throbber
Cell Metabolism
`
`Article
`
`Cytosolic Monothiol Glutaredoxins Function in
`Intracellular Iron Sensing and Trafficking via
`Their Bound Iron-Sulfur Cluster
`
`Ulrich Mu¨ hlenhoff,1 Sabine Molik,1 Jose´ R. Godoy,1 Marta A. Uzarska,1 Nadine Richter,1 Andreas Seubert,2 Yan Zhang,3
`JoAnne Stubbe,3 Fabien Pierrel,4 Enrique Herrero,5 Christopher Horst Lillig,1 and Roland Lill1,*
`1Institut fu¨ r Zytobiologie und Zytopathologie
`2Fachbereich Chemie, Philipps-Universita¨ t Marburg
`35032 Marburg, Germany
`3Chemistry Department, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
`4Laboratoire de Chimie et Biologie des Me´ taux, CEA Grenoble, 38054 Grenoble Cedex 9, France
`5Departament de Cie` ncies Me` diques Ba` siques, IRB Lleida, Universitat de Lleida, Lleida 25198, Spain
`*Correspondence: lill@staff.uni-marburg.de
`DOI 10.1016/j.cmet.2010.08.001
`
`SUMMARY
`
`Iron is an essential nutrient for cells. It is unknown
`how iron, after
`its import
`into the cytosol,
`is
`specifically delivered to iron-dependent processes
`in various cellular compartments. Here, we identify
`an essential function of the conserved cytosolic
`monothiol glutaredoxins Grx3 and Grx4 in intracel-
`lular iron trafficking and sensing. Depletion of Grx3/4
`specifically impaired all
`iron-requiring reactions in
`the cytosol, mitochondria, and nucleus, including
`the synthesis of Fe/S clusters, heme, and di-iron
`centers. These defects were caused by impairment
`of iron insertion into proteins and iron transfer to mito-
`chondria,
`indicating that intracellular iron is not
`bioavailable, despite highly elevated cytosolic levels.
`The crucial task of Grx3/4 is mediated by a bridging,
`glutathione-containing Fe/S center that functions
`both as an iron sensor and in intracellular iron
`delivery. Collectively, our study uncovers an impor-
`tant role of monothiol glutaredoxins in cellular iron
`metabolism, with a surprising connection to cellular
`redox and sulfur metabolisms.
`
`INTRODUCTION
`
`Iron is essential for virtually all organisms, because it functions as
`a cofactor in central cellular processes such as respiration, DNA
`synthesis and repair, ribosome biogenesis, and metabolism.
`Research over the past decade has uncovered sophisticated
`systems facilitating the specific transport of iron across the
`plasma and various intracellular membranes (Hentze et al.,
`2004; Kaplan and Kaplan, 2009; Philpott and Protchenko, 2008;
`Vergara and Thiele, 2008). Despite its central metabolic function,
`little is known about the passage of iron through the eukaryotic
`cytosol to become incorporated into proteins and transported
`into various subcellular compartments. A soluble, low-molec-
`ular-mass form of iron was described in the 1970s, but ever since
`
`the discovery of this ‘‘labile iron pool,’’ its physiological impor-
`tance and composition have been under debate (Crichton and
`Charloteaux-Wauters, 1987; Richardson and Ponka, 1997).
`Presumably, iron may also be bound to dedicated proteins
`ensuring specific delivery and insertion into iron-requiring sites.
`A metallo-chaperone function has been worked out for insertion
`of copper and nickel into respective metal-containing enzymes
`(Finney and O’Halloran, 2003; Lyons and Eide, 2007), but proteins
`performing a general role in iron trafficking or insertion are
`unknown. An iron donor function has been suggested for frataxin
`in mitochondrial Fe/S cluster biosynthesis (Bencze et al., 2006;
`Lill, 2009). In humans, the poly (rC) binding protein 1 (PCBP1)
`was shown to specifically deliver bound iron to ferritin, the major
`iron storage protein in higher eukaryotes (Shi et al., 2008). The
`apparently specific role of the PCBP1 iron chaperone and the
`fact that both ferritin and PCBP1 are not universally conserved
`leave open the possibility that other proteins with a general
`importance for iron trafficking exist. Clearly, the mode of specific
`iron delivery within the eukaryotic cytosol remains one of the
`fundamental unresolved problems of iron homeostasis.
`Because iron is not only essential but also toxic at higher
`levels, cells have developed sophisticated systems for ensuring
`a tightly regulated iron homeostasis (Hentze et al., 2004; Kaplan
`and Kaplan, 2009). In mammals this process is executed by
`iron-regulatory proteins in a posttranscriptional fashion, and
`the yeast Saccharomyces cerevisiae uses the iron-sensing
`transcription factors Aft1 and Aft2. Under iron deprivation,
`Aft1-Aft2 activate transcription of genes of the iron regulon
`encoding cell-surface iron transporters and proteins involved in
`intracellular iron utilization (Kaplan and Kaplan, 2009; Philpott
`and Protchenko, 2008). Sensing of intracellular iron by Aft1
`also requires the regulatory proteins Fra1-Fra2 and the cyto-
`solic-nuclear monothiol glutaredoxins Grx3 and Grx4, which
`are essential for the nuclear export of Aft1 in response to iron
`sufficiency (Kumanovics et al., 2008; Ojeda et al., 2006; Pujol-
`Carrion et al., 2006). The regulatory role of Grx3/4 is functionally
`conserved in fungi
`that utilize iron-regulated transcription
`systems unrelated to those from S. cerevisiae (Haas et al.,
`2008; Kaplan and Kaplan, 2009; Mercier and Labbe, 2009).
`Grx3/4 belong to the large thioredoxin (Trx) fold family and are
`composed of an N-terminal Trx and a C-terminal monothiol
`
`Cell Metabolism 12, 373–385, October 6, 2010 ª2010 Elsevier Inc. 373
`
`

`

`Role of Cytosolic Glutaredoxin in Iron Trafficking
`
`Cell Metabolism
`
`glutaredoxin (Grx) domain (Herrero and de la Torre-Ruiz, 2007;
`Lillig et al., 2008). Although the Grx3/4 subfamily of multidomain
`monothiol glutaredoxins is conserved in eukaryotes, no universal
`function has been assigned to this family so far. In contrast to
`most members of the Grx protein family that catalyze dithiol-di-
`sulfide redox reactions, monothiol Grx proteins rarely possess
`oxidoreductase activity. Instead, after in vitro reconstitution or
`upon overexpression in Escherichia coli, they are capable of
`binding a bridging [2Fe-2S] cluster utilizing the active-site
`cysteine residue of the Grx domain and glutathione (GSH) as
`ligands (Li et al., 2009; Picciocchi et al., 2007). The existence
`of this unusual Fe/S center under physiological conditions,
`however, has not been demonstrated, and its functional role
`has remained unclear.
`Here, we have used yeast as a model to define an essential
`role of Grx3/4 in intracellular iron trafficking. Depletion of Grx3/
`4 led to functional
`impairment of virtually all
`iron-dependent
`processes,
`including heme biosynthesis, mitochondrial and
`cytosolic Fe/S protein biogenesis, and the formation of di-iron
`centers in mitochondria and the cytosol, eventually leading to
`the loss of cell viability. We provide evidence for the in vivo
`binding of a bridging Fe/S cluster to Grx3/4 and we assign
`a crucial physiological function to this cofactor both in cytosolic
`iron trafficking and as an iron sensor. Thus, the conserved cyto-
`solic monothiol glutaredoxins use their bound Fe/S cofactor for
`a general role in intracellular iron trafficking.
`
`RESULTS
`
`Deficiency in Grx3/4 Is Associated with Defects
`in Iron-Dependent Enzymes
`To facilitate the functional analysis of Grx3/4, we constructed
`a regulatable yeast strain (Gal-GRX4; strain background W303-
`1A; see Table S1 available online) in which GRX3 was deleted
`and GRX4 was expressed under the control of the glucose-
`repressible GAL-L promoter. Upon Grx4 depletion, Gal-GRX4
`cells failed to grow on both fermentable and nonfermentable
`carbon sources (Figure 1A; see below). Likewise, double deletion
`of GRX3-GRX4 was lethal
`in the W303 strain background,
`distinguishing these cells from strain BY4742 grx3/4D, which
`shows only severely retarded growth (Figure 1A) (Ojeda et al.,
`2006). The strong effect of Grx3/4 deficiency on cell viability is
`not explained by their role in iron regulation, since Aft1 is not
`essential under iron-replete conditions (Kaplan and Kaplan,
`2009). These data and the general conservation of Grx3/4 in
`eukaryotes suggest
`that
`these proteins perform a so far
`unknown, important function.
`Gal-GRX4 cells were used to investigate the immediate conse-
`quences of Grx3/4 deficiency. Gal-GRX4 cells were cultivated in
`minimal medium supplemented with glucose and iron chloride to
`gradually deplete Grx4 (Figures 1B and 1C). A strong activation
`of the Aft1-dependent FET3 gene was observed using a FET3
`promoter-GFP fusion as a reporter (Figure 1B) (Ojeda et al.,
`2006). Surprisingly, the activities of the mitochondrial Fe/S
`protein aconitase and cytosolic catalase, a heme-containing
`protein, drastically decreased, despite the presumed sufficient
`cellular iron supply. These effects resemble those upon deple-
`tion of Ssq1, a component the iron-sulfur cluster (ISC) assembly
`machinery, even though the changes occurred later as a result of
`
`slower depletion of Ssq1 (strain Gal-SSQ1). Grx3/4 deficiency
`was associated with a severe activity loss of
`respiratory
`complexes II (succinate dehydrogenase) and IV (cytochrome
`oxidase) but was fully complemented by expression of GRX4
`from a plasmid (Figure 1D). Likewise, low activities of both
`aconitase and respiratory complexes III and IV were observed
`in BY4742 grx3/4D cells (Figure S1A), consistent with our earlier
`observation of an impaired 55Fe/S cluster incorporation into
`aconitase (Ojeda et al., 2006). Immunostaining of cell extracts
`from Grx4-depleted Gal-GRX4 cells and BY4742 grx3/4D cells
`further revealed changes in the levels of several iron-containing
`proteins, including the aconitase-type Fe/S proteins Aco1 and
`Leu1, ferrochelatase Hem15, and the core mitochondrial ISC
`assembly protein Isu1 (Figure 1C). These changes of protein
`levels correlate with those of the transcriptome of both iron-
`deprived and ISC machinery–compromised cells (Hausmann
`et al., 2008; Shakoury-Elizeh et al., 2004). In contrast, other
`iron-dependent proteins, such as succinate dehydrogenase
`subunit 2 (Sdh2) and the ubiquinone biosynthesis enzyme
`Coq7, were hardly changed and behaved similarly to the noniron
`proteins mitochondrial cytochrome oxidase subunit 4 (Cox4) and
`porin (Por1), cytosolic Hsp70, and ribosomal subunit Rps3.
`Together, these findings indicate that Grx3/4-deficient cells
`develop severe defects in several mitochondrial and cytosolic
`iron-dependent proteins, despite the induction of the Aft1-
`dependent iron uptake system. Notably, these global
`iron-
`related defects are not detected upon the constitutive activation
`of Aft1 (Hausmann et al., 2008; Ihrig et al., 2010), suggesting that
`these consequences of Grx3/4 deficiency occur independently
`of Aft1 and a deregulated iron homeostasis.
`
`Deficiency in Grx3/4 Impairs the De Novo Synthesis
`of Cellular Fe/S Clusters and Heme
`We asked whether the decreased Fe/S protein activities in Grx3/
`4-depleted cells might be explained by an impaired de novo
`synthesis of their Fe/S clusters and addressed this problem by
`using an established 55Fe radiolabeling and immunoprecipitation
`assay (Molik et al., 2007). First, the essential cytosolic Fe/S
`proteins Rli1, Dre2, and Nar1 were analyzed by expressing these
`proteins from a high-copy vector in wild-type and Gal-GRX4
`cells. Fe/S cluster insertion into all three Fe/S protein targets
`was decreased 4–10-fold upon Grx4 depletion (Figure 2A). The
`amount of Dre2 in Gal-GRX4 cells was comparable to that in
`wild-type cells,
`indicating a specific Fe/S cluster assembly
`defect (Figure 2A, inset). In the case of Rli1 and Nar1, protein
`levels were diminished, likely suggesting that the apoforms of
`these Fe/S proteins were degraded. Similar apoprotein insta-
`bility is frequently observed upon strong defects in Fe/S protein
`biogenesis (Balk et al., 2004). Analysis of 55Fe incorporation into
`the mitochondrial Fe/S proteins Bio2 (biotin synthase) and Ilv3
`(dihydroxyacid dehydratase) and the essential mitochondrial
`ISC scaffold protein Isu1 revealed an up to 4-fold lower 55Fe
`incorporation upon Grx4 depletion (Figures 2B and 2C). Protein
`levels of Bio2, Ilv3, and Isu1 did not change upon depletion of
`Grx4 (Figures 2B and 2C,
`insets). These findings indicate
`a general impairment in the de novo assembly of Fe/S proteins
`upon depletion of Grx3/4.
`In principle, the defect of Fe/S protein maturation upon Grx3/4
`depletion could be explained by a primary mitochondrial Fe/S
`
`374 Cell Metabolism 12, 373–385, October 6, 2010 ª2010 Elsevier Inc.
`
`

`

`Cell Metabolism
`
`Role of Cytosolic Glutaredoxin in Iron Trafficking
`
`Figure 1. Deficiency in Grx3/4 Is Associated with Defects in Iron-Dependent Enzymes
`(A) Wild-type (WT), Gal-GRX4, grx3D (strain background W303-1A), and grx3/4D (strain background BY4742) were grown in SD medium for 40 hr. Ten-fold serial
`
`C.
`dilutions were spotted onto YPEG plates and incubated for 2 days at 30
`(B and C) WT, Gal-GRX4, and Gal-SSQ1 strains harboring plasmid pFET3-GFP were grown in SD minimal medium. At the indicated times, FET3 promoter activ-
`ities were determined by measuring the GFP-specific fluorescence emission of cells (B), and cell extracts were assayed for aconitase and catalase activities, or
`were analyzed for the indicated proteins by immunostaining (C).
`(D) Enzyme activities of respiratory complexes II (SDH) and IV (COX) were determined relative to malate dehydrogenase (MDH) in mitochondria isolated from
`Gal-GRX4 cells cultivated in rich glucose medium for 40 hr and 64 hr, and from Gal-GRX4 cells expressing GRX4 from vector pCM189 (+Grx4). Error bars indicate
`the SEM (n R 4).
`
`protein assembly defect, because mitochondria are involved in
`generation of all cellular Fe/S proteins (Lill and Mu¨ hlenhoff,
`2008). However, we note that the observed effects were less
`severe in mitochondria compared to the cytosol. To directly
`test the functionality of the mitochondrial ISC assembly ma-
`chinery, we made use of an anaerobic in vitro system analyzing
`the capacity of mitochondrial detergent extracts to support Fe/S
`cluster insertion into the apoform of purified Yah1, a [2Fe-2S]
`
`ferredoxin (Molik et al., 2007). Apo-Yah1 was incubated with
`mitochondrial lysates and 55Fe. The radiolabeled Yah1 holopro-
`tein was bound to Q-sepharose, and the amount of radioactivity
`associated with holo-Yah1 was quantified by scintillation count-
`ing. Remarkably, extracts derived from Grx3/4-depleted mito-
`chondria were even more competent in synthesizing the Fe/S
`cluster on Yah1 than Grx4-complemented Gal-GRX4 cells
`(Figure 2D). This finding strongly suggests that the mitochondrial
`
`Cell Metabolism 12, 373–385, October 6, 2010 ª2010 Elsevier Inc. 375
`
`

`

`Role of Cytosolic Glutaredoxin in Iron Trafficking
`
`Cell Metabolism
`
`Figure 2. Deficiency in Grx3/4 Impairs the De Novo Synthesis of Cellular Fe/S Clusters and Heme
`(A–C) Wild-type (WT) and Grx4-depleted Gal-GRX4 cells overproducing the cytosolic Fe/S proteins Rli1-HA, Dre2 or Nar1 (A), mitochondrial Bio2, Ilv3-Myc (B), or
`Isu1 (C) were radiolabeled with 10 mCi 55Fe for 2 hr. The Fe/S proteins were immunoprecipitated, and bound 55Fe was quantified by scintillation counting. Protein
`levels were assessed by immunostaining. Porin (Por1) served as a loading control. Gal-GRX4 cells were depleted for Grx4 by growth in SD medium for 40 hr and
`64 hr (in case of Isu1).
`(D) Purified apo-Yah1 was incubated under anaerobic conditions in the presence of 55Fe and cysteine either alone (-) or with detergent extracts of mitochondria
`isolated from 40 hr or 64 hr depleted Gal-GRX4 cells or Gal-GRX4 cells overproducing Grx4 (+Grx4). 55Fe/S cluster reconstitution on re-isolated Yah1 was quan-
`tified by scintillation counting.
`(E) WT and Gal-GRX4 cells (40 hr depletion) harboring either vector pCM189 (-) or pCM189-GRX4 (+Grx4) were radiolabeled with 55Fe. 55Fe-heme was extracted
`with butyl-acetate and quantified by scintillation counting. Error bars indicate the SEM (n R 4).
`
`in Grx3/4-depleted cells,
`ISC assembly system is functional
`rendering it likely that the decreased Fe/S cluster incorporation
`into apoproteins is explained by impaired iron supply.
`Consistent with this idea, 55Fe insertion into heme was 5.5-fold
`lower
`in Grx4-depleted Gal-GRX4 cells, compared with
`wild-type cells or Gal-GRX4 cells complemented by GRX4
`(Figure 2E). This diminished heme synthesis activity may explain
`the loss of function of heme-dependent enzymes, such as cata-
`lase and cytochrome oxidase, upon depletion of Grx3/4 (see
`above). In summary, Grx3/4-depleted cells are strongly impaired
`in both cellular Fe/S protein maturation and heme biosynthesis.
`Such defects are not observed in Aft1-activated cells (Haus-
`mann et al., 2008; Ihrig et al., 2010).
`
`Deficiency in Grx3/4 Leads to Impairment of Di-Iron
`Enzymes Despite Cytosolic Iron Overload
`The strong decrease of cellular Fe/S clusters and heme in Grx3/
`4-deficient cells is somewhat paradoxical, because these cells
`are expected to accumulate iron as the result of a constitutively
`activated cellular iron uptake system (Ojeda et al., 2006). To
`verify this idea, we measured the cellular iron content by ICP-MS
`analysis of wild-type and Grx4-depleted Gal-GRX4 cells grown
`in minimal medium supplemented with 100 mM FeCl3. Total
`cellular
`iron increased 6-fold upon depletion of Grx4
`
`(Figure 3A). The level of chelatable iron increased similarly and
`was mainly present as Fe2+ (Figure S2). Cellular levels of other
`metals, with the exception of Zn (3-fold higher), were hardly
`changed. In contrast, mitochondrial iron levels were up to 2.3-
`fold lower in Grx4-depleted Gal-GRX4 cells compared with
`wild-type levels (Figure 3B). Mitochondria from BY4742 grx3/
`4D cells contained even 7.5-fold less iron. Mitochondrial Mn,
`Co, and Zn levels were hardly altered, but Cu changed in parallel
`to iron. The decrease in mitochondrial
`iron levels in Grx3/4-
`depleted cells is the more remarkable, because cells with mito-
`chondrial Fe/S protein assembly defects usually display strongly
`elevated mitochondrial iron levels (Lill and Mu¨ hlenhoff, 2008).
`The fact that this was not observed, despite high levels of total
`cellular iron, indicates a defective delivery of iron to mitochondria
`in Grx3/4-deficient cells.
`A reasonable explanation for these general defects in iron
`handling in the absence of Grx3/4 may be a sequestration of
`iron into the vacuole, the major iron storage compartment in
`fungi
`(Kaplan and Kaplan, 2009; Philpott and Protchenko,
`2008). To test this idea, we varied the amount of Ccc1, the major
`importer of divalent metals into the vacuole. Deletion of CCC1
`did not restore growth of Grx4-depleted Gal-GRX4 cells and
`did not increase the low enzyme activities of aconitase and
`catalase (Figure 3C,
`top panel, and Figure 3D). Similarly,
`
`376 Cell Metabolism 12, 373–385, October 6, 2010 ª2010 Elsevier Inc.
`
`

`

`Cell Metabolism
`
`Role of Cytosolic Glutaredoxin in Iron Trafficking
`
`Figure 3. Deficiency in Grx3/4 Results in Cytosolic Iron and GSH Accumulation
`(A and B) The metal content of (A) wild-type (WT) and Gal-GRX4 cells (depleted for 64 hr) and (B) mitochondria isolated from the indicated strains was determined
`by ICP-MS.
`(C) The indicated strains lacking (ccc1D) or overproducing Ccc1 (Ccc1[) were cultivated in SD medium for 40 hr. Ten-fold serial dilutions were spotted onto SC
`C under aerobic (+O2) or anaerobic (O2) conditions.
`
`agar plates containing glycerol (Glyc) or glucose (Glc), and were cultivated at 30
`(D) WT, Gal-GRX4 and Gal-GRX4/ccc1D cells were grown in SD medium for 64 hr, and aconitase and catalase enzyme activities were determined.
`(E) GSH levels were determined in cell extracts from WT, BY4742 grx3/4D, Gal-GRX4 (depleted for 40 hr or 64 hr) and Gal-GRX4 cells expressing GRX4 from
`a plasmid (+Grx4).
`
`overproduction of Ccc1 failed to restore growth (Figure 3C,
`middle panel). These data suggest that the accumulated iron is
`not stored in the vacuole.
`Grx4-depleted Gal-GRX4 cells failed to grow under anaerobic
`conditions (Figure 3C, bottom panel). Thus, reactive oxygen
`species (due to increased iron levels) are not responsible for
`the lethal phenotype of Grx3/4-depleted cells. Moreover,
`oxidized glutathione levels (GSSG; measured under anaerobic
`conditions) were below the detection limit (not shown). Rather,
`reduced glutathione (GSH) was strongly elevated in Grx4-
`in BY4742 grx3/4D
`depleted Gal-GRX4 cells, but not
`(Figure 3E). Together, these results and the predominant pres-
`ence of iron in its ferrous form (Figure S2) indicate that reducing
`conditions prevail in Grx3/4-deficient cells.
`
`The experiments presented above showed a maturation
`defect in cellular Fe/S and heme proteins in Grx3/4-deficient
`cells despite a cytosolic iron accumulation. Because this
`indicated a defective delivery of iron, we asked whether other
`iron-dependent enzymes were affected by Grx3/4 deficiency.
`First, the iron status of ribonucleotide reductase (Rnr), a cytosolic
`diferric-tyrosyl radical enzyme essential for deoxyribonucleotide
`synthesis, was analyzed (Perlstein et al., 2005). Upon depletion
`of Grx4 in Gal-GRX4 cells, the protein levels of Rnr2 decreased
`slightly (Figure S3). Nevertheless, the specific activity of Rnr
`was 6-fold lower compared with wild-type cells (Figure 4A).
`This was likely due to inefficient metallation, because 55Fe inser-
`tion into Rnr in vivo (followed by immunoprecipitation of subunit
`Rnr2) was 5–7-fold decreased in Grx4-depleted cells (Figure 4B).
`
`Cell Metabolism 12, 373–385, October 6, 2010 ª2010 Elsevier Inc. 377
`
`

`

`Role of Cytosolic Glutaredoxin in Iron Trafficking
`
`Cell Metabolism
`
`Figure 4. Deficiency in Grx3/4 Leads to Functional Impairment of Di-iIron Enzymes
`(A) Permeabilized wild-type (WT) and Grx4-depleted Gal-GRX4 cells were assayed for specific ribonucleotide reductase activity (see also Figure S3).
`(B) WT and Gal-GRX4 cells were grown in SD medium for 40 and 64 hr and were radiolabeled, and 55Fe binding to Rnr2 was analyzed by immunoprecipitation and
`scintillation counting. Protein levels of Rnr2 and Por1 were determined by immunoblotting (inset).
`(C) The substrate (demethoxyubiquinol DMQ6) and product (ubiquinone CoQ6) of mitochondrial mono-oxygenase Coq7 were analyzed by electrochemical
`detection coupled to HPLC separation of mitochondrial lipid extracts from WT and Grx4-depleted Gal-GRX4 cells. CoQ4 is a commercial standard.
`(D) Ratio of DMQ6 and CoQ6 levels was determined in mitochondria isolated from WT Gal-GRX4 cells (depleted for 40 hr or 64 h) containing either vector pCM189
`or pCM189-GRX4 (+Grx4).
`(E) WT and Grx4-depleted Gal-GRX4 cells overproducing Fe-only superoxide dismutase from E. coli (FeSod) were analyzed for superoxide dismutase in-gel
`activities and FeSod by immunoblotting.
`
`As a second di-iron protein, we analyzed the mitochondrial
`mono-oxygenase Coq7, which catalyzes the hydroxylation of
`demethoxyubiquinol (DMQ6), the penultimate reaction of ubiqui-
`none (CoQ6) biosynthesis (Tran et al., 2006). HPLC analysis of
`mitochondrial
`lipid extracts revealed diminished CoQ6 and
`increased DMQ6 levels upon Grx4 depletion (Figure 4C). This
`effect was reversed to wild-type ratios by expression of GRX4
`from a plasmid (Figure 4D). The simultaneous accumulation of
`the substrate (DMQ6) and decrease of the product (CoQ6) of
`the enzyme Coq7 indicates a diminished activity of this di-iron
`mono-oxygenase upon depletion of Grx3/4. Collectively, these
`
`data demonstrate that a Grx3/4 deficiency causes a severe
`defect in cellular di-iron enzymes.
`Are the observed defects in Grx3/4-depleted cells specific for
`iron-dependent proteins? This question was addressed by
`analyzing the activities and protein levels of several metal-
`dependent enzymes. The in-gel activities of the endogenous
`Cu/Zn- and Mn-dependent superoxide dismutases (Sod1 and
`Sod2,
`respectively)
`remained unchanged in Grx4-depleted
`Gal-GRX4 cells (Figure 4E). In marked contrast, both the level
`and activity of ectopically expressed iron-only superoxide
`dismutase (FeSod) from E. coli strongly declined. Furthermore,
`
`378 Cell Metabolism 12, 373–385, October 6, 2010 ª2010 Elsevier Inc.
`
`

`

`Cell Metabolism
`
`Role of Cytosolic Glutaredoxin in Iron Trafficking
`
`the Zn-dependent alcohol dehydrogenase (ADH) was 3-fold
`more active upon Grx4 depletion (Figure S1B). This increased
`ADH activity is characteristic for a switch toward fermentative
`metabolism and is typically observed upon iron deprivation.
`The normal function of several metal-reliant enzymes indicates
`that the described defects in Grx3/4-depleted cells are specific
`for iron-related processes. We conclude that Grx3/4 perform
`an essential
`role in cellular
`iron trafficking,
`in addition to
`and independently of their nonessential function in iron uptake
`regulation.
`
`Grx3/4 Assemble a Bridging Fe/S Cluster Independently
`of the CIA Machinery
`For a function of monothiol glutaredoxins in cellular iron traf-
`ficking, iron binding may be a necessary prerequisite. Recent
`in vitro studies have shown that various glutaredoxins, including
`yeast Grx3/4, can bind a bridging, GSH-liganded [2Fe-2S]
`cluster (Bandyopadhyay et al., 2008; Berndt et al., 2007; Johans-
`son et al., 2007; Li et al., 2009). We asked whether this unusual
`Fe/S cofactor is of physiological relevance and can be observed
`in a native environment using the 55Fe radiolabelling assay.
`Significant amounts of 55Fe were coimmunoprecipitated with
`anti-Grx4-antibodies from cell extracts derived from wild-type
`cells (strain BY4742), whereas only background levels were
`observed in grx3/4D cells (Figure 5A). Similar amounts of 55Fe
`were coimmunoprecipitated from both grx3D and grx4D cells,
`indicating that the homologous Grx3 and Grx4 bind iron with
`similar efficiency and independently of each other. The amount
`of Grx4-bound 55Fe was unchanged both in wild-type cells of
`our standard strain W303-1A, and, remarkably, under anaerobic
`or oxidative stress conditions prevailing after addition of H2O2 or
`upon deletion of SOD1 (sod1D cells), indicating that iron binding
`to Grx3/4 was insensitive to oxidative stress (Figure 5B).
`To analyze whether the iron bound to Grx3/4 is part of an
`Fe/S cluster, we investigated the requirement of 55Fe binding
`for components of Fe/S protein biogenesis (Lill and Mu¨ hlenhoff,
`2008). We first analyzed the role of the cysteine desulfurase
`complex Nfs1-Isd11, which serves as the sulfur donor for
`Fe/S protein biogenesis. For regulated depletion of Nfs1 or
`Isd11, we used Gal-NFS1 or Gal-ISD11 cells, respectively.
`55Fe binding to Grx4 (Figure 5C) and to the cytosolic Fe/S
`protein Leu1 as a control (Figure S4) declined to background
`levels upon depletion of either Nfs1 or Isd11. The same result
`was seen in Gal-ISU1Disu2 cells depleted for the core ISC
`scaffold proteins Isu1/2. No recovery of iron binding to Grx3/
`4 was observed when a cytosolic-nuclear version of Nfs1
`(DN-Nfs1) was produced in Gal-NFS1 cells lacking mitochon-
`drial Nfs1 (Figure 5C). We found during these experiments
`that the Grx4 protein levels decreased upon Nfs1 depletion,
`whereas the Grx3 levels remained unchanged. Although the
`reason for this specific decrease is unknown, we note that
`the remaining Grx3 did not bind 55Fe above background levels,
`unlike in grx4D cells (Figure 5A). The dependence of
`iron
`binding to Grx3/4 on core components of the mitochondrial
`ISC assembly system demonstrates that Grx3/4 bind an Fe/S
`cluster in vivo.
`Does incorporation of the Fe/S cluster into Grx3/4 also require
`the cytosolic iron-sulfur protein assembly (CIA) system (Lill,
`2009)? Depletion of the essential CIA components Nbp35 and
`
`Dre2 in the respective GAL promoter-exchange mutants signifi-
`cantly increased rather than impaired 55Fe binding to Grx3/4
`(Figure 5D), whereas hardly any 55Fe binding was observed to
`Leu1 as a control (Figure S4). This surprising finding suggests
`that incorporation of the Fe/S cluster into Grx3/4, although
`requiring mitochondrial Nfs1-Isd11-Isu1, does not depend on
`the cytosolic CIA machinery.
`The noncanonical pathway used for Fe/S cluster assembly on
`Grx3/4 may be due to the special nature of this cofactor, with
`a conserved Cys and GSH serving as ligands of a [2Fe-2S]
`cluster bridged between two Grx monomers, as observed after
`in vitro reconstitution or after overexpression in E. coli (Li et al.,
`2009; Picciocchi et al., 2007). We therefore sought to obtain
`in vivo evidence for this configuration. First, we determined
`which ligands might coordinate the Fe/S cluster. The importance
`of the conserved active-site cysteine residues C171 in the
`C-terminal Grx domain and C37 in the N-terminal Trx domain
`(Figure 5E, top) for 55Fe binding was tested in wild-type cells
`expressing a plasmid-encoded Myc-tagged Grx4 (Grx4-Myc)
`in which these residues were mutated to serine (mutant proteins
`C171S and C37S) or alanine (C171A). Myc-tagged wild-type
`Grx4 was fully functional
`(see below) and was produced at
`similar levels as the mutant proteins (Figure 5E, bottom). Mutant
`proteins C171S and C171A did not bind 55Fe above background
`levels (Figure 5E, middle). In contrast, mutant protein C37S
`bound wild-type amounts of 55Fe, suggesting that iron is bound
`via C171. Second, the importance of GSH for 55Fe binding to
`Grx3/4 in vivo was tested using the GSH synthesis-deficient
`mutant gsh1D that can be depleted for GSH upon growth in
`media lacking GSH. Although significant amounts of 55Fe were
`bound to Grx4-Myc in the presence of exogenously added
`GSH, only background levels of 55Fe were found in GSH-
`deprived cells (Figure 5F). Together, these findings suggest
`that the active-site C171 of the Grx domain and GSH serve as
`ligands of the Grx3/4-bound Fe/S cluster.
`Finally, we tried to find in vivo evidence for the presence of
`a bridging Fe/S cluster on Grx3/4. A yeast strain was constructed
`that simultaneously expressed C-terminally HA- and Myc-
`tagged Grx4. Cell extracts were subjected to immunoprecipita-
`tion with anti-HA or anti-Myc immunobeads followed by
`immunoblotting. Immunoprecipitation with anti-HA antibodies
`led to coisolation of Grx4-HA and a smaller amount of Grx4-
`Myc (Figure 5G). The same result, yet with inversed intensities,
`was observed using anti-Myc beads, whereas no cross-reacting
`bands were visible in wild-type cells. In cells expressing a C171S
`mutant Grx4-Myc and wild-type Grx4-HA, coimmunoprecipita-
`tion was far less efficient but still detectable. These observations
`document the importance of residue C171 for efficient Grx4
`dimer formation and thus are consistent with the idea of
`a bridging Fe/S cluster between two Grx monomers. Collec-
`tively, our findings suggest that, under physiological conditions,
`Grx3/4 bind a bridging Fe/S cluster that is coordinated by the
`active-site cysteine and GSH. These results are consistent with
`the structure of glutaredoxins reconstituted in vitro.
`
`The Grx3/4-Bound Fe/S Cluster Is Important for Iron
`Metabolism
`Is the Grx3/4-bound Fe/S cluster important for the function of
`Grx3/4 in iron trafficking and is this cofactor also required for
`
`Cell Metabolism 12, 373–385, October 6, 2010 ª2010 Elsevier Inc. 379
`
`

`

`Role of Cytosolic Glutaredoxin in Iron Trafficking
`
`Cell Metabolism
`
`380 Cell Metabolism 12, 373–385, October 6, 2010 ª2010 Elsevier Inc.
`
`

`

`Cell Metabolism
`
`Role of Cytosolic Glutaredoxin in Iron Trafficking
`
`iron sensing? As shown above, the C171S and C171A Grx4-Myc
`mutant proteins have lost the ability to stably bind the Fe/S cluster
`(Figure 5E). Grx4-depleted Gal-GRX4 cells expressing these
`mutant proteins from the endogenous GRX4 promoter failed to
`grow, whereas wild-type and C37S Grx4-Myc that retained
`normal
`iron binding supported wild-type growth (Figure 6A,
`top). Aft1-dependent transcription of FET3 was fully activated,
`and virtually no aconitase and catalase activities were observed
`in Gal-GRX4 cells producing C171S and C171A Grx4-Myc
`instead of wild-type Grx4 (Figure 6B). In contrast, wild-type and
`C37S Grx4-Myc-expressing Gal-GRX4 cells showed wild-type
`signals in these assays. All Grx4-Myc proteins were present at
`similar levels, with the exception of C171A, which apparently
`was less stable (Figure 6A, bottom). These results demonstrate
`the crucial role of residue C171 and thus the Grx3/4-bound
`Fe/S cluster for both iron regulation and iron trafficking.
`Formally, it is possible that the active-site C171 performs its
`essential role in Grx4 via thiol-dependent redox chemistry
`(Herrero and de la Torre-Ruiz, 2007; Lillig et al., 2008) rather
`than by coordination of the Fe/S cluster. Such a function can
`be excluded from the observation that overexpression (from
`a Tet promoter) of the C171S (but not C171A) Grx4-Myc mutant
`protein in Grx4-depleted Gal-GRX4 cells restored wild-type
`growth (Figure 6C). We noted that Grx4 overexpression generally
`diminished FET3 induction (Figure 6D). The corresponding
`C171S cells still displayed strong FET3 activation and wild-
`type aconitase, and strongly increased catalase activities. The
`restored iron loading of these and

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket