section 3.3 dealing with IscU). These observationssuggest that there exists a specific conformation of IscU [2Fe–2S] ableto interact with chaperones. If this in vitro specificity for [2Fe–2S] vs.[4Fe–4S] clusters truly reflects the physiological form of IscU clusters,it is a very interesting point.

    Based on recent in vitro studies, two mechanisms have been pro-posed for Fe/S cluster release from free IscU by the HscA–HscB system.

    Both of these which involve an ATP-Fe/S-bound HscA–HscB–IscU com-plex are presented Fig. 5 and discussed in Section 4.2.

    3.7. IscA

    IscA is a small 12-kDa protein belonging to the A-type protein familywhich also includes SufA and ErpA proteins (67). The precise role of IscAin Fe/S assembly has not yet been clearly identified as an iscA mutantstrain has no specific phenotype, and in vitro results concerning itsmetallic center have been variable as all experiments performed aredifferent. The key point is what is the right experiment? meaningwhich results are really addressing how the process works in vivo?.Thus, it is currently unclear whether IscAmetallic center is an Fe/S clus-ter or a mononuclear iron. Moreover, crystallographic studies of E. coliapo-IscA have shown that the protein can exist as a dimer or tetramerand that the three conserved cysteine residues form a ‘cysteine pocket’that could accommodate either a mononuclear iron atom or an Fe/Scluster (68,69). Recent in vitro advances on IscA suggest that both me-tallic sites may exist and therefore may play separate roles in Fe/Sbiogenesis.

    3.7.1. IscA is an Fe/S-binding proteinInitially, in vitro reconstitution of the apo form of bacterial IscA with

    Fe2+ and sodium sulfite or Fe3+ and L-Cys/IscS under anaerobic condi-tions demonstrated the formation of [2Fe–2S] or [4Fe–4S] clusters withcomplete cysteinyl ligation, as characterized by various spectroscopicmethods (57,70). The holo-form of IscA is a dimer, and quantitation ofthe iron and sulfur it contains indicates that there is one cluster perdimer. An unusual feature of IscA is that it contains only three conservedcysteines, it has therefore been suggested that the Fe/S cluster (of eithertype) bridges the gap between two subunits (70). The [2Fe–2S] and[4Fe–4S] forms of NifIscA were recently further characterized, and the[4Fe–4S] cluster was shown to be very sensitive to oxygen and to rapid-ly convert to [2Fe–2S] (71). This conversion process is reversible in vitroas adding DTT caused the [4Fe–4S] cluster to re-form. Fe/S-loading onIscA was also demonstrated in vivo on as-isolated proteins. Thesecontained an [2Fe–2S] (72) or an [4Fe–4S] cluster (73), thus bacterialIscA is undoubtedly an Fe/S-binding protein.

    3.7.2. IscA is an Fe-binding proteinWhen incubated in the presence of oxygen with Fe2+/DTT or the

    thioredoxin reductase system, E. coli IscA was shown to bind one ironatom per homodimer with an association constant of 1.1019 M−1 (74,75). A similar strong affinity for iron has been observed for other bacte-rial IscA, including that from A. vinelandii (NifIscA) (76) andA. ferrooxidans (77) as well as for IscA homologues from humans (78)and yeast (79). For E. coli IscA, mutation of any of its three conservedcysteine residues to serine producedmutant proteinswith a diminishediron-binding capacity (80). An in-depth spectroscopic study on NifIscArevealed that it can bind one Fe2+ or one Fe3+ atom per dimer (76).The ferric iron is most likely 5-coordinatedwith two or three cysteinateligands, whereas the ferrous iron site is ligated with three cysteinate li-gands and one or two oxygenic ligands. One of these oxygenic ligandsmight be a tyrosine as recent results on E. coli IscA, showed that theY40F mutation greatly diminished its iron-binding capacity (81). Theferric and ferrous forms of IscA can interconvert, and L-cysteine is effec-tive in mediating release of Fe2+ from the Fe3+ form (76,81). Finally,both IscA iron forms can mediate Fe/S cluster assembly on IscU in thepresence of L-cysteine and IscS. In vivo evidence for IscA's Fe-bindingcapacity was demonstrated with IscA isolated from E. coli cells grownunder aerobic conditions. This protein displays the same biochemicaland spectroscopic properties as those observed for the in vitro Fe-loaded IscA (80,81). However, IscA isolated under these conditions con-tains very little amount of iron (0.1 iron/monomer).

    Depending on whether IscA was characterized as an Fe/S- or Fe-binding protein, different roles have been proposed for it in the Fe/S

  • 1440 B. Blanc et al. / Biochimica et Biophysica Acta 1853 (2015) 1436–1447

    assembly process: it could be an alternative Fe/S scaffold under aerobicor oxidative stress conditions; or an Fe/S carrier between scaffold IscUand target proteins; or a metallochaperone for Fe delivery to IscU or toacceptor proteins facilitating in situ assembly of [4Fe–4S] fromdegraded[2Fe–2S] clusters.

    3.7.3. Proposed functions for IscA proteinWhen IscA was initially found to be an Fe/S cluster protein, it was

    believed to act as an alternative scaffold (57,70,72,82) since it wasshown to transfer its Fe/S cluster to Fe/S proteins (57,82). This functionwas excluded from genetic and biochemical experiments (17,67), andIscA was rather proposed as an Fe/S carrier which receives an Fe/S clus-ter from the IscU scaffold and delivers it to apo-target proteins (67,83,84). This would place IscA in the trafficking/delivery step (like SufAand ErpA A-type proteins). Support for this Fe/S-carrier function ofIscAwas recently provided by in vitro and genetic studies (67). Interest-ingly, recent in vivo experiments hinted that IscA could fulfill this Fe/S-carrier function for the specific maturation of [4Fe–4S] proteins such asaconitase, ThiC, IlvD (85), formate dehydrogenase N, nitrate reductase(Nar) and small subunits of hydrogen-oxidizing hydrogenases (86,87).If IscA is an [2Fe–2S] protein, its cluster would need to be reductivelycoupled to generate an [4Fe–4S] cluster prior to transfer to these apo-target proteins. The reductive equivalents could be provided by ferre-doxin which has been shown to interact with IscA (57).

    In vivo and in vitro experiments for iron-binding by IscA suggest thatIscAmay act as an iron chaperone for the biogenesis of iron–sulfur clus-ters in E. coli cells under aerobic conditions (74,80). Recently, it wasshown that mutation of the tyrosine residue at position 40 into phenyl-alanine (Y40F) produces an inactive IscA protein unable to complementan iscA mutant in vivo. However, a key control showing that IscAY40Fprotein accumulates in the “complemented” strains was not done,preventing to conclude on the role of this residue and therefore on therole of IscA as an Fe-chaperone.

    Bacterial cells over-expressing IscA analyzed by Mössbauer spec-troscopy could beused to determinewhether iron and/or an Fe/S clusteris bound to IscA under physiological conditions. This would help to dis-criminate between its proposed functions as a Fe-chaperone and/or anFe/S carrier. In themeantime, these apparentlymutually exclusive func-tions of IscA could be reconciled as suggested by Johnson et al., by sup-posing that both [2Fe–2S]-IscA and Fe3+-bound IscA coexist in vivo andare both implicated in the maturation or repair of [4Fe–4S] proteinsunder oxygen or oxidative stress conditions (Fig. 3). In an Fe-loadedform, IscA can provide iron for (i) Fe/S cluster assembly on IscU when

    Fig. 3. Possible roles for IscA in Fe/S assembly and repair. In the Fe-loaded form, IscA provides irrepair from a cysteine persulfide-ligated [2Fe–2S] cluster (76,120). As an [2Fe–2S] carrier protecluster (71,72).

    incubated with L-cysteine, IscS, and electrons under anaerobic condi-tions (76,81) or (ii) [4Fe–4S] repair from a cysteine persulfide-ligated[2Fe–2S] cluster (88) as recently suggested for E. coli RIC protein (89).As an [2Fe–2S] carrier protein, IscA could be involved in [4Fe–4S] pro-tein maturation after reductive coupling of its [2Fe–2S] cluster.

    3.8. The regulator, IscR

    IscR is an [2Fe–2S] transcriptional regulator controlling the expres-sion ofmore than 40 genes in E. coli, including those encoding for anaer-obic respiratory Fe/S enzymes and proteins involved in Fe/S formationsuch as ErpA, NfuA and the SUF system (90). IscR contains three con-served cysteine residues (92, 98, 104 in E. coli), which are required forFe/S ligation. Recently, a histidine residue (His107 in E. coli) was pro-posed to act as the fourth ligand (91). The presence of an [2Fe–2S]-con-taining protein was demonstrated by in vitro and in vivo analyses (91,92). IscR maturation can occur in the total absence of Fe/S carrier pro-teins, i.e., IscR can obtain its Fe/S cluster directly from scaffold proteinsas it plays a central role in Fe/S homeostasis (92). Mutagenic analyseshave revealed the existence of two classes of IscR-binding sites onDNA: these are known as Type 1 and Type 2 sites (90). Holo-IscR (the[2Fe–2S]-bound IscR) was shown to bind the Type 1 site with a higheraffinity than apo-IscR, whereas the holo- and apo-IscR bind with similaraffinity to the Type 2 site (93). Holo-IscR represses its own expression aswell as that of the remainder of the isc operon by binding to Type 1 sites,while under its apo-form, IscR activates expression of the suf operonwhich has a Type 2 DNA-binding site. The most recent advances withregard to IscR were provided by in vivo studies, giving important in-sights into how repression of the isc operon by [2Fe–2S]-IscR couldhelp cells to maintain homeostasis by regulating the synthesis of en-zymes involved in the ISC Fe/S biogenesis pathway (94).

    4. ISC protein complexes and Fe/S cluster biogenesis

    4.1. Complexes relating to the de novo assembly step

    Over the last couple of years, structural and functional characteriza-tion of binary (IscS–IscU, IscS–CyaY, IscS–IscX and IscS–Fdx) and ternary(IscS–IscU–CyaY, IscS–IscU–IscX and IscS–IscU–Fdx) complexes havesignificantly increased what we know about the early steps in the Fe/Scluster biogenesis mechanism, namely Fe/S cluster assembly on IscU.

    on for (i) Fe/S cluster assembly on IscUwith L-cysteine, IscS, and electrons or (ii) [4Fe–4S]in, IscA is involved in [4Fe–4S] protein maturation after reductive coupling of its [2Fe–2S]

  • 1441B. Blanc et al. / Biochimica et Biophysica Acta 1853 (2015) 1436–1447

    4.1.1. Binary complexes

    4.1.1.1. The IscS–IscU complex. An IscS–IscU complex from E. coliwas iso-lated in solution (95) and Isothermal calorimetry (ITC) experimentsshowed that IscU interacts with IscS in a 1:1 stoichiometry; a Kd valueof 1.3 μM (96) was determined. Two X-ray structures of the IscS–IscUcomplexwere solved, one in an apo-form, the other with an Fe/S clusterbound to IscU (15,97). Both of these structures showed that the com-plex is not formed by association between a dimer of IscS and a dimerof IscU; rather each IscU monomer binds near the C-terminal portionof the IscS dimer, forming a very elongated S-shaped complex measur-ing 150 Å long and 65 Å wide (15). Complex formation is associatedwith minor changes in the conformation of the IscS dimer, predomi-nantly of surface side chains. The IscU protein projects its conservedcysteine residues toward the loop on IscS where the catalytic Cys328 isfound. In themodel, the distance between Cys328 on IscS and any cyste-ine on IscU is greater than 12 Å, suggesting that a hugemovement of theloop must occur to allow successful sulfur transfer from the catalyticIscS cysteine to IscU cysteines. This range of movement is possiblesince in the Archeoglobus fulfidus complex, in which IscUD35A stabilizesan [2Fe–2S] cluster, the catalytic cysteine of IscS is a ligand of the IscUFe/S cluster, together with the three conserved cysteines (97). Interest-ingly, this particular IscS–IscUD35A complex displays no cysteinedesulfurase activity (97). This is probably because A. fulgidus can growin a hydrogen sulfide-rich environment and therefore does not need toenzymatically produce sulfide for Fe/S assembly (98). Thus, the IscS pro-tein in A. fulgidusmight rather provide a ligand for Fe/S cluster coordina-tionwith IscU. In E. coli, IscS displays a cysteine desulfurase activity.Whenit was measured in vitro after mixing IscS with a large excess of IscUcontradictory results were obtained, making it difficult to conclude onthe effect of IscU on the cysteine desulfurase activity of IscS (49,99,100).Indeed, in some cases, a two-fold decrease in activity was observed com-pared to that measured with free IscS (49), while in other cases, IscUwasfound to have no effect on sulfur production (99). The molecular mecha-nisms controlling the inhibitory effect are not known, and further exper-iments using labeled cysteine and as-purified IscS–IscU complex in a 1:1stoichiometry will be necessary to clarify this point, as has been donefor the eukaryotic system (101). A 1:1 complex between the cysteinedesulfurase and the scaffold was also characterized with eukaryotic pro-teins after heterologous co-expression of ISCU, NFS1 and its partnerISD11 in E. coli. This confirmed the physiological relevance of a tight asso-ciation between the desulfurase and the scaffold protein (102).

    4.1.1.2. The IscS–CyaY and IscS–IscX complexes. CyaY and IscX both con-tain a large negatively-charged patch on their surface and behave simi-larly with iron, suggesting that their functions are redundant (48). ITCmade it possible to characterize a first IscS–CyaY complex. This complexdisplays a 1:1 stoichiometry and medium-range affinity between IscSand CyaY proteins was determined (Kd: 18.5 μM) showing that IscShas a lower affinity for CyaY than for IscU. This is supported by thefact that it has been impossible to isolate an IscS–CyaY complex by gelfiltration, whereas it was possible to purify the IscS–IscU complex(95). Ferrous iron neither affects the affinity constant nor IscS–CyaYcomplex formation. An ab initio bead model was constructed based onsmall-angle X-ray scattering (SAXS) data and the X-ray structures ofthe isolated proteins. Like for IscU, CyaY monomers rather than adimer of CyaY interact with IscS. However, in contrast to IscU, theCyaY molecule binds at a site near the IscS dimer interface, and not atthe periphery, leading to a more globular complex (96). In vitro studiesdemonstrated that the IscS residues necessary for interactionwith CyaYare R220, R223 and R225 (96). Negatively-charged residues present onthe acidic ridge of CyaY interact directly with these positively-chargedamino acids on IscS. CyaY binding to IscS does not interfere with sulfurproduction, as even in large excess it only has a moderate effect on itscysteine desulfurase activity (max 1.2-fold decrease) (49,99,103). AnIscS–IscX complex was also characterized in vitro and a structural

    model was built. This model suggests that IscX binds near the dimeriza-tion interface of IscS, like CyaY (15,49). Ferrous iron does not strengthenthe interaction between IscS and IscX. Like for CyaY, IscX does not altersulfur production by IscS in the presence of L-cysteine (49). Thus, theseresults indicate that CyaY and IscX behave similarly in terms of interac-tion with IscS, but as they have no effect on its activity it is difficult todetermine their function based on binary IscS–CyaY and IscS–IscXcomplexes.

    4.1.1.3. The IscS–Fdx complex. An IscS–Fdx complex was recently charac-terized using a combination of nuclear magnetic resonance (NMR),mutagenesis and SAXS experiments (104,105). The results of this char-acterization demonstrated that [2Fe–2S]-Fdx (in either oxidation state)interacts with IscS in a 1:1 stoichiometry, and with an affinity of thesame order of magnitude as that of IscU for IscS (Kd = 1.5 μM) (104,105). The interaction face on Fdx includes residues close to its Fe/S clus-ter (104), suggesting a role for the Fdx Fe/S cluster in IscS catalysis.These studies also showed that IscS and Fdx interact through electro-static interactions involving complementary surfaces of oppositecharges: D70, D71 and D74 on Fdx and R220, R223 and R237 on theIscS protomer. Therefore, IscS–Fdx interaction is very similar to that ofIscS and CyaY or IscX, and a globular complex will be formed in allcases. The importance of the IscS–Fdx interaction for the function ofFdx in the Fe/S assembly process was demonstrated in vivo in a Δfdxstrain using IscR as a reporter for Fe/S protein maturation (105). Inthese experiments, whereas a plasmid carrying wild-type fdx gene cancomplement the mutant strain, a plasmid carrying fdxD70KD74K couldnot. Recently, in vitro experiments showed that the presence of reducedFdxwas sufficient for the IscS enzymatic reaction to occur in the absenceof any additional exogenous reducing agents (such as DTT) (104). Inthese experiments, the electron from the reduced Fdx was shown tobe transferred to IscS only in the presence of an L-cysteine substrate.In contrast, with the IscSC328S variant (in which the catalytic cysteineresidue is mutated) the electron is not transferred. Thus, Fdx appearsto be able to supply one electron to IscS in its persulfide-state onCys328, produced by conversion of cysteine to alanine. However, to re-duce a persulfide two electrons are necessary, and Fdx has not beenshown to transfer two successive electrons to fully reduce the IscSpersulfide. Thus, the role played by Fdx in the reduction of IscSpersulfide remains quite intriguing. To add to the conundrum, it isknown that the persulfide on IscS can be spontaneously transferred toIscU through a transpersulfuration reaction without requiring electrons(106). Therefore, it appears more likely that Fdx plays a role in thereduction of the persulfide on IscU for Fe/S formation. Even though nointeraction between bacterial Fdx and IscU has been observed, a specificinteraction between the reduced form of FDX2 and ISU1 was recentlyobserved in eukaryotes strongly suggesting that FDX2 works with ISU1rather than cysteine desulfurase during Fe/S formation (Muhlenhoff, U.unpublished results). All these results support the idea that Fdx is involvedin the sulfur delivery step but do not exclude that Fdx also has a functionin Fe/S trafficking from the scaffold to apo-targets.

    From all these studies it clearly appears that CyaY, IscX, Fdx and IscUcan interact with IscS to form binary complexes, with all three partners(CyaY, IscX, Fdx) occupying the same binding site. Whereas ITC studiessuggest the following order of affinity for IscS and its partners:IscU ≥ Fdx N CyaY (105), NMR studies appear to suggest an inverseorder of affinity: CyaY N Fdx N IscU (104). Thismakes it difficult to estab-lish a model for sequential interactions of these proteins with IscS, andthus hinders our comprehension of step by step Fe/S cluster assemblyon IscU.

    4.1.2. Ternary complexes

    4.1.2.1. The IscS–IscU–CyaY complex. Pull-down experiments demon-strated that IscU can bind IscS at the same time as CyaY, which stronglysuggests the formation of an IscS–IscU–CyaY complex (15). An IscS–

  • 1442 B. Blanc et al. / Biochimica et Biophysica Acta 1853 (2015) 1436–1447

    IscU–CyaY complexwas also recently captured through chemical cross-linking experiments (104). Using SAXS, X-ray structures of the singleproteins and NMR data, a model of a ternary complex containing IscS,IscU and CyaY with a 1:1:1 stoichiometry was developed (96). In thismolecular model, a monomer of CyaY and a monomer of IscU interactwith one subunit of the IscS dimer. In this configuration, CyaY is posi-tioned in a clearly distinct site to that occupied by IscU, even thoughthey are next to each other. Thus, CyaY fits into the cavity delimitedby the IscS dimer interface and the IscU binding site. CyaY locatesclose to the catalytic cysteine loop of IscS, but far from the PLP, in agree-ment with the observation that CyaY does not interfere with IscS activ-ity. In vitro experiments demonstrated that CyaY enhances the affinityof IscU for IscS, with a Kd value for IscS–IscU interaction in the presenceof CyaY that is approximately one order of magnitude lower than thatobserved in its absence (hundred nM vs. μM). When the same experi-ment was performed the other way around, to determine the dissocia-tion constant of IscS–CyaY in the absence or presence of IscU, IscU wasalso found to increase IscS–CyaY affinity (35 nM vs. 23 μM). Thus, thepresence of the third component significantly modulates the affinitiesbetween partners involved in IscS–IscU and IscS–CyaY binary com-plexes, forming a tight ternary complex. The presence of iron has no in-fluence on this ternary complex, either in terms of formation or affinityconstants (96). Curiously, the cysteine desulfurase activity of the terna-ry complex is reduced (by 1.2–2.2-fold) compared to IscS alone (49,99,100), but it is difficult to determine whether this moderate inhibition isrelated to the presence of IscU and/or CyaY due to the contradictory re-sults obtained with the binary IscS–IscU complex, as mentioned above(49,99,100). Nevertheless, it is quite clear that CyaY influences the ki-netics of enzymatic Fe/S assembly on IscU. Indeed, CyaY has an inhibito-ry effect on the Fe/S formation on IscU, particularly by reducing theinitial reaction rate (96,99,103). The nature of the Fe/S clusters on IscU(mixture of [2Fe–2S] and [4Fe–4S]) is unchanged in the presence or ab-sence of CyaY (99) indicating that CyaY does not influence the nature ofthe cluster formed on IscU. However, currently it is difficult to deter-mine the molecular mechanism behind the inhibitory effect of CyaYon Fe/S assembly. It is worth noting that in these experiments investi-gating Fe/S cluster formation on IscU, CyaY, IscU and IscS were mixedin non-stoichiometric amounts, generally using an IscS:IscU:CyaY ratioof 1:50:5. The large excess of IscU could explain the decrease in IscS ac-tivity observed by some groups (49,99). Similarly, the excess of CyaYcould explain the decrease in Fe/S formation. Hence, it is difficult to in-terpret results obtained from these experiments even though proteinsmixed in solution will equilibrate with protein complexes. At present,it is urgent to investigate the biochemical properties (cysteinedesulfurase activity, iron-binding, Fe/S formation and stability, confor-mational changes) of an isolated ternary complex of known stoichiom-etry (IscS–IscU–CyaY)2, as determined by biophysical analyses. Thisshould be possible since this complex can be isolated in a stable form(Ollagnier de Choudens et al. unpublished results). This characterizationwill be extremely useful for understanding the frataxin-dependent in-hibitory effect on Fe/S formation at a molecular level, and to unravelthe general molecular mechanism of Fe/S assembly. This notion to in-vestigate bacterial Fe/S assembly using specific protein complex is par-ticularly important since a complex composed of mammalian cysteinedesulfurase (and its partner ISD11), scaffold and frataxin proteins((NFS1)2-(ISD11)2-(ISCU)2-(FXN)2) was recently isolated after co-expression of the proteins in E. coli. This complex has provided key in-formation helping to unravel the eukaryotic Fe/S assembly processand frataxin function (102). In particular, it showed that frataxin con-trols iron entry into the complex by enhancing cysteine desulfurase ac-tivity. This contrasts with observations in bacteria, where frataxin(CyaY) is found to have no influence on the cysteine desulfurase activityof IscS. This can be explained by the fact that IscS has a significant sulfurproduction activity, in contrast to NFS1-ISD11which displays almost noactivity unless it is activated. Therefore, bacterial frataxin, CyaY, mayplay only a regulatory role with regard to iron mobilization when

    mixed with IscS, IscU, cysteine and iron. By controlling iron entry,fewer, but sufficient, Fe/S clusters could be synthesized compared toin the absence of frataxin.

    Another ternary complex, involving IscS, IscU and IscX, was also re-cently characterized (15,49). The main biochemical properties of thiscomplex are that it has a lower (2-fold lower) cysteine desulfurase ac-tivity than the IscS–IscU complex and a poorer ability to form Fe/S clus-ters on IscU (49). Therefore, IscX appears to repress Fe/S formation onIscU. Like for CyaY, the advantage of this inhibitory effect is not yetunderstood.

    4.1.2.2. The IscS–IscU–Fdx complex. In vitro experiments provided evi-dence that IscU added to IscS-holoFdx does not displace Fdx, and thatIscU does not alter the affinity of the IscS–Fdx complex. This stronglysuggests that a ternary IscS-holoFdx complex forms (105). Superimpo-sition of the HADDOCKmodel of holoFdx-IscS with the crystal structureof the IscU–IscS complex showed that IscS can indeed accommodateboth holoFdx and IscU in a ternary holoFdx–IscS–IscU complex (105).In this model, which provides the first structural information on theholoFdx in complex and its importance for cluster formation, the[2Fe–2S] cluster of Fdx is oriented between the active site cysteineloop of IscS and the cysteine ligands of IscU. The C-terminus of [2Fe–2S]–Fdx, which contains a histidine (His 107) and a tyrosine (Tyr103), points toward the interface between IscS and IscU, and, througha relay mechanism, could assist electron transfer from Fdx to IscS and/or IscU (105). In thismodel, acidic amino acids on Fdx interact with con-served, positively-charged arginine residues on IscS, like in the binaryIscS–Fdx complex. CyaY was shown to displace Fdx from the IscS–Fdxcomplex indicating that Fdx and CyaY compete for the same bindingsites on the IscS–IscU complex (49,105). Future experiments will aimto isolate and structurally and functionally characterize the IscS–IscU–Fdx complex. In particular, it will be interesting to investigate Fdx abilityto reduce the persulfide on the IscU protein within the IscS–IscU–Fdxcomplex since the reaction of the IscS–IscU–Fdx complex with L-cysteine first generates a persulfide on IscS, the terminal sulfur atomof which is directly transferred through transpersulfuration to a cyste-ine residue on IscU, generating another persulfide species. The stabilityand formation of the cluster on IscU formed in the IscS–IscU–Fdx com-plex should also be compared to that produced with the IscS–IscU–CyaY complex, as well as the reductive coupling of [2Fe–2S] clusters toform an [4Fe–4S] cluster on IscU in the presence of reduced Fdx.

    All these studies on protein complexes have increased what weknow about the Fe/S assembly step on IscU, in particular by revealingthe existence of various ternary complexes: IscS–IscU–CyaY, IscS–IscU–Fdx and IscS–IscU–IscX, in which CyaY, IscX and Fdx occupy thesame binding site. One challenge that will need to be addressed in thefuture will be to place each ternary complex in sequence in the land-scape of Fe/S cluster formation on IscU. In addition, it will be interestingto investigate kinetics of Fe/S assembly on IscU using IscS–IscU–CyaYand IscS–IscU–Fdx ternary complexes in combination with Fdx andCyaY respectively. Indeed, in vitro kinetics for Fe/S assembly on IscUmeasured so far using isolated proteins are not reasonable (hours) toexplain an in vivo process (few min.) and one expect a faster processusing complexes. However, in line with the most recent results and ob-servations, two models for Fe/S cluster assembly on IscU can be pro-posed (Fig. 4). Since little information is available for IscX, this laterwas not included in the mechanisms.

    (A) An IscS dimer interactswith IscU to generate an (IscS–IscU)2 com-plex. In the presence of CyaY, a ternary complex (IscS–IscU–CyaY)2 isformed. IscS produces sulfide as persulfide, which is spontaneously trans-ferred to IscU. In themeantime, CyaY controls iron entry into the complexvia conformational changes making the acidic ridge available for irondocking. Then, Fdxdisplaces CyaY to formanew(IscS–IscU–Fdx)2 ternarycomplex. Fdx reduces the persulfides on IscU, allowing Fe/S formationand cluster transfer can then occur. In model (B), a similar (IscS–IscU)2complex is formed by interaction between IscS and IscU. In the presence

  • Fig. 4.Models proposed for Fe/S assembly on IscU. Model (A): frataxin binds the IscS–IscU complex to regulate the iron entry. Cysteine desulfurase produces sulfide from L-cysteine. Thepersulfide generates on IscS is transfered through a transpersulfuration reaction to IscU. Then, Fdx displaces CyaY and reduces persulfide on IscU allowing Fe/S cluster formation. Model(B): frataxin stabilizes Fe/S and regulates the Fe/S transfer step. In this model, there is no regulation of iron entry; reduced Fdx binds the IscS–IscU complex after the iron entry and thecysteine desulfuration reaction allowing persulfide reduction on IscU and Fe/S formation. CyaYdisplaces oxidized Fdx from the IscS–IscU complex protecting Fe/S cluster and/or facilitatingFe/S transfer to targets (49,105).

    1443B. Blanc et al. / Biochimica et Biophysica Acta 1853 (2015) 1436–1447

    of L-cysteine and iron, persulfide formation and iron-binding occur onIscU. Then, Fdx interactswith the binary (IscS–IscU)2 complex to generatean (IscS–IscU–Fdx)2 complex. Fdx reduces thepersulfides on IscU to allowFe/S formation. CyaY displaces Fdx to form a new ternary complex (IscS–IscU–CyaY)2. The role of CyaY in this complex would be to stabilize thecluster, as reported in eukaryotes (102), and/or to induce conformationalchanges of the complex to promote optimal Fe/S transfer. Consistentwitha role of bacterial frataxin, CyaY, in the transfer step, the cyay gene co-occurs with chaperone genes in genomes (25), and deletion of this geneis not essential in bacteria for Fe/S metabolism. This second attractivemodel could thus explain the inhibitory effect of CyaY on Fe/S formation,by reducing access of sulfide and iron to IscU when CyaY is present withIscU, IscS, L-cysteine and iron.

    4.2. Complexes involved in the Fe/S cluster transfer step

    4.2.1. The Fe/S cluster transfer stepOnce the Fe/S cluster has formed on IscU, it is transferred to apo-

    target proteins with the help of the HscA–HscB system. As discussedin Section 3.6, the interactions between IscU and HscA and HscB havebeen characterized (46) and based on recent in vitro studies, twomech-anisms have been proposed for Fe/S cluster release from IscU by theHscA/B system (Fig. 5). The first model (107) (Fig. 5 (left)) involves astructured [2Fe–2S] form (S) and a disordered (D) apo-form of IscU(see Section 3). HscB preferentially binds the structured (S) form ofIscU, whereas HscA alone or in complex with ADP binds and stabilizesthe disordered (D) form of IscU (107). Apo-IscU (D form) is releasedwhen ATP binds HscA, suggesting a mechanism in which transfer of

    the Fe/S cluster is coupled to ATP hydrolysis by HscA. This would con-vert IscU from the (S) state to the (D) state and simultaneously releasethe Fe/S cluster and HscB from IscU. Concomitantly, ATP hydrolysisinduces a transition of HscA from the ATP-bound state (T) to the ADP-bound state (R). In this model, as the cluster is transferred to its target,apo-IscU remains bound to the HscA chaperone until a new ATP mole-cule binds, inducing its release (Fig. 5 (left)). The second model (Fig. 5(right)) is based on the possible existence of two isomers of [2Fe–2S]–IscU2 protein which differ by their affinity for the [2Fe–2S] cluster dueto different metal coordination (108). In this model, HscA stabilizesthe isomer in which the [2Fe–2S] cluster is less tightly bound to IscU(108). The molecular bases for this coordination change are notknown, but it is possible that conformational changes related to theHscA (T) → (R) transition during ATP hydrolysis contribute toconverting IscU to the favorable isomer state for Fe/S transfer in thepresence of HscB (109). In this model, Fe/S cluster transfer occursafter the release of [2Fe–2S]-IscU from the chaperones (Fig. 5 (right)).Whatever the model, it seems likely that the mechanism whereby thechaperones enhance [2Fe–2S] cluster transfer involves conformationalchanges to both the HscA (T/R) and IscU (S/D) proteins, rather thansimply through direct interaction of the chaperones with the cluster.

    However, even though the Fe/S cluster becomes more labile in thepresence of HscA–HscB, it is not released into themediumduring trans-fer to a target. Numerous in vitro experiments have demonstrated thatFe/S transfer is a protected process which can be divided into threesteps: (1) formation of a complex between the Fe/S donor (IscU) andthe Fe/S acceptor (target), with cysteines on the acceptor playing norole in this interaction; (2) a unidirectional and rapid cluster transfer

  • Fig. 5.Models proposed for chaperone-mediated Fe/S transfer from IscU to targets. In the first model (107) (1): First, HscB bound to IscU in its S state targets the ATP-bound form of HscA(T-state) leading to a temporary HscA–HscB–holoIscU complex. The approach of a Fe/S acceptor leads to hydrolysis of ATP and a concomitant conformational change of HscA from the T-state to theADP-bound form (R-state), which stabilizes thedisordered form (D) of IscU. This leads to the release of its Fe/S cluster and ofHscB. Disordered IscU remains bound toHscA afterrelease of ADP, but subsequent binding of ATP changes the conformation of HscA to the T-state, which releases apo-IscU under its D state and a new cycle can be engaged. In the secondmodel (108) (2), there are two isomers of [2Fe–2S]–IscU2 proteinwhich differ by their affinity for the [2Fe–2S] cluster due to differentmetal coordination (109). After ATP hydrolysis, HscBis released from the temporaryHscA–HscB–holoIscU complex and the [2Fe–2S]–IscU2 protein shifts to the isomer inwhich the [2Fe–2S] cluster is less tightly bound to IscU that is stabilizedby ADP bound-HscA or HscA alone. The transition to the ATP-bound state of HscA leads to the release of the [2Fe–2S]–IscU2 protein then able to transfer its Fe/S cluster to the apo-target.

    1444 B. Blanc et al. / Biochimica et Biophysica Acta 1853 (2015) 1436–1447

    based on a cysteine-thiol exchange mechanism between the scaffoldcluster and the free thiols on the target protein; (3) dissociation ofthe complex (82). Even though this proposed mechanism for Fe/Stransfer is likely to be correct, it has a weakness: it is based on exper-iments performed only with the scaffold IscU and a target, not takingchaperones into account which are known to enhance dramaticallyFe/S cluster transfer to targets (20,66). From our current knowledgeof protein–protein interactions (Section 4.1), the Fe/S transfer pro-cess is likely to be much more complex, with an apo-target that in-teracts with IscU-containing binary or ternary complexes. Whetherthe chaperones intervene just before or concomitantly with targetarrival is not known, but they certainly further make the situation

    more complex. So far, no large complexes involved in the Fe/S clustertransfer process have been characterized. The world is thereforewaiting for information on the following complexes: IscS–IscU–HscA–HscB, IscS–IscU–CyaY–HscA–HscB (IscS–IscU–IscX–HscA–HscB) or IscS–IscU–Fdx–HscA–HscB complexes and their equiva-lents with an apo-target.

    In vitro and in vivo experiments have demonstrated that Fe/S clustertransfer from IscU to apo-targets occurs either directly or via Fe/S carrierproteins (IscA in the case of the ISC system) (67,92). The interactionbetween IscA and IscU was characterized in vitro (82), but no interac-tions between IscA and the IscU-containing binary or ternary complexesdescribed above have yet been observed.

  • 1445B. Blanc et al. / Biochimica et Biophysica Acta 1853 (2015) 1436–1447

    Even though they do not belong to the ISC system, other proteins inbacteria have been shown to play an Fe/S shuttle role during Fe/S trans-fer. These include monothiol glutaredoxin (Grx4 and Grx5 in E. coli andA. vinelandii respectively), NfuA and ErpA (83,110). Some laboratories,mainly through genetic approaches, are investigating trafficking ofthese Fe/S carriers so as to be able to determine which carrier gives itscluster to which protein and to gain an overview of the Fe/S transportnetwork in cells (111). Some interesting biochemical datawere recentlyobtained with IscA and Grx.

    4.2.2. A Grx–IscA complexA specific interaction between IscA and glutaredoxin (Grx) was re-

    cently characterized in vitro and in vivo both in bacteria and eukaryotes(112–114). This interaction is considered to be an efficient complex forFe/S transfer. The initial evidence for the role of monothiol glutaredoxinin Fe/S biogenesis came from yeast gene knockout studies, in which dele-tion of the grx5 gene, which codes for mitochondrial Grx, resulted in de-creased maturation of [4Fe–4S] proteins (115). Moreover, radiolabeled55Fe immuno-precipitation studies with grx5 knockout mutantsrevealed that yeast Grx5 is involved in the Fe/S transfer step from IscUto apo-targets (65). Upon purification, monothiol glutaredoxins containbound [2Fe–2S] clusters. These clusters are ligated by the active site cys-teines of two monomers and two GSH molecules (116,117).

    Recently, a ready, unidirectional and complete [2Fe–2S] clustertransfer exchange between Grx and IscA was demonstrated usingA. vinelandii proteins (114). The existence of a Grx–IscA complex andFe/S transfer from Grx to IscA was also recently confirmed with yeastproteins (118). By a structural approach, the molecular recognitionbetween Grx5 and IscA1/2 was investigated, and the cluster transferprocess between themwas characterized at amolecular level. This dem-onstrated that a switch between two conformational states of [2Fe–2S]-bound Grx drives the cluster transfer event, which occurs through aspecific protein-protein recognition process (118). Consistent withdata on IscU (see Section 3.6 with chaperones), these results revealthe general mode of Fe/S transfer which seems to be driven by a switchbetween two conformational states in the Fe/S donor. Unidirectionaland quantitative Fe/S cluster transfer from IscU to Grx5 was also dem-onstrated in vitro with A. vinelandii proteins (20) which suggests thatFe/S is transferred first from IscU to Grx and then to IscA. This order ofFe/S transfermay occur in some specific conditions (environmental, tar-get) since it is known that IscU can directly transfer its cluster to IscAwithout transiting via Grx (82). A challenge in the future will be todetermine, at a molecular level, the specific role played by Grx withinthe IscA–Grx complex and how it differs from that played by IscAalone in the [4Fe–4S]-protein maturation process, as IscA is known tobe capable of maturing such proteins without assistance. The role ofGrx might be to increase the rate of Fe/S transfer as second order kinet-ics measured with bacterial Grx are always significantly faster thanthose reported in its absence (20,66,114).

    4.2.3. IscA–target complexesProteins that directly receive an Fe/S cluster from IscA are consid-

    ered to be targets of IscA. These targets can be Fe/S carrier proteins orfinal apo-acceptors requiring an Fe/S cluster to be functional. Examplesof these include aconitase, succinate dehydrogenase, radical-SAMenzymes, etc. Few complexes between these proteins and IscA havebeen characterized in vitro, probably due to fast dissociation of thecomplex once the Fe/S is transferred. It was, however, possible to trapa complex between IscA and its target protein biotin synthase (BioB)using a BioB mutant protein in which one of its cysteine ligands wasmutated to alanine (82). Despite a lack of characterization of complexesbetween IscA and its targets in vitro, in vivo experiments have revealedsome specific interactions between IscA and the anaerobic respiratoryenzymes FdhN (formate dehydrogenase N), nitrate reductase (Nar),IspG/H enzymes and NiFe hydrogenase in Escherchia coli (67,86,87,119). Complexes between IscA and other Fe/S carrier proteins have

    yet to be characterized, although it is known that a unidirectional Fe/Stransfer from IscA to ErpA occurs (67). Since Fe/S carrier proteins(ErpA, SufA, NfuA, etc.) function under specific environmental condi-tions, we could hypothesize that hetero-complexes between differentFe/S carriers might exist under these conditions, allowing a better pro-tection of the cluster compared with isolated Fe/S carrier proteins. Allof these questions will have to be addressed in the coming years.

    All these results obtained with IscA and its interactors lead credenceto the hypothesis of IscA as an Fe/S protein rather than an Fe-protein.

    5. Conclusion

    Iron–sulfur cluster biogenesis is an essential process in biology, inbacteria it is governed by three distinct protein systems: NIF, ISC andSUF. Over the last twelve years, a great effort has been made to charac-terize isolated proteins. This has made it possible to identify the mainbasic steps of this process and the corresponding protein actors. Howev-er, recent studies have revealed the limitations of investigating this typeof complex process byworkingwith only one protein in a test tube, con-sidering the number of proteins involved in vivo. More recent in vitrocharacterization of protein complexes has challenged some of the re-sults obtained with isolated proteins meaning that some experimentswill have to be repeated with complexes, in particular those relatingto Fe/S assembly on IscU, which is the most extensively studied step.There is no doubt that investigations using such complexeswill advanceour molecular understanding of the Fe/S biogenesis process immeasur-ably in the coming years. With a view to working with a more integrat-ed system better mimicking the physiological pathway, environmentalconditions (stress, anaerobiosis) should be taken into account inin vitro experiments. Combined with in vivo studies, these should great-ly enhance our knowledge of Fe/S assembly in bacteria. There are stillmany crucial points remaining to be investigated in the next years tofully elucidate this fascinating biological process.

    Acknowledgments

    Work in our laboratory was generously supported by grants fromthe ANR Blanc (FeStreS 11BSV302202). We thank Marc Fontecave fordiscussion and support as well as our longstanding collaborators fromFrédéric Barras group in Marseille (LCB).

    References

    [1] H. Beinert, Iron–sulfur proteins: ancient structures, still full of surprises, J. Biol.Inorg. Chem. 5 (2000) 2–15.

    [2] M. Fontecave, Iron–sulfur clusters: ever-expanding roles, Nat. Chem. Biol. 2 (2006)171–174.

    [3] R. Lill, Function and biogenesis of iron–sulphur proteins, Nature 460 (2009)831–838.

    [4] B. Py, P.L. Moreau, F. Barras, Fe–S clusters, fragile sentinels of the cell, Curr. Opin.Microbiol. 14 (2011) 218–223.

    [5] M.R. Jacobson, et al., Biochemical and genetic analysis of the nifUSVWZM clusterfrom Azotobacter vinelandii, Mol. Gen. Genet. 219 (1989) 49–57.

    [6] J. Frazzon, D.R. Dean, Formation of iron–sulfur clusters in bacteria: an emergingfield in bioinorganic chemistry, Curr. Opin. Chem. Biol. 7 (2003) 166–173.

    [7] L. Zheng, V.L. Cash, D.H. Flint, D.R. Dean, Assembly of iron–sulfur clusters, Identifi-cation of an iscSUA–hscBA–fdx gene cluster from Azotobacter vinelandii.J. Biol.Chem. 273 (1998) 13264–13272.

    [8] Y. Takahashi, U. Tokumoto, A third bacterial system for the assembly of iron–sulfurclusters with homologs in archaea and plastids, J. Biol. Chem. 277 (2002)28380–28383.

    [9] L. Nachin, L. Loiseau, D. Expert, F. Barras, SufC: an unorthodox cytoplasmic ABC/ATPase required for [Fe–S] biogenesis under oxidative stress, EMBO J. 22 (2003)427–437.

    [10] J.W. Olson, J.N. Agar, M.K. Johnson, R.J. Maier, Characterization of the NifU and NifSFe–S cluster formation proteins essential for viability in Helicobacter pylori, Bio-chemistry 39 (2000) 16213–16219.

    [11] F. Prischi, et al., Structural bases for the interaction of frataxin with the centralcomponents of iron–sulphur cluster assembly, Nat. Commun. 1 (2010) 95.

    [12] C.J. Schwartz, O. Djaman, J.A. Imlay, P.J. Kiley, The cysteine desulfurase, IscS, has amajor role in in vivo Fe–S cluster formation in Escherichia coli, Proc. Natl. Acad.Sci. U. S. A. 97 (2000) 9009–9014.

    http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0005http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0005http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0010http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0010http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0015http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0015http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0020http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0020http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0025http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0025http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0030http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0030http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0585http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0585http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0585http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0040http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0040http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0040http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0045http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0045http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0045http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0050http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0050http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0050http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0055http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0055http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0060http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0060http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0060

  • 1446 B. Blanc et al. / Biochimica et Biophysica Acta 1853 (2015) 1436–1447

    [13] H. Mihara, T. Kurihara, T. Yoshimura, N. Esaki, Kinetic and mutational studies of threeNifS homologs from Escherichia coli: mechanistic difference between L-cysteinedesulfurase and L-selenocysteine lyase reactions, J. Biochem. Tokyo 127 (2000)559–567.

    [14] J.R. Cupp-Vickery, H. Urbina, L.E. Vickery, Crystal structure of IscS, a cysteinedesulfurase from Escherichia coli, J. Mol. Biol. 330 (2003) 1049–1059.

    [15] R. Shi, et al., Structural basis for Fe–S cluster assembly and tRNA thiolation mediat-ed by IscS protein–protein interactions, PLoS Biol. 8 (2010) e1000354.

    [16] J.N. Agar, et al., IscU as a scaffold for iron–sulfur cluster biosynthesis: sequential as-sembly of [2Fe–2S] and [4Fe–4S] clusters in IscU, Biochemistry 39 (2000)7856–7862.

    [17] U. Tokumoto, Y. Takahashi, Genetic analysis of the isc operon in Escherichia coli in-volved in the biogenesis of cellular iron–sulfur proteins, J. Biochem. Tokyo 130(2001) 63–71.

    [18] E.C. Raulfs, I.P. O'Carroll, P.C. Dos Santos, M.C. Unciuleac, D.R. Dean, In vivo iron–sulfur cluster formation, Proc. Natl. Acad. Sci. U. S. A. 105 (2008) 8591–8596.

    [19] Y. Shimomura, K.Wada, K. Fukuyama, Y. Takahashi, The asymmetric trimeric archi-tecture of [2Fe–2S] IscU: implications for its scaffolding during iron–sulfur clusterbiosynthesis, J. Mol. Biol. 383 (2008) 133–143.

    [20] P. Shakamuri, B. Zhang, M.K. Johnson, Monothiol glutaredoxins function in storingand transporting [Fe2S2] clusters assembled on IscU scaffold proteins, J. Am. Chem.Soc. 134 (2012) 15213–15216.

    [21] M.C. Unciuleac, et al., In vitro activation of apo-aconitase using a [4Fe–4S] cluster-loaded form of the IscU [Fe–S] cluster scaffolding protein, Biochemistry 46 (2007)6812–6821.

    [22] S.S. Mansy, G. Wu, K.K. Surerus, J.A. Cowan, Iron–sulfur cluster biosynthesis,Thermatoga maritima IscU is a structured iron–sulfur cluster assembly protein.J.Biol. Chem. 277 (2002) 21397–21404.

    [23] T.A. Ramelot, et al., Solution NMR structure of the iron–sulfur cluster assembly pro-tein U (IscU) with zinc bound at the active site, J. Mol. Biol. 344 (2004) 567–583.

    [24] J.H. Kim, et al., Structure and dynamics of the iron–sulfur cluster assembly scaffoldprotein IscU and its interaction with the cochaperone HscB, Biochemistry 48(2009) 6062–6071.

    [25] M.A. Huynen, B. Snel, P. Bork, T.J. Gibson, The phylogenetic distribution of frataxinindicates a role in iron–sulfur cluster protein assembly, Hum. Mol. Genet. 10(2001) 2463–2468.

    [26] H. Puccio, et al., Mouse models for Friedreich ataxia exhibit cardiomyopathy, sen-sory nerve defect and Fe–S enzyme deficiency followed by intramitochondrialiron deposits, Nat. Genet. 27 (2001) 181–186.

    [27] U. Muhlenhoff, N. Richhardt, M. Ristow, G. Kispal, R. Lill, The yeast frataxin homo-log Yfh1p plays a specific role in thematuration of cellular Fe/S proteins, Hum.Mol.Genet. 11 (2002) 2025–2036.

    [28] A. Rotig, et al., Aconitase and mitochondrial iron–sulphur protein deficiency inFriedreich ataxia, Nat. Genet. 17 (1997) 215–217.

    [29] E. Vivas, E. Skovran, D.M. Downs, Salmonella enterica strains lacking the frataxinhomolog CyaY show defects in Fe–S cluster metabolism in vivo, J. Bacteriol. 188(2006) 1175–1179.

    [30] D.S. Li, K. Ohshima, S. Jiralerspong, M.W. Bojanowski, M. Pandolfo, Knock-out of thecyaY gene in Escherichia coli does not affect cellular iron content and sensitivity tooxidants, FEBS Lett. 456 (1999) 13–16.

    [31] A.G. Albrecht, et al., The frataxin homologue Fra plays a key role in intracellulariron channeling in Bacillus subtilis, Chembiochem 12 (2011) 2052–2061.

    [32] J. Velayudhan, et al., Distinct roles of the Salmonella enterica serovar TyphimuriumCyaY and YggX proteins in the biosynthesis and repair of iron–sulfur clusters, In-fect. Immun. 82 (2014) 1390–1401.

    [33] T. Yoon, J.A. Cowan, Iron–sulfur cluster biosynthesis. Characterization of frataxin asan iron donor for assembly of [2Fe–2S] clusters in ISU-type proteins, J. Am. Chem.Soc. 125 (2003) 6078–6084.

    [34] M. Nair, et al., Solution structure of the bacterial frataxin ortholog, CyaY: mappingthe iron binding sites, Structure 12 (2004) 2037–2048.

    [35] J.D. Cook, et al., Monomeric yeast frataxin is an iron-binding protein, Biochemistry45 (2006) 7767–7777.

    [36] M. Nair, et al., Solution structure of the bacterial frataxin ortholog, CyaY: mappingthe iron binding sites, Struct. Camb. 12 (2004) 2037–2048.

    [37] G. Layer, S. Ollagnier-de Choudens, Y. Sanakis, M. Fontecave, Iron–sulfur cluster bio-synthesis: characterization of Escherichia coli CYaY as an iron donor for the assemblyof [2Fe–2S] clusters in the scaffold IscU, J. Biol. Chem. 281 (2006) 16256–16263.

    [38] O. Gakh, et al., Mitochondrial iron detoxification is a primary function of frataxinthat limits oxidative damage and preserves cell longevity, Hum. Mol. Genet. 15(2006) 467–479.

    [39] J. Adamec, et al., Iron-dependent self-assembly of recombinant yeast frataxin: im-plications for Friedreich ataxia, Am. J. Hum. Genet. 67 (2000) 549–562.

    [40] J. Gerber, U. Muhlenhoff, R. Lill, An interaction between frataxin and Isu1/Nfs1 thatis crucial for Fe/S cluster synthesis on Isu1, EMBO Rep. 4 (2003) 906–911.

    [41] K. Aloria, B. Schilke, A. Andrew, E.A. Craig, Iron-induced oligomerization of yeastfrataxin homologue Yfh1 is dispensable in vivo, EMBO Rep. 5 (2004) 1096–1101.

    [42] O. Gakh, et al., Normal and Friedreich ataxia cells express different isoforms offrataxin with complementary roles in iron–sulfur cluster assembly, J. Biol. Chem.285 (2010) 38486–38501.

    [43] S. Schmucker, et al., Mammalian frataxin: an essential function for cellular viabilitythrough an interaction with a preformed ISCU/NFS1/ISD11 iron–sulfur assemblycomplex, PLoS One 6 (2011) e16199.

    [44] S. Adinolfi, et al., Bacterial frataxin CyaY is the gatekeeper of iron–sulfur cluster for-mation catalyzed by IscS, Nat. Struct. Mol. Biol. 16 (2009) 390–396.

    [45] C.L. Tsai, D.P. Barondeau, Human frataxin is an allosteric switch that activates theFe–S cluster biosynthetic complex, Biochemistry 49 (2010) 9132–9139.

    [46] U. Tokumoto, et al., Network of protein–protein interactions among iron–sulfurcluster assembly proteins in Escherichia coli, J. Biochem. Tokyo 131 (2002)713–719.

    [47] Y. Shimomura, Y. Takahashi, Y. Kakuta, K. Fukuyama, Crystal structure ofEscherichia coli YfhJ protein, a member of the ISC machinery involved in assemblyof iron–sulfur clusters, Proteins 60 (2005) 566–569.

    [48] C. Pastore, et al., YfhJ, a molecular adaptor in iron–sulfur cluster formation or afrataxin-like protein? Structure 14 (2006) 857–867.

    [49] J.H. Kim, J.R. Bothe, R.O. Frederick, J.C. Holder, J.L. Markley, Role of IscX in iron–sul-fur cluster biogenesis in Escherichia coli, J. Am. Chem. Soc. (2014) 7933–7942.

    [50] Y.H. Lee, et al., Binding energetics of ferredoxin-NADP+ reductase with ferredoxinand its relation to function, Chembiochem 12 (2011) 2062–2070.

    [51] P. Mulo, Chloroplast-targeted ferredoxin-NADP(+) oxidoreductase (FNR): struc-ture, function and location, Biochim. Biophys. Acta 1807 (2011) 927–934.

    [52] C.R. Staples, et al., The function and properties of the iron–sulfur center in spinachferredoxin: thioredoxin reductase: a new biological role for iron–sulfur clusters,Biochemistry 35 (1996) 11425–11434.

    [53] T.G. Spiro, Iron–Sulfur Proteins, 1982. (New York).[54] Y. Kakuta, T. Horio, Y. Takahashi, K. Fukuyama, Crystal structure of Escherichia coli

    Fdx, an adrenodoxin-type ferredoxin involved in the assembly of iron–sulfur clus-ters, Biochemistry 40 (2001) 11007–11012.

    [55] H. Lange, A. Kaut, G. Kispal, R. Lill, A mitochondrial ferredoxin is essential for bio-genesis of cellular iron–sulfur proteins, Proc. Natl. Acad. Sci. U. S. A. 97 (2000)1050–1055.

    [56] K. Chandramouli, et al., Formation and properties of [4Fe–4S] clusters on the IscUscaffold protein, Biochemistry 46 (2007) 6804–6811.

    [57] S. Ollagnier-de-Choudens, T. Mattioli, Y. Takahashi, M. Fontecave, Iron–sulfur clus-ter assembly: characterization of IscA and evidence for a specific and functionalcomplex with ferredoxin, J. Biol. Chem. 276 (2001) 22604–22607.

    [58] K.G. Hoff, J.J. Silberg, L.E. Vickery, Interaction of the iron–sulfur cluster assemblyprotein IscU with the Hsc66/Hsc20 molecular chaperone system of Escherichiacoli, Proc. Natl. Acad. Sci. U. S. A. 97 (2000) 7790–7795.

    [59] J.J. Silberg, K.G. Hoff, T.L. Tapley, L.E. Vickery, The Fe/S assembly protein IscU be-haves as a substrate for the molecular chaperone Hsc66 from Escherichia coli, J.Biol. Chem. 276 (2001) 1696–1700.

    [60] K.G. Hoff, J.R. Cupp-Vickery, L.E. Vickery, Contributions of the LPPVK motif of theiron–sulfur template protein IscU to interactions with the Hsc66–Hsc20 chaperonesystem, J. Biol. Chem. 278 (2003) 37582–37589.

    [61] J.R. Cupp-Vickery, J.C. Peterson, D.T. Ta, L.E. Vickery, Crystal structure of the molec-ular chaperone HscA substrate binding domain complexed with the IscU recogni-tion peptide ELPPVKIHC, J. Mol. Biol. 342 (2004) 1265–1278.

    [62] T.L. Tapley, L.E. Vickery, Preferential substrate binding orientation by themolecularchaperone HscA, J. Biol. Chem. 279 (2004) 28435–28442.

    [63] A.K. Fuzery, et al., Solution structure of the iron–sulfur cluster cochaperone HscBand its binding surface for the iron–sulfur assembly scaffold protein IscU, Biochem-istry 47 (2008) 9394–9404.

    [64] S.P. Wu, S.S. Mansy, J.A. Cowan, Iron–sulfur cluster biosynthesis, Molecular chaper-one DnaK promotes IscU-bound [2Fe–2S] cluster stability and inhibits clustertransfer activity.Biochemistry 44 (2005) 4284–4293.

    [65] U. Muhlenhoff, J. Gerber, N. Richhardt, R. Lill, Components involved in assemblyand dislocation of iron–sulfur clusters on the scaffold protein Isu1p, Embo J. 22(2003) 4815–4825.

    [66] K. Chandramouli, M.K. Johnson, HscA and HscB stimulate [2Fe–2S] cluster transferfrom IscU to apoferredoxin in an ATP-dependent reaction, Biochemistry 45 (2006)11087–11095.

    [67] D. Vinella, C. Brochier-Armanet, L. Loiseau, E. Talla, F. Barras, Iron–sulfur (Fe/S) pro-tein biogenesis: phylogenomic and genetic studies of A-type carriers, PLoS Genet. 5(2009) e1000497.

    [68] P.W. Bilder, H. Ding, M.E. Newcomer, Crystal structure of the ancient, Fe–S scaffoldIscA reveals a novel protein fold, Biochemistry 43 (2004) 133–139.

    [69] J.R. Cupp-Vickery, J.J. Silberg, D.T. Ta, L.E. Vickery, Crystal structure of IscA, an iron–sulfur cluster assembly protein from Escherichia coli, J. Mol. Biol. 338 (2004)127–137.

    [70] C. Krebs, et al., IscA, an alternate scaffold for Fe–S cluster biosynthesis, Biochemis-try 40 (2001) 14069–14080.

    [71] D.T. Mapolelo, B. Zhang, S.G. Naik, B.H. Huynh, M.K. Johnson, Spectroscopic andfunctional characterization of iron–sulfur cluster-bound forms of Azotobactervinelandii (Nif)IscA, Biochemistry 51 (2012) 8071–8084.

    [72] K. Morimoto, et al., The asymmetric IscA homodimer with an exposed [2Fe–2S]cluster suggests the structural basis of the Fe–S cluster biosynthetic scaffold, J.Mol. Biol. 360 (2006) 117–132.

    [73] J. Zeng, et al., The IscA from Acidithiobacillus ferrooxidans is an iron–sulfur proteinwhich assemble the [Fe(4)S(4)] cluster with intracellular iron and sulfur, Arch.Biochem. Biophys. 463 (2007) 237–244.

    [74] H. Ding, K. Harrison, J. Lu, Thioredoxin reductase system mediates iron binding inIscA and iron delivery for the iron–sulfur cluster assembly in IscU, J. Biol. Chem.280 (2005) 30432–30437.

    [75] H. Ding, R.J. Clark, Characterization of iron binding in IscA, an ancient iron–sulphurcluster assembly protein, Biochem. J. 379 (2004) 433–440.

    [76] D.T. Mapolelo, B. Zhang, S.G. Naik, B.H. Huynh, M.K. Johnson, Spectroscopic andfunctional characterization of iron-bound forms of Azotobacter vinelandii (Nif)IscA, Biochemistry 51 (2012) 8056–8070.

    [77] L. Qian, C. Zheng, J. Liu, Characterization of iron–sulfur cluster assembly proteinIscA from Acidithiobacillus ferrooxidans, Biochemistry (Mosc) 78 (2013) 244–251.

    [78] W. Wang, et al., In vivo evidence for the iron-binding activity of an iron–sulfurcluster assembly protein IscA in Escherichia coli, Biochem. J. 432 (2010) 429–436.

    http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0065http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0065http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0065http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0065http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0070http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0070http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0075http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0075http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0080http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0080http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0080http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0085http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0085http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0085http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0090http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0090http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0095http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0095http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0095http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0100http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0100http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0100http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0105http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0105http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0105http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0590http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0590http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0590http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0115http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0115http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0120http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0120http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0120http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0125http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0125http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0125http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0130http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0130http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0130http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0135http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0135http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0135http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0140http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0140http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0145http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0145http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0145http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0150http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0150http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0150http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0155http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0155http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0160http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0160http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0160http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0165http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0165http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0165http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0595http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0595http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0175http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0175http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0180http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0180http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0185http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0185http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0185http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0190http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0190http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0190http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0195http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0195http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0200http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0200http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0205http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0205http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0210http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0210http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0210http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0215http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0215http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0215http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0220http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0220http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0225http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0225http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0230http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0230http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0230http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0235http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0235http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0235http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0240http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0240http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0600http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0600http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0245http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0245http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0245http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0250http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0250http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0255http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0255http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0255http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0605http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0260http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0260http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0260http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0265http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0265http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0265http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0270http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0270http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0275http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0275http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0275http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0280http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0280http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0280http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0285http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0285http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0285http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0290http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0290http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0290http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0295http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0295http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0295http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0300http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0300http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0305http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0305http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0305http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0610http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0610http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0610http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0315http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0315http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0315http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0320http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0320http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0320http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0325http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0325http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0325http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0615http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0615http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0335http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0335http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0335http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0340http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0340http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0345http://refhub.elsevier.com/S0167-4889(14)00437-6/rf0345http://refhub.elsevier.com/S0167-4889(14