+ All Categories
Home > Documents > Retromer-mediated direct sorting is required for proper ... · Divalent metal transporter 1 (DMT1...

Retromer-mediated direct sorting is required for proper ... · Divalent metal transporter 1 (DMT1...

Date post: 29-Sep-2020
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
11
756 Research Article Introduction The steady-state localization of transmembrane (TM) proteins in the endocytic system is a consequence of many sorting events that occur at various points throughout the endosomal pathway. The sorting decisions are governed by a complex system of sorting signals in the itinerant proteins and molecular machineries that recognize those signals and deliver the proteins to their intended destinations (Bonifacino and Traub, 2003; Seaman, 2008). In general, sorting signals are located in the cytoplasmic domain of TM proteins, and some TM proteins have two or more sorting signals in their individual cytoplasmic domains, which allow them a more complex and unique itinerary in the endosomal pathway. For instance, the cation-independent mannose-6-phosphate receptor (CI-MPR), which sorts lysosomal hydrolase precursors to lysosomes, shuttles between the trans-Golgi network (TGN) and endosomes by the actions of several different sorting mechanisms (Ghosh et al., 2003; Seaman, 2005; Bonifacino and Hurley, 2008). At the TGN, CI-MPR binds lysosomal acid hydrolase and is then recognized by the Golgi-associated, -adaptin-ear-containing ARF- binding (GGA) proteins, via the acidic cluster dileucine signal located in its cytoplasmic domain, and sorted into clathrin-coated vesicles for delivery to the endosomes (Puertollano et al., 2001; Takatsu et al., 2001; Zhu et al., 2001). After delivery to the endosomes, CI-MPR dissociates from its ligand and is recognized by the retromer complex, via the aromatic motif located in its cytoplasmic domain, and is then retrieved to the TGN for further rounds of lysosomal hydrolase sorting (Arighi et al., 2004; Carlton et al., 2004; Seaman, 2004; Seaman, 2007). In addition to the sorting signals for GGAs and the retromer, the cytoplasmic domain of CI- MPR has several other sorting signals for substances such as PACS1, TIP47 and the clathrin adaptor complexes AP1 and AP2 (Ghosh et al., 2003). Thus, intracellular traffic of CI-MPR is regulated by several different sorting signals coupled with each sorting mechanism, which ensures its proper recycling between the TGN and the endosomes. Unlike single-pass TM proteins such as CI- MPR, the multipass TM proteins, such as nutrient transporters, have several cytoplasmic domains that make it more difficult to understand the molecular mechanisms for their sorting events in the endosomal pathway. Divalent metal transporter 1 (DMT1 or SLC11A2, formerly called NRAMP2/DCT1) is an integral membrane protein consisting of 12 predicted TM domains with two potential N-glycosylation sites; both of its N- and C-terminal tail domains face the cytosol (Gunshin et al., 1997). DMT1 has been shown to transport a number of divalent metals including Fe 2+ , Mn 2+ , Co 2+ , Cu 2+ , Ni 2+ , Pb 2+ , Zn 2+ and Cd 2+ by a proton cotransport mechanism (Gunshin et al., 1997). A mutation in Dmt1 (G185R) has been identified in microcytic anemia (mk) mice and Belgrade (b) rats (Fleming et al., 1997; Fleming et al., 1998), which have significant defects in intestinal iron absorption and assimilation of iron by erythroid precursor cells, indicating that the protein has lost the iron transport function in those tissues of the mutant animals. Indeed, DMT1 localizes at the brush border of duodenal enterocytes (Canonne- Hergaux et al., 1999), where it is responsible for dietary iron absorption across the apical plasma membrane, and in the endocytic compartments of cells of peripheral tissues (Su et al., 1998; Gruenheid et al., 1999; Tabuchi et al., 2000; Tabuchi et al., 2002), where it is responsible for the transport across endosomal membranes of iron internalized via the transferrin (Tf)-transferrin receptor (TfR) complex. Dmt1 produces at least two distinct mRNAs by alternative splicing of two 3 exons encoding different 3 untranslated regions Retromer-mediated direct sorting is required for proper endosomal recycling of the mammalian iron transporter DMT1 Mitsuaki Tabuchi* ,‡ , Izumi Yanatori, Yasuhiro Kawai and Fumio Kishi Department of Molecular Genetics, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan *Present address: Department of Life Science, Faculty of Agriculture, Kagawa University, Miki-cho, Kagawa 761-0795, Japan Authors for correspondence ([email protected]; [email protected]) Accepted 12 December 2009 Journal of Cell Science 123, 756-766 © 2010. Published by The Company of Biologists Ltd doi:10.1242/jcs.060574 Summary Endosomal recycling of the mammalian iron transporter DMT1 is assumed to be important for efficient and rapid uptake of iron across the endosomal membrane in the transferrin cycle. Here, we show that the retromer, a complex that mediates retrograde transport of transmembrane cargoes from endosomes to the trans-Golgi network, is required for endosomal recycling of DMT1-II, an alternative splicing isoform of DMT1. Bacterially expressed Vps26-Vsp29-Vsp35 trimer, a retromer cargo recognition complex, specifically binds to the cytoplasmic tail domain of DMT1-II in vitro. In particular, this binding is dependent on a specific hydrophobic motif of DMT1- II, which is required for its endosomal recycling. DMT1-II colocalizes with the Vps35 subunit of the retromer in TfR-positive endosomes. Depletion of the retromer by siRNA against Vps35 leads to mis-sorting of DMT1-II to LAMP2-positive structures, and expression of siRNA-resistant Vps35 can rescue this effect. These findings demonstrate that the retromer recognizes the recycling signal of DMT1- II and ensures its proper endosomal recycling. Key words: Endosome, Retromer, Recycling, DMT1, Iron metabolism, Transferrin cycle Journal of Cell Science
Transcript
Page 1: Retromer-mediated direct sorting is required for proper ... · Divalent metal transporter 1 (DMT1 or SLC11A2, formerly called NRAMP2/DCT1) is an integral membrane protein consisting

756 Research Article

IntroductionThe steady-state localization of transmembrane (TM) proteins inthe endocytic system is a consequence of many sorting events thatoccur at various points throughout the endosomal pathway. Thesorting decisions are governed by a complex system of sortingsignals in the itinerant proteins and molecular machineries thatrecognize those signals and deliver the proteins to their intendeddestinations (Bonifacino and Traub, 2003; Seaman, 2008). Ingeneral, sorting signals are located in the cytoplasmic domain ofTM proteins, and some TM proteins have two or more sortingsignals in their individual cytoplasmic domains, which allow thema more complex and unique itinerary in the endosomal pathway.For instance, the cation-independent mannose-6-phosphate receptor(CI-MPR), which sorts lysosomal hydrolase precursors tolysosomes, shuttles between the trans-Golgi network (TGN) andendosomes by the actions of several different sorting mechanisms(Ghosh et al., 2003; Seaman, 2005; Bonifacino and Hurley, 2008).At the TGN, CI-MPR binds lysosomal acid hydrolase and is thenrecognized by the Golgi-associated, -adaptin-ear-containing ARF-binding (GGA) proteins, via the acidic cluster dileucine signallocated in its cytoplasmic domain, and sorted into clathrin-coatedvesicles for delivery to the endosomes (Puertollano et al., 2001;Takatsu et al., 2001; Zhu et al., 2001). After delivery to theendosomes, CI-MPR dissociates from its ligand and is recognizedby the retromer complex, via the aromatic motif located in itscytoplasmic domain, and is then retrieved to the TGN for furtherrounds of lysosomal hydrolase sorting (Arighi et al., 2004; Carltonet al., 2004; Seaman, 2004; Seaman, 2007). In addition to the sortingsignals for GGAs and the retromer, the cytoplasmic domain of CI-MPR has several other sorting signals for substances such as PACS1,TIP47 and the clathrin adaptor complexes AP1 and AP2 (Ghosh et

al., 2003). Thus, intracellular traffic of CI-MPR is regulated byseveral different sorting signals coupled with each sortingmechanism, which ensures its proper recycling between the TGNand the endosomes. Unlike single-pass TM proteins such as CI-MPR, the multipass TM proteins, such as nutrient transporters, haveseveral cytoplasmic domains that make it more difficult tounderstand the molecular mechanisms for their sorting events inthe endosomal pathway.

Divalent metal transporter 1 (DMT1 or SLC11A2, formerlycalled NRAMP2/DCT1) is an integral membrane protein consistingof 12 predicted TM domains with two potential N-glycosylationsites; both of its N- and C-terminal tail domains face the cytosol(Gunshin et al., 1997). DMT1 has been shown to transport a numberof divalent metals including Fe2+, Mn2+, Co2+, Cu2+, Ni2+, Pb2+,Zn2+ and Cd2+ by a proton cotransport mechanism (Gunshin et al.,1997). A mutation in Dmt1 (G185R) has been identified inmicrocytic anemia (mk) mice and Belgrade (b) rats (Fleming et al.,1997; Fleming et al., 1998), which have significant defects inintestinal iron absorption and assimilation of iron by erythroidprecursor cells, indicating that the protein has lost the iron transportfunction in those tissues of the mutant animals. Indeed, DMT1localizes at the brush border of duodenal enterocytes (Canonne-Hergaux et al., 1999), where it is responsible for dietary ironabsorption across the apical plasma membrane, and in the endocyticcompartments of cells of peripheral tissues (Su et al., 1998;Gruenheid et al., 1999; Tabuchi et al., 2000; Tabuchi et al., 2002),where it is responsible for the transport across endosomalmembranes of iron internalized via the transferrin (Tf)-transferrinreceptor (TfR) complex.

Dmt1 produces at least two distinct mRNAs by alternativesplicing of two 3� exons encoding different 3� untranslated regions

Retromer-mediated direct sorting is required forproper endosomal recycling of the mammalian irontransporter DMT1Mitsuaki Tabuchi*,‡, Izumi Yanatori, Yasuhiro Kawai and Fumio Kishi‡

Department of Molecular Genetics, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan*Present address: Department of Life Science, Faculty of Agriculture, Kagawa University, Miki-cho, Kagawa 761-0795, Japan‡Authors for correspondence ([email protected]; [email protected])

Accepted 12 December 2009Journal of Cell Science 123, 756-766 © 2010. Published by The Company of Biologists Ltddoi:10.1242/jcs.060574

SummaryEndosomal recycling of the mammalian iron transporter DMT1 is assumed to be important for efficient and rapid uptake of iron acrossthe endosomal membrane in the transferrin cycle. Here, we show that the retromer, a complex that mediates retrograde transport oftransmembrane cargoes from endosomes to the trans-Golgi network, is required for endosomal recycling of DMT1-II, an alternativesplicing isoform of DMT1. Bacterially expressed Vps26-Vsp29-Vsp35 trimer, a retromer cargo recognition complex, specifically bindsto the cytoplasmic tail domain of DMT1-II in vitro. In particular, this binding is dependent on a specific hydrophobic motif of DMT1-II, which is required for its endosomal recycling. DMT1-II colocalizes with the Vps35 subunit of the retromer in TfR-positive endosomes.Depletion of the retromer by siRNA against Vps35 leads to mis-sorting of DMT1-II to LAMP2-positive structures, and expression ofsiRNA-resistant Vps35 can rescue this effect. These findings demonstrate that the retromer recognizes the recycling signal of DMT1-II and ensures its proper endosomal recycling.

Key words: Endosome, Retromer, Recycling, DMT1, Iron metabolism, Transferrin cycle

Jour

nal o

f Cel

l Sci

ence

Page 2: Retromer-mediated direct sorting is required for proper ... · Divalent metal transporter 1 (DMT1 or SLC11A2, formerly called NRAMP2/DCT1) is an integral membrane protein consisting

757Retromer-mediated sorting of DMT1

(UTRs) and protein products with distinct C-termini (Lee et al.,1998). The DMT1 isoform I (DMT1-I, +IRE) contains an iron-responsive element (IRE) in its 3� UTR and is expressed at thebrush border of duodenal enterocytes, where its expression isinduced by dietary iron deprivation (Canonne-Hergaux et al.,1999). DMT1 isoform II (DMT1-II, –IRE) lacks the IRE andencodes a protein that has a different C-terminal 25 amino acidsegment instead of the 18 amino acid segment of DMT1-I (see Fig.1A). DMT1-II is expressed preferentially in nonepithelial cells andis very abundant in reticulocytes. Its expression is induced by thehematopoietic hormone erythropoietin (Canonne-Hergaux et al.,2001). Recently, additional isoforms of Dmt1 mRNA have beenidentified based on alternative promoter usage at exon 1 (exon1Avs 1B) (Hubert and Hentze, 2002). This alternative promoter usageis assumed to produce a DMT1 protein bearing an additional 29amino acids (exon1A) upstream of the previously identified startcodon of DMT1-I and II (exon1B) (Hubert and Hentze, 2002). Therole of these additional residues in the function and targeting ofDMT1 has not yet been studied. However, recently, it has beenshown that DMT1-I expression from the exon1A promoter isactivated by hypoxia-inducible factor signaling induced in theduodenum following an acute iron deficiency (Shah et al., 2009).This indicates that DMT1-I, with an N-terminus derived fromexon1A, specifically functions in dietary iron absorption from theapical plasma membrane in the duodenum. These observationssuggest that the cytoplasmic tail domains of the N- and C-terminiof the DMT1 molecule are substituted by the alternative promoterusage and the alternative splicing, respectively, in a tissue-specificmanner, and that these substitutions adapt the DMT1 localizationto the tissue-specific iron acquisition (i.e. the dietary iron absorptionfrom the apical plasma membrane in the duodenum and endosomaliron acquisition in the Tf cycle).

Previously, we demonstrated that isoforms DMT1-I and DMT1-II, with their N-termini derived from exon1B, are targeted to distinctendosomal compartments. We also identified critical amino acidsin the cytoplasmic tail domain of DMT1-II as a determinant for theisoform-specific localization (Tabuchi et al., 2002). It has recentlybeen shown that this signal is required for endosomal recycling ofDMT1-II, and sorting of DMT1-II to the recycling pathway occursat the endosomes (Touret et al., 2003; Lam-Yuk-Tseung et al., 2005).In contrast to DMT1-II, DMT1-I is not efficiently recycled but issubsequently targeted to LAMP2-positive structures (Tabuchi et al.,2000; Tabuchi et al., 2002; Lam-Yuk-Tseung and Gros, 2006). Thesedata indicate that a putative sorting receptor specifically recognizesthe recycling signal of DMT1-II and ensures its endosomalrecycling. However, the molecular basis for the sorting of signal-dependent endosomal recycling of DMT1-II has yet to be elucidated.

In this study, we report a molecular mechanism underlying thesorting signal-dependent endosomal recycling of DMT1-II. Wefound that the retromer is required for proper endosomal recyclingof DMT1-II. We discuss the molecular mechanism for retromer-mediated sorting of DMT1-II to the recycling pathway and its roleon iron acquisition in the Tf cycle.

ResultsStructural requirements for endosomal recycling ofDMT1-IITo identify the structural requirements for the recycling signal ofDMT1-II, we performed a detailed mutational analysis of thecytoplasmic tail sequence. We expected that this approach mightreveal a conserved sequence motif by comparison with known

sorting signal sequences and then allow us to identify themolecule(s) required for endosomal recycling of DMT1-II. Weconstructed various GFP-tagged mutants of DMT1-II, which haddeletions or amino acid substitutions within the 25-amino acidsequence specific for DMT1-II, to narrow the region required forendosomal recycling of DMT1-II, and to identify the critical aminoacids and the structural requirements (Fig. 1A and listed in Fig.1G). These constructs were expressed in HEp-2 cells that wereimmunostained with antibodies against GFP and LAMP2, a markerfor organelles of the late endocytic pathway. Their localizations wereanalyzed by confocal microscopy. To measure the colocalizationof DMT1-II mutants with LAMP2, pixel-by-pixel analysis usingPearson’s correlation coefficient (Pc) was used and Pc with LAMP2(PcLAMP2) was calculated. A Pc value of 1 indicates perfectcolocalization. During the course of this analysis, we noticed thatthe localization of these mutants could be divided into three classesbased on the comparison between their images and PcLAMP2 counts(typical patterns of their localization and PcLAMP2 counts are shownin Fig. 1B,C): ‘class A’ mutants, similarly to DMT1-I, displayed apredominant localization in the perinuclear region of transfectedcells and significantly colocalized with LAMP2 (PcLAMP2≥0.8),indicating a severe defect of endosomal recycling of DMT1-II, astypified by the DMT1-II L557A mutant. Mutants classified as ‘classB’ displayed a dual localization in both the perinuclear region andin punctate structures diffusely scattered throughout the cytoplasm,and a partial colocalization of GFP signals with LAMP2 wasobserved exclusively in the perinuclear region (0.5<PcLAMP2<0.8),indicating a partial defect of endosomal recycling of DMT1-II, astypified by the DMT1-II Y555A mutant. ‘Class C’ mutants displayeda predominant localization in punctate structures scatteredthroughout the cytoplasm, and almost no colocalization of GFPsignals with LAMP2 (PcLAMP≤0.5) was observed, indicating normalendosomal recycling of DMT1-II, as typified by the DMT1-II wild-type. Thirty cells for each sample were examined and theirlocalizations were scored by the above criteria (Fig. 1D-F).

Deletion analysis of the cytoplasmic tail domain of DMT1-IImapped the most important element for endosomal recycling withinthe eight amino acid stretch 551-558 of DMT1-II (Fig. 1D,G).Alanine-scanning analysis in this region confirmed that Tyr555 andLeu557 are the crucial amino acids for endosomal recycling ofDMT1-II, which is in good agreement with our previous report(Tabuchi et al., 2002). Notably, the L557A mutant displayed a severemis-sorting of DMT1-II to LAMP2-positive structures, whereas theY555A mutant displayed a modest mis-sorting (PcLAMP2 L557A vsY555A0.86±0.024; n30 vs 0.71±0.126; n30, P<0.0001, Fig.1E), indicating that Leu557 is a more important residue for properendosomal recycling of DMT1-II than Tyr555. Deletion of theY555LL sequence led to a complete mis-sorting of DMT1-II toLAMP2-positive structures (PcLAMP20.91±0.018; n30, Fig. 1E),indicating that this mutation drastically reduces the ability ofendosomal recycling of DMT1-II. Although each single amino acidsubstitution to alanine within the Q551PEL sequence did not affectthe proper localization of DMT1-II, deletion of this region orsubstitution of all amino acids to alanine in this region led to a mis-sorting of DMT1-II to LAMP2-positive structures (PcLAMP20.63±0.107; n30, Fig. 1E). Importantly, deletion analysis indicatedthat the distance from the TM domain is also important for thissignal (Fig. 1E,G). The crucial amino acid Tyr555 must be morethan 15 amino acids away from the TM domain (Fig. 1G).

We next examined the structural requirements for the specificamino acid residues present at positions 555 and 557. Substitution

Jour

nal o

f Cel

l Sci

ence

Page 3: Retromer-mediated direct sorting is required for proper ... · Divalent metal transporter 1 (DMT1 or SLC11A2, formerly called NRAMP2/DCT1) is an integral membrane protein consisting

758

of Tyr555 with Ser led to a significant mis-sorting of DMT1-II toLAMP2-positive structures (PcLAMP20.86±0.043; n30, Fig. 1F),whereas substitution by Phe, Trp or Leu did not affect their

Journal of Cell Science 123 (5)

localizations (Fig. 1F,G), suggesting that any hydrophobic aminoacid at position 555, either aromatic or aliphatic, is sufficient forendosomal recycling of DMT1-II. Substitution of Leu557 by any

Fig. 1. Mutational analysis of the cytoplasmic tail domain of DMT1-II reveals the structural requirements for endosomal recycling of DMT1-II.(A)�Predicted secondary structure of DMT1. DMT1 is an integral membrane protein, which consists of 12 putative transmembrane domains and two potentialglycosylation sites. Two isoforms of DMT1 mRNA result from alternative splicing of a single gene product in mammalian cells. The two polypeptides (DMT1-Iand DMT-II) share 543 residues on the N-terminal portion but differ primarily in the last 18 or 25 C-terminal residues. (B)�Isoform-specific localization of DMT1.DMT1-I localizes in LAMP2-positive structures (class A), whereas DMT1-II localizes in maturing endosomes or recycling endosomes (TfR-positive endosomes)(class C). Some DMT1-II mutants show a dual localization of both TfR-positive endosomes and LAMP2-positive structures (class B). (C)�The localization of GFP-tagged DMT1-II was split into three classes. HEp-2 cells expressing GFP-DMT1-I (class A), GFP-DMT1-II Y555A (class B) or GFP-DMT1-II wild-type (class C)were fixed and immunostained with anti-GFP (green) and LAMP2 (red) antibodies. Scale bar: 20�m. Pixel-by-pixel colocalization analysis yielded a Pearson’scorrelation coefficient (Pc) with LAMP2 (PcLAMP2). The PcLAMP2 counts in each selected region are indicated. Pc1 indicates perfect colocalization.(D-F)�Quantification of the localization of various mutants of DMT1 in HEp-2 cell. Samples were inspected by confocal microscopy and the PcLAMP2 counts wereanalyzed (n30 cells per sample). Error bars represent s.d. The cut-off criteria for each class of localization were determined by the comparison of their images andPcLAMP2 counts. Class A, PcLAMP2≥0.8; Class B, 0.5<PcLAMP2<0.8; Class C, PcLAMP2≤0.5. (G)�Summary of the mutational analysis of the cytoplasmic tail domain ofDMT1-II. Amino acid sequences of the cytoplasmic tail domain of wild-type DMT1-I and DMT1-II and their various mutants, together with their class oflocalization. Based on the results from the mutational analysis of DMT1-II, we concluded that the sequence Øx(Leu/Met)x is a structural requirement for arecycling signal of DMT1-II (x indicates any amino acid; Ø indicates any hydrophobic amino acid).

Jour

nal o

f Cel

l Sci

ence

Page 4: Retromer-mediated direct sorting is required for proper ... · Divalent metal transporter 1 (DMT1 or SLC11A2, formerly called NRAMP2/DCT1) is an integral membrane protein consisting

759Retromer-mediated sorting of DMT1

hydrophobic amino acid except for Met led to a striking mis-sortingof DMT1-II to LAMP2-positive structures (Fig. 1F,G). On the basisof the detailed mutational analysis of the cytoplasmic tail domainof DMT1-II in this study, we conclude that a Øx(Leu/Met)x motif(where Ø can be any hydrophobic amino acid and x can be anyamino acid) is the recycling signal responsible for proper endosomalrecycling of DMT1-II.

Structural similarity between the recycling signal of DMT1-II and the retromer recognition sequence of CI-MPRTo find sequence motif(s) that are similar to the recycling signal ofDMT1-II, we investigated the known sorting motifs and found astructurally similar sequence motif WLM, which has been recentlyidentified in the CI-MPR tail as a sorting motif required for retromer-mediated endosome-to-TGN retrieval (Seaman, 2007). The retromerwas originally identified as an essential protein complex in yeastfor the proper retrieval of the vacuolar hydrolase receptor Vps10,a functional analog of mammalian CI-MPR, from endosomes backto the TGN (Seaman et al., 1998), and the mammalian retromerhas also been shown to function in endosome-to-TGN retrieval ofCI-MPR (Arighi et al., 2004; Carlton et al., 2004; Seaman, 2004).The mammalian retromer comprises a sorting nexin (SNX) dimercomposed of four possible combinations of SNX1-SNX5, SNX1-SNX6, SNX2-SNX5 and SNX2-SNX6 (Wassmer et al., 2009), anda cargo-recognition complex composed of Vps26, Vps29 and Vps35(Bonifacino and Hurley, 2008). Intriguingly, recent reports showedthat the retromer also functions in the endosomal recycling of severalTM proteins including the yeast iron transporter Fet3-Ftr1 complex(Strochlic et al., 2007) and Wntless, a membrane protein dedicatedto the secretion of the morphogen Wnt proteins (Eaton, 2008). Thesequence similarity and recent findings of the retromer function inmembrane protein recycling prompted us to examine the potentialinvolvement of the retromer in endosomal recycling of DMT1-II.We first swapped the YLL motif in the cytoplasmic tail domain ofDMT1-II for WLM and analyzed the localization of this mutant byimmunofluorescence confocal microscopy. This substitution did notaffect the proper localization of DMT1-II, indicating that the WLMmotif is sufficient for endosomal recycling, even in the case ofDMT1-II (Fig. 1F,G). Seaman (Seaman, 2007) has also reportedthe FLV motif in sortilin as a retromer recognition sequence.However, the FLV motif is not sufficient for endosomal recyclingin the case of DMT1-II (Fig. 1F,G).

Retromer directly interacts with the recycling signal ofDMT1-II in vitroWe next examined whether the recycling signal of DMT1-II coulddirectly bind to the retromer complex. Bacterially expressedglutathione S-transferase (GST)-DMT1-II tail domain (Fig. 2A)was used to pull down a bacterially expressed FLAG-taggedretromer cargo-recognition complex, which consists of 3�FLAG-Vps26-Vps29-Vps35-His6 (Fig. 2B). Since a previous studyshowed that the cytoplasmic tail domain of CI-MPR interacts withVps35 using a yeast two-hybrid assay (Arighi et al., 2004), weused the GST-CI-MPR tail (Fig. 2A) as a positive control.Immunoblot analysis showed that both GST-CI-MPR and GST-DMT1-II tails, but not GST alone, efficiently pulled down therecombinant retromer in a dose-dependent manner (Fig. 2C),showing that the cytoplasmic tail domain of DMT1-II can directlybind to the retromer. Strikingly, this interaction was almostcompletely abrogated by deletion of the YLL sequence from theDMT1-II tail domain (Fig. 2C), which leads to a complete mis-

sorting of DMT1-II to LAMP2-positive structures in HEp-2 cells(Fig. 1E,G).

Using the results of the in vivo mutagenesis experiments (Fig.1G), we next expressed GST-DMT1-II tail fusion proteins withnonfunctional mutations Y555A or L557A , and GST-DMT1-II tailfusion proteins with the functional mutation L557M. These proteinswere used for pull-down assays to investigate the binding specificityof the DMT1-II-cytoplasmic tail domain to the retromer. As shownin Fig. 2D, both the GST-DMT1-II tail Y555A and L557A mutantpulled down the retromer less efficiently than the GST-DMT1-IItail wild type (reduced to <65% compared with that of the wildtype). Notably, the GST-DMT1-II tail L557M mutant pulled downthe retromer more efficiently (increased to more than 125%compared with the wild type), indicating that this L557M mutanthas higher affinity to the retromer than the wild type. The bindingspecificity of the DMT1-II tail domain to the retromer in vitro waswell correlated with the structural requirements for the recyclingsignal of DMT1-II in cultured cells.

DMT1-II colocalization with the retromer in TfR-positiveendosomesWe next determined the subcellular localization of DMT1-II and theretromer. HEp-2 cells stably expressing 3�HA-tagged DMT1-II wereimmunostained with antibodies against HA epitope Vps35 andvarious organellar markers. A triple-staining experiment showed thatDMT1-II colocalized with Vps35 in relatively large punctae, whereTfR, a marker for organelles of the early endocytic and recyclingpathways, also colocalized with Vps35 (Fig. 3Ae-h, arrows). Toclarify the localization of DMT1-II and Vps35 double-positivepunctate-structures, antibody against EEA1, a marker for organellesof the early endocytic pathway, was used for triple staining insteadof TfR (Fig. 3B). Only the small patches of the DMT1-II and Vps35double-positive punctate structures overlapped with EEA1, whereasthe majority of the double-positive punctate structures werejuxtaposed to EEA1 and not colocalized with EEA1 (Fig. 3Be-h,arrows). Furthermore, these double-positive punctate structurescolocalized particularly with CI-MPR in the perinuclear region(supplementary material Fig. S1Ae-h, arrows) but not with TGNmarker p230 or recycling endosome marker Rab11 (supplementarymaterial Fig. S1B,C). These data showed that DMT1-II colocalizeswith the retromer in TfR-positive endosomes, whereas EEA1 ismostly excluded from this configuration. These results support thehypothesis that the retromer might regulate endosomal recycling ofDMT1-II at maturing endosomes (see Discussion).

Retromer depletion by RNAi leads to mis-sorting of DMT1-II to LAMP2-positive structuresTo examine directly whether the retromer is required for endosomalrecycling of DMT1-II, we knocked down VPS35 mRNA using smallinterfering RNA (siRNA). HEp-2 cells stably expressing 3�HA-tagged DMT1-II were transfected with two types of siRNAoligonucleotide pairs against VPS35: VPS35 siRNA#1 and #2. Aftertwo rounds of transfection at 72 hour intervals with each of thesesiRNAs, the protein expression of Vps35 decreased to undetectablelevels in both samples of siRNA-treated cells, which was confirmedby immunoblot analysis using anti-Vps35 antibody (Fig. 4A). Theprotein expression of DMT1-II was not significantly affected bydepletion of Vps35 (Fig. 4A). Notably, an additional band atapproximately 40 kDa was observed in Vps35-depleted cells butnot in control siRNA-treated cells (Fig. 4A, indicated with a singleasterisk).

Jour

nal o

f Cel

l Sci

ence

Page 5: Retromer-mediated direct sorting is required for proper ... · Divalent metal transporter 1 (DMT1 or SLC11A2, formerly called NRAMP2/DCT1) is an integral membrane protein consisting

760

We next examined the localization of DMT1-II in Vps35-depletedcells. We observed that DMT1-II colocalized with TfR but not withLAMP2 in control siRNA-treated cells (Fig. 4Ba-d and i-l,respectively). However, upon depletion of Vps35 by treatment withVPS35 siRNA#1, localization of DMT1-II changed dramatically. Weobserved almost no colocalization of DMT1-II with TfR in Vps35-depleted cells (Fig. 4Be-h). Instead, DMT1-II colocalized well withLAMP2 (Fig. 4Bm-p). To quantify this effect, we performed pixel-by-pixel colocalization analysis and Pc was calculated. Depletion ofVps35 caused a significant reduction of a PcTfR (Pc with TfR) (controlsiRNA vs VPS35 siRNA0.61±0.09; n50 vs 0.28±0.14; n50,

Journal of Cell Science 123 (5)

P<0.0001; Fig. 4C), and conversely a significant increase of PcLAMP2

(control siRNA vs VPS35 siRNA0.12±0.10; n50 vs 0.77±0.06;n50, P<0.0001; Fig. 4D). This analysis statistically confirmed thatDMT1-II was mis-sorted to LAMP2-positive structures in Vps35-depleted cells, indicating that endosomal recycling of DMT1-II wasperturbed by the loss of retromer function.

Phenotypic rescue of Vps35-depleted cells by expressionof siRNA-resistant Vps35To further confirm the retromer function in endosomal recyclingof DMT1-II, we monitored the rescue of this phenotype by

Fig. 2. Retromer complex binds to the cytoplasmic tail domain of DMT1-II. (A)�Schematic representation of the structure of GST-fusion proteins used for GSTpull-down assay. (B)�Schematic representation for the bacterial expression and purification of 3�FLAG-tagged retromer complex. (C)�Purified recombinantFLAG-tagged retromer complex (consisting of proteins, 3�FLAG-Vps26-Vps29-Vps35-His6) was incubated with purified GST or GST fusion proteins bearing thewild-type tails of CI-MPR or DMT1-II, or the tail with deletion of the YLL recycling motif of DMT1-II, as shown in Fig. 1G (GST-DMT1-IIYLL tail), and then theproteins were pulled down with glutathione-Sepharose 4B beads. Serial dilutions of proteins bound to the beads were resolved by SDS-PAGE. Bound FLAG-tagged retromer components were detected by immunoblotting with antibody to the FLAG-tag, and proteins were visualized by Coomassie blue staining.(D)�Recombinant retromer complex was tested for interaction with GST or GST fusion proteins bearing the wild-type tails or mutant tails of DMT1-II, as above.Interaction of the retromer complex with Y555A or L557A mutant tails of DMT1-II, whose full-length proteins are mis-sorted to LAMP2-positive strucures inHEp-2 cells shown in Fig. 1E, was significantly decreased compared with the wild-type tail of DMT-II, whereas interaction of the retromer complex with theL557M mutant tail of DMT1-II, whose full-length protein is normally sorted to TfR-positive endosomes, was notably increased compared with the wild-type tail ofDMT1-II. We observed less 3�FLAG-Vps35-His6 protein than 3�FLAG-Vps26, or 3�FLAG-Vps29 proteins, presumably because of an inefficient transfer of theprotein to the PVDF membrane or a partial proteolytical digestion of the N-terminal region during protein purification.

Jour

nal o

f Cel

l Sci

ence

Page 6: Retromer-mediated direct sorting is required for proper ... · Divalent metal transporter 1 (DMT1 or SLC11A2, formerly called NRAMP2/DCT1) is an integral membrane protein consisting

761Retromer-mediated sorting of DMT1

transfecting Vps35-depleted cells with GFP alone, or GFP-taggedsiRNA-resistant Vps35 wild-type or GFP-tagged nonfunctionalmutant Vps35 R107A (Gokool et al., 2007). The expression of thesiRNA-resistant Vps35 wild-type in Vps35-depleted cells wellrestored the mis-sorting of DMT1-II. GFP-Vps35-positive cells inVps35-depleted cells displayed almost no colocalization of DMT1-II with LAMP2 (Fig. 5Aa�-e�), but significant colocalization ofDMT1-II with TfR (supplementary material Fig. S2Aa�-e�). Tomeasure the colocalization of DMT1-II with LAMP2 in both GFP-Vps35-positive and -negative cells, pixel-by-pixel analysis was usedand Pc with LAMP2 was calculated. This analysis revealed almostno colocalization of DMT1-II with LAMP2 in GFP-Vps35-positivecells (PcLAMP20.24±0.16; n50; Fig. 5Aa�-e�), but did showsignificant colocalization in GFP-Vps35-negative cells (PcLAMP20.62±0.10; n50; Fig. 5Aa�-e�). These results confirmed thatexpression of GFP-Vps35 rescued the defect of endosomal recyclingof DMT1-II in endogenous Vps35-depleted cells.

In contrast to the wild-type GFP-Vps35, neither GFP alone norGFP-tagged nonfunctional mutant Vps35 R107A could rescue thisphenotype (supplementary material Fig. S2B,C). More than 90%of cells displayed colocalization of DMT1-II with LAMP2 in cellspositive for GFP alone or GFP-Vps35 R107A, whereas less than3% of cells displayed colocalization of DMT1-II with LAMP2 incells positive for wild-type GFP-Vps35 (Fig. 5B; supplementarymaterial Fig. S2C,D), confirming that the rescue of DMT1-II

localization by ectopic expression of siRNA-resistant Vps35 inVps35-depleted cells is due to the restoration of normal retromerfunction. Thus, these results demonstrate that the defect ofendosomal recycling of DMT1-II in Vps35-depleted cells is a directconsequence of loss of retromer function.

In conclusion, these results strongly suggest that DMT1-II isrecognized by a retromer cargo-recognition complex via itsrecycling signal at maturing endosomes, thereby becomingincorporated into tubular vesicles formed by the retromer and sortingDMT1-II to the recycling pathway.

DiscussionIn this study, we report a molecular mechanism underlying thesorting-signal-dependent endosomal recycling of DMT1-II. First,we identified the structural requirements for the recycling signalof DMT1-II and found the retromer to be a candidate moleculerequired for endosomal recycling of DMT1-II. Second, wedemonstrated that the retromer cargo-recognition complex coulddirectly bind to the cytoplasmic tail domain of DMT1-II in vitroin a recycling-signal-dependent manner. Third, we showed thatDMT1-II colocalized with a Vps35 subunit of the retromer in TfR-positive endosomes. Finally, we showed that depletion of Vps35by RNAi led to a complete mis-sorting of DMT1-II to LAMP2-positive structures, and ectopic expression of siRNA-resistantVps35 rescued this effect.

Fig. 3. DMT1-II and Vps35 colocalizes in TfR-positiveendosomes in HEp-2 cells. HEp-2 cells stably expressing a 3�HA-tagged DMT1-II were fixed, permeabilized, and immunostainedwith rabbit anti-HA pAb (Aa,Ba), mouse anti-TfR mAb (Ab) oranti-EEA1 mAb (Bb) and goat anti-human Vps35 pAb (Ac,Bc),followed by Alexa Fluor 555-conjugated donkey anti-rabbit IgG,Alexa Fluor 488-conjugated donkey anti-mouse IgG, and AlexaFluor 633-conjugated donkey anti-goat IgG. Cells were examinedby confocal microscopy. Merged images show colocalization of3�HA-DMT1-II with Vps35 (Ae,Be), 3�HA-DMT1 with TfR(Af) and Vps35 with TfR (Ag) in relatively large puncta (arrows),but almost no colocalization of 3�HA-DMT1-II with EEA1 (Bf)and Vps35 with EEA1 (Bg). Scale bars: 5�m.Jour

nal o

f Cel

l Sci

ence

Page 7: Retromer-mediated direct sorting is required for proper ... · Divalent metal transporter 1 (DMT1 or SLC11A2, formerly called NRAMP2/DCT1) is an integral membrane protein consisting

762

Cumulatively, these data argue that the retromer can directly bindand sort the DMT1-II molecule at maturing endosomes (Fig. 6B),thereby ensuring proper endosomal recycling of this molecule.

Retromer cargo-recognition mechanismDetailed mutational analysis of the cytoplasmic tail domain ofDMT1-II revealed the structural requirements for the recyclingsignal of DMT1-II and identified the Øx(Leu/Met)x motif as itsrecycling signal. This motif is also required for the binding of theDMT1-II-cytoplasmic tail domain to the retromer in vitro. Thesedata suggest that this motif represents a novel retromer-bindingmotif. So far, no strong consensus retromer-binding motifs havebeen identified, and none have been characterized at a structurallevel (Bonifacino and Hurley, 2008; Collins, 2008). The WLM motifof CI-MPR (Seaman, 2007) fits the structural requirement for the

Journal of Cell Science 123 (5)

recycling signal of DMT1-II and the swapping of the YLL motifin DMT1-II for the WLM motif did not affect its proper localizationin HEp-2 cells, showing that the WLM motif is sufficient forendosomal recycling of DMT1-II. Other known retromer-recognition sequences, such as FLV in sortilin (Seaman, 2007) orFxFxD in yeast DPAP-A (Nothwehr et al., 2000), are structurallysimilar, but the residues Val and Phe at amino acid position 2 inthese sequences do not fit the structural requirement for therecycling signal of DMT1-II (Fig. 6A). Futhermore, the FLV motifis not sufficient for endosomal recycling in the case of DMT1-II.We found that the upstream and downstream sequences of the crucialamino acids Y555LL were also important for the proper recyclingof DMT1-II (Fig. 1D,E,G). The upstream and downstreamsequences of the FLV motif in sortilin might also be important forthe efficient binding of this motif to the retromer.

Fig. 4. Mis-sorting of DMT1-II to LAMP2-positive structures upon retromer depletion.(A)�Depletion of Vps35 by RNAi is assessedby immunoblotting of each siRNA-treated celllysate. Tubulin is assumed to be unaffected bythe siRNA treatment and can serve as aloading control in this experiment. A singleasterisk indicates the protein bands found inVPS35-depleted cells, which presumablycorrespond to some degradation product of3�HA-DMT1-II protein. A double asteriskindicates the immature protein bands of3�HA-DMT1-II (Tabuchi et al., 2002).(B)�HEp-2 cells stably expressing a 3�HA-DMT1-II were transfected twice at 72 hourintervals with control siRNA or siRNAsdirected against human VPS35. At 24 hoursafter the second transfection, cells were seededon coverslips and cultured for an additional 48hours. Cells were fixed at 72 hours after thesecond transfection, double-labeled with ratanti-HA mAb (b,f,j,n) and mouse anti-TfRmAb (c,g) or mouse anti-LAMP2 mAb (k,o),followed by Alexa Fluor 555-conjugated goatanti-rat IgG and Alexa Fluor 633-conjugatedgoat anti-mouse IgG. Cells were examined byconfocal microscopy. Scale bar: 10�m.(C,D) Pixel-by-pixel colocalization analysisyielded a Pearson’s correlation coefficient withTfR (PcTfR) or LAMP2 (PcLAMP2). A total of50 cells were analyzed for each sample.Depletion of Vps35 significantly reducedPcTfR, and inversely increased PcLAMP2. Errorbars indicate s.d. *P<0.0001.

Jour

nal o

f Cel

l Sci

ence

Page 8: Retromer-mediated direct sorting is required for proper ... · Divalent metal transporter 1 (DMT1 or SLC11A2, formerly called NRAMP2/DCT1) is an integral membrane protein consisting

763Retromer-mediated sorting of DMT1

In addition to the known retromer-binding sequences, we foundstructurally similar sequences F746LLQ and Y532KLT in thecytoplasmic tail domains of human polymeric immunoglobulinreceptor (pIgR) and human Wntless, respectively, both of whichare known to be retromer cargo proteins (Verges et al., 2004; Yanget al., 2008). Mostov and colleagues have shown that the retromerdirectly binds to the cytoplasmic tail domain of pIgR and regulatestranscytosis of pIgR from the basolateral to the apical surface(Verges et al., 2004). Interestingly, they showed that a pIgR mutantlacking the 30 C-terminal residues of its cytoplasmic domain, whichcompletely lacks the F746LLQ sequence, impaired its retromer-binding ability. Furthermore, the same group recently found thatthe F746LLQ sequence of pIgR is sufficient to transcytose the pIgRfrom the basolateral to the apical surface (Luton et al., 2009). Thesedata suggest that the F746LLQ sequence functions as a retromer-binding sequence, which is in good agreement with our prediction.The relative contribution of the Y532KLT to retromer-dependentrecycling of human Wntless remains to be determined. Furtherexperiments are needed to define the consensus retromer-bindingmotif.

Endosomal sorting pathways for DMT1 isoformsEndosomes are highly dynamic organelles that comprise distinctsubdomains occupied by different Rab proteins within the samecontinuous membrane. For example, during endocytosis ofdegradative cargos, such as low-density lipoproteins to lysosomes,endosomes containing the cargos gradually mature from early tolate stages with a continuous conversion of Rab5 to Rab7 (Rink et

al., 2005). It has recently been reported that the retromer cargo-recognition complex is recruited onto the endosomal membrane bythe action of GTP-bound Rab7 (Rojas et al., 2008; Seaman et al.,2009). Based on our present results and these recent findings, weassume that the endosomal recycling pathway for DMT1-II is asfollows: Rab5-positive endosomes containing DMT1-II graduallymature by recruiting the GTP-bound Rab7, which further recruitsthe retromer cargo-recognition complex onto its endosomalmembrane (Fig. 6B). DMT1-II is subsequently recognized by theretromer cargo-recognition complex via its recycling signal andincorporated into the tubular vesicle formed by the retromer cargo-recognition complex and the BAR-domain-containing proteinsSNXs, which stimulate membrane-curvature formation (McMahonand Gallop, 2005). DMT1-II is then transported to the TGN beforeendosomes are completely matured. At the TGN, DMT1-II istransported via the recycling endosome en route to the plasmamembrane. Since DMT1-I cannot be recognized by the retromer,this isoform cannot escape from maturing endosomes and isconsequently transported to LAMP2-positive structures.

Although depletion of the retromer by treatment with VPS35siRNA caused a complete mis-sorting of DMT1-II to LAMP2-positive structures, we did not observe any significant reduction ofthe DMT1-II protein level in Vps35-depleted cells. Nevertheless,we did observe a faint band at ~40 kDa, which presumablycorresponds to a degradation product of DMT1-II (Fig. 3A). Inaddition, Vps35-depleted cells treated with cycloheximide for 12hours displayed a DMT1-II protein level that was comparable withthat in control cells treated with ethanol for 12 hours (supplementary

Fig. 5. Rescue of DMT1-II localization by expression of siRNA-resistant Vps35. (A)�Cells transfected withVPS35 siRNA#1 followed by transfection of plasmid encoding VPS35 siRNA#1-resistant GFP-tagged VPS35 wildtype were fixed, immunostained with rabbit anti-GFP pAb, rat anti-HA mAb and mouse anti-LAMP2 mAbfollowed by Alexa Fluor 488-conjugated goat anti-rabbit IgG, Alexa Fluor 555-conjugated goat anti-rat IgG andAlexa Fluor 633-conjugated goat anti-mouse IgG. Cells were examined by confocal microscopy. Scale bars:10�m. PcLAMP2 in GFP-positive cells0.24±0.16 (n50), PcLAMP2 in GFP-negative cells0.62±0.10 (n50).(B)�Quantification of the rescue of DMT1-II mislocalization to LAMP2-positive structures in Vps35-depleted cellsafter transfection with GFP alone, or siRNA-resistant Vps35 wild type or R107A mutant. Samples were inspectedby confocal microscopy and scored for colocalization of DMT1-II with LAMP2. The average counts werecalculated from three independent experiments, each analyzing at least 45 cells per condition. Errors bars indicates.d. *P<0.0001.

Jour

nal o

f Cel

l Sci

ence

Page 9: Retromer-mediated direct sorting is required for proper ... · Divalent metal transporter 1 (DMT1 or SLC11A2, formerly called NRAMP2/DCT1) is an integral membrane protein consisting

764

material Fig. S3), showing that the turnover of DMT1-II protein isvery slow at LAMP2-positive structures. These data indicate thata small amount of DMT1-II is degraded in lysosomes, presumablythrough sorting into a multivesicular body pathway (Katzmann etal., 2002), but the majority of DMT1-II is probably transported tothe limiting membrane of LAMP2-positive structures in Vps35-depleted cells. Likewise, DMT1-I must be transported to thelimiting membrane of LAMP2-positive structures, where it couldfunction as an iron transporter (Fig. 6B).

Role for retromer-mediated endosomal recycling of DMT1-II in the Tf cycleIn the Tf cycle, DMT1-II and TfR are functionally coupled tooperate pH-dependent iron uptake across the endosomal membrane,and both molecules are recycled between the plasma membrane

Journal of Cell Science 123 (5)

and endosomes (Andrews, 1999; Touret et al., 2003; Andrews,2008). However, the molecular mechanisms underlying theirrecycling from endosomes to the plasma membrane are apparentlydistinct. DMT1-II is directly recognized by the retromer cargo-recognition complex and transported through the TGN en route tothe recycling endosome, whereas TfR is directly transported to therecycling endosome by a retromer-independent mechanism (Traeret al., 2007), and subsequently both molecules are recycled backto the plasma membrane for another round of the Tf cycle (Fig.6B). Since endosomes are highly dynamic organelles forming acontinuum of maturing organelles from early to late stages(Bonifacino and Rojas, 2006), the retromer-mediated dynamicrecycling system must be important for retention of the irontransporter at TfR-positive endosomes to facilitate the endosomaliron uptake in the Tf cycle, which is particularly important for theproduction of hemoglobin in mitochondria in erythroid precursors(Fig. 6B). Further studies are required to gain a better understandingof the relative contribution of retromer-mediated endosomalrecycling of DMT1-II on endosomal iron uptake in the Tf cycle.

Materials and MethodsAntibodies and reagentsWe used mouse or rat monoclonal antibodies (mAbs) against the following proteinsor epitope tags: EEA1, p230 and Rab11 (BD Biosciences); CI-MPR (2G11, AffinityBioReagentsTM); LAMP2 (H4B4, Developmental Studies Hybridoma Bank); -tubulin (GE Healthcare); HA tag (mouse mAb HA.11, Covance; rat mAb 3F10, RocheApplied Science); FLAG tag (M2, Sigma); TfR [N2, provided by Tamotsu Yoshimori,Osaka University, Osaka, Japan (Yoshimori et al., 1988)]. A rabbit polyclonal antibody(pAb) to the FLAG tag was obtained from Sigma, and one to the HA tag was obtainedfrom MBL, Japan. A goat pAb to Vps35 was obtained from Imgenex. We raised arabbit pAb against the GFP. A rabbit was immunized with a maltose-binding protein-GFP fusion protein, and the anti-GFP antiserum was affinity purified using a GST-GFP fusion-protein-immobilized column. Goat anti-rabbit IgG or donkey anti-mouseIgG conjugated with Alexa Fluor 488, goat anti-rat IgG or donkey anti-rabbit IgGconjugated with Alexa Fluor 555, and goat anti-mouse IgG or donkey anti-goat IgGconjugated with Alexa Fluor 633 were purchased from Invitrogen. Horseradish-peroxidase-conjugated anti-mouse and anti-rabbit IgG were purchased from Cellsignaling technology. Horseradish-peroxidase-conjugated anti-goat IgG was purchasedfrom R&D systems. Most of the other general reagents were from Wako chemicals,Nacalai Tesque or Sigma.

Recombinant DNA proceduresTo introduce the mutations into the cytoplasmic domain of DMT1-II, the pUC13-DMT1-II plasmid (Tabuchi et al., 2002) was used for site-directed mutagenesis tocreate the deletion or alanine-scanning mutants. Site-directed mutagenesis wasperformed by the inverse PCR method (Imai et al., 1991). The plasmids carrying mutantconstructs were prepared from the Escherichia coli strain SCS110 (Stratagene) anddigested with BclI. The BclI fragments containing the mutated constructs of DMT1-II were ligated into the BamHI site of pIRES neo-GFP plasmid (Clontech) to generatethe mammalian expression plasmids carrying GFP-tagged DMT1-II mutants.

To generate plasmids encoding GFP or FLAG-tagged VPS26, VPS29 and VPS35,DNA encoding full-length human Vps26, Vps29 and Vps35 was amplified by PCRusing KOD plus DNA polymerase (Toyobo). Each amplified fragment was clonedinto pDONR221 by BP reaction using GatewayTM technology (Invitrogen) togenerate entry clones for VPS26, VPS29, and VPS35. Each entry clone was transferredinto pIRES neo-GFP or 3�FLAG-based Gateway vectors by LR reaction to generatepIRES neo-GFP or 3�FLAG-VPS26, -VPS29 and -VPS35 plasmids.

To generate a plasmid encoding VPS35 siRNA#1-resistant wild type of VPS35,pDONR221-siRNAR-VPS35WT, inverse PCR was performed with primers (theforward primer: 5�-GTA GTT AGC TTA CAA GTC TCT-3�; the reverse primer: 5�-GTC TTC ACT AGG CAT GTC TTG-3�; substituted nucleotides without an aminoacid change are underlined) corresponding to the sequence of VPS35 siRNA#1 usingthe pDONR221-VPS35 plasmid as a template, and the PCR product was self-ligatedwith T4 DNA ligase and T4 polynucleotide kinase (see supplementary material Fig.S4 for further details). The resulting clone was transferred into a pIRES neo-GFP-based Gateway vector by LR reaction to generate a pIRES neo-GFP-siRNAR-VPS35WT

plasmid.To generate a plasmid expressing the GST-CI-MPR tail fusion protein in E. coli,

the fragment containing the region encompassing residues 2328-2491 of the humanCI-MPR was amplified by PCR from a human skin fibroblast cDNA library. Theamplified fragment was digested with BamHI and XhoI, and the BamHI/XhoI fragmentwas cloned into the BamHI-XhoI sites of pGEX-5X-1 (GE Healthcare) to generatea pGEX-5X-1-CI-MPR tail plasmid.

Fig. 6. A model for retromer-dependent endosomal recycling.(A)�Representative retromer-binding motifs. (B)�Schematic diagram of the roleof the retromer for endosomal recycling of DMT1-II in the Tf cycle. DMT1-IIand TfR are functionally coupled to operate pH-dependent iron uptake acrossthe endosomal membrane, and both molecules are recycled between theplasma membrane and endosomes. TfR is directly transported to the recyclingendosome, whereas DMT1-II is recognized by the retromer cargo-recognitioncomplex and transported through the TGN en route to the recycling endosome;subsequently, both molecules are recycled back to the plasma membrane foranother round of the Tf cycle.

Jour

nal o

f Cel

l Sci

ence

Page 10: Retromer-mediated direct sorting is required for proper ... · Divalent metal transporter 1 (DMT1 or SLC11A2, formerly called NRAMP2/DCT1) is an integral membrane protein consisting

765Retromer-mediated sorting of DMT1

To generate plasmids expressing fusion proteins of GST-DMT1-II tail wild-typeor various mutants in E. coli, the fragment containing the region encompassing residues533-568 of human DMT1-II was amplified by PCR from pUC13-DMT1-II plasmidscontaining a wild-type or mutant version of DMT1-II, as described above. Theamplified fragments were digested with EcoRI and XhoI, and the EcoRI-XhoIfragments were cloned into the EcoRI-XhoI sites of pGEX 5X-1 to generate plasmidsof pGEX-5X-1-DMT1-II tail wild type or various mutants.

To generate a plasmid expressing the 3�FLAG-tagged retromer complex in E.coli, DNA fragments encoding 3�FLAG-tagged full-length molecules of each Vps26,Vps29 and Vps35 with a Shine-Dalgarno sequence at 5�-upstream of each ORF, wereamplified by PCR from pIRES neo-3�FLAG-VPS26, -VPS29 and -VPS35 plasmids,respectively. The amplified fragments were sequentially cloned into pET23d plasmidin the order of 3�FLAG-VPS26, 3�FLAG-VPS29 and then 3�FLAG-VPS35 togenerate a pET23d-3�FLAG-Retromer plasmid. 3�FLAG-Vps35 was fused to a C-terminal His6 tag.

The cloned regions of all plasmids were completely sequenced to ensure thatunplanned mutations were not introduced during the cloning procedures.

Protein expression and purificationFor purification of the GST-fusion protein, Rosseta-gamiB (DE3) cells (EMD)transformed with protein expression plasmids were grown in Luria broth with 100g/ml ampicillin at 37°C to an OD600 of 0.8, and then protein expression was inducedby the addition of isopropyl-1-thio--D-galactopyranoside (IPTG) to 0.3 mM for 3hours at 37°C. Cell pellets were resuspended in buffer A (0.5% Triton X-100 in PBS)containing 1 mM phenylmethylsulfonyl fluoride (PMSF) and 1 mg/ml lysozyme, anddisrupted by sonication. Lysates were cleared by centrifugation at 20,000 g for 30minutes at 4°C and GST-tagged proteins were purified from the supernatant usingglutathione-Sepharose 4B beads (GE Healthcare). After incubation for 2 hours withthe beads, the beads were washed five times with buffer A, and then proteins wereeluted with G buffer (10 mM reduced glutathione, 50 mM Tris-HCl, pH 9.0). Proteinswere subsequently dialyzed in PBS and protein concentrations were determined usinga micro BCA protein assay kit (Pierce).

For the purification of the FLAG-tagged retromer complex, Rosseta-gami2 (DE3)pLys cells (EMD) transformed with pET23d-3�FLAG-Retromer plasmid were grownin Terrific broth (Invitrogen) with 34 g/ml chloramphenicol, 50 g/ml streptomycin,12.5 g/ml tetracycline, and 100 g/ml ampicillin at 37°C to an OD600 of 0.8. Thebacteria were then moved to 16°C for 30 minutes and protein expression was inducedby the addition of IPTG to 0.3 mM for 19 hours. The cell pellet was resuspended inbuffer B (50 mM NaH2PO4, 300 mM NaCl, 5 mM imidazole, pH 8.0) containingcomplete EDTA-free protease inhibitor cocktail (Roche Molecular Biochemicals), 1mM PMSF and 1 mg/ml lysozyme, and disrupted by sonication. The lysate was clearedby centrifugation at 20,000 g for 30 minutes at 4°C and the cleared lysate was appliedto the Ni-NTA agarose column (Qiagen) equilibrated with buffer B. The column wasthen washed with 10 column volumes of wash buffer (50 mM NaH2PO4, 300 mMNaCl, 10 mM imidazole, pH 8.0) and eluted with elution buffer (50 mM NaH2PO4,300 mM NaCl, 250 mM imidazole, pH 8.0). Fractions containing the recombinantFLAG-retromer complex were collected and subsequently dialyzed into PBS, andprotein concentrations were determined using a micro BCA protein assay kit (Pierce).

GST pull-down assayGST or GST fusion proteins (0.1 mg/ml) were incubated with recombinant FLAG-tagged retromer cargo recognition complex (3�FLAG-Vps26, -Vps29 and -Vps35-His6) in 0.5 ml GST pull-down buffer [50 mM HEPES-KOH, pH 7.4, 50 mM KCl,1 mM MgCl2, 10% (v/v) glycerol, 2 mM dithiothreitol, and 0.6 mg/ml bovine serumalbumin] for 2 hours at 4°C. Glutathione-Sepharose 4B beads (50 l) were addedand the incubation was continued for an additional 30 minutes. The beads were washedwith 1 ml GST pull-down buffer five times and the bound proteins were eluted with50 l of G buffer (10 mM glutathione, 50 mM Tris-HCl, pH 9.0). Samples wereanalyzed by immunoblot analysis. Immunoblot analysis was performed using mouseanti-FLAG M2 mAb and horseradish peroxidase-conjugated anti-mouse IgG. Signalswere detected using Pierce SuperSignal West Pico chemiluminescent detection reagent(Pierce).

Cell transfection and immunofluorescence microscopyHuman HEp-2 epithelial cells were cultured on 12-well dishes at 37°C in DME high-glucose medium (Sigma) supplemented with 10% (v/v) FBS. For mutational analysisof the DMT1-II-recycling signal, plasmids encoding various GFP-tagged mutants ofDMT1-II were transfected using FuGENE 6 transfection reagent (Roche MolecularBiochemicals) into HEp-2 cells according to the manufacturer’s instructions. At 24hours after transfection, the cells were stripped with trypsin-EDTA, and seeded ontoglass coverslips, and cultured for an additional 48 hours. Cells were fixed,permeabilized, and immunostained with rabbit anti-GFP pAb and mouse anti-LAMP2mAb, followed by Alexa Fluor 488-conjugated goat anti-rabbit IgG and Alexa Fluor594-conjugated goat anti-Mouse IgG. Cell images were obtained using a confocalmicroscope (TSC SP-2; Leica) equipped with 488-nm Ar-Kr, 543/594 nm He-Ne,and 633 nm He-Ne lasers. Images were acquired in a sequential mode using a 63�Plan Apochromat NA/1.4 oil objective and the appropriate filter combination. Allimages were saved as TIFF files, their contrast was adjusted with Photoshop (versionCS2; Adobe), and the images were imported into Illustrator (version CS2; Adobe).

Pixel-by-pixel colocalization analysis was performed using ImageJ (plug-in JACoP)to determine the Pc with marker proteins (Bolte and Cordelieres, 2006).

To generate HEp-2 cells stably expressing 3�HA-DMT1-II, HEp-2 cells weregrown at 50-60% confluency in a 10 cm dish and transfected with 5 g pIRES neo-3�HA-DMT1-II plasmid using FuGENE 6 transfection reagent. At 24 hours afterthe transfection, cells were stripped with trypsin-EDTA and cultured in 96-well dishesin DMEM supplemented with 10% FBS containing 0.75 mg/ml G418 (NacalaiTesque). The medium was changed every 3 days until colonies were formed. Theexpression of the G418-resistant clones was tested by immunostaining and immunoblotanalysis, and clones with normal localization of DMT1-II and normal cell morphologywere selected.

For triple staining of 3�HA-DMT1-II, Vps35 and various organellar markers, HEp-2 cells stably expressing 3�HA-DMT1-II were grown on glass coverslips for 72hours and fixed, permeabilized, and immunostained with mouse anti-TfR, EEA1, CI-MPR, p230, or Rab11 mAb, rabbit anti-HA pAb, and goat anti-Vps35 pAb followedby donkey Alexa Fluor 555-conjugated anti-rabbit IgG, donkey Alexa Fluor 488-conjugated anti-mouse IgG, and donkey Alexa Fluor 633-conjugated anti-goat IgG.

RNA interferenceThe siRNA knockdown oligonucleotides were obtained from Invitrogen(StealthTM/siRNA duplex oligoribonucleotides) and resuspended in water accordingto the manufacturer’s instructions. HEp-2 cells stably expressing 3�HA-DMT1-IIwere grown to 30% confluency and transfected twice at 72-hours intervals with 33nM siRNA oligonucleotides using LipofectamineTM RNAiMAX (Invitrogen). Formost experiments, cells were analyzed 72 hours after the second round of transfection.

In rescue experiments, cDNA encoding GFP-tagged siRNA-resistant Vps35 wild-type or R107A mutant, nonfunctional mutant Vps35 (Gokool et al., 2007), or GFPalone were transfected using FuGENE 6 transfection reagent (Roche MolecularBiochemicals) into Vps35-depleted cells 24 hours after the second siRNA treatment.Twelve hours after the plasmid transfection, cells were stripped with trypsin-EDTAand seeded onto glass coverslips, and cultured for an additional 48 hours. Cells werefixed, permeabilized, and immunostained with rabbit anti-GFP pAb, rat anti-HA mAb,and mouse anti-LAMP2 or TfR mAb, followed by Alexa Fluor 488-conjugated anti-rabbit IgG, Alexa Fluor 555-conjugated anti-rat IgG, and Alexa Fluor 633-conjugatedanti-mouse IgG.

The following oligonucleotides were used to ablate expression of Vps35 (VSP35siRNA#1, 5�-G CCU UCA GAG GAU GUU GUA UCU UUA-3�; VPS35 siRNA#2,5�-GC AUG AGU UGU UAU GUG CUU AGU AA-3�).

We thank Scott D. Emr and Kazuhisa Nakayama for helpfuldiscussions and their critical reading of the manuscript. This work wassupported in part by the Asahi Glass Foundation, and also partlysupported by the Research Project Grants from Kawasaki MedicalSchool and the Okayama Medical Foundation.

Supplementary material available online athttp://jcs.biologists.org/cgi/content/full/123/5/756/DC1

ReferencesAndrews, N. C. (1999). Disorders of iron metabolism. N. Engl. J. Med. 341, 1986-1995.Andrews, N. C. (2008). Forging a field: the golden age of iron biology. Blood 112, 219-

230.Arighi, C. N., Hartnell, L. M., Aguilar, R. C., Haft, C. R. and Bonifacino, J. S. (2004).

Role of the mammalian retromer in sorting of the cation-independent mannose 6-phosphate receptor. J. Cell Biol. 165, 123-133.

Bolte, S. and Cordelieres, F. P. (2006). A guided tour into subcellular colocalization analysisin light microscopy. J. Microsc. 224, 213-232.

Bonifacino, J. S. and Traub, L. M. (2003). Signals for sorting of transmembrane proteinsto endosomes and lysosomes. Annu. Rev. Biochem. 72, 395-447.

Bonifacino, J. S. and Rojas, R. (2006). Retrograde transport from endosomes to the trans-Golgi network. Nat. Rev. Mol. Cell. Biol. 7, 568-579.

Bonifacino, J. S. and Hurley, J. H. (2008). Retromer. Curr. Opin. Cell Biol. 20, 427-436.Canonne-Hergaux, F., Gruenheid, S., Ponka, P. and Gros, P. (1999). Cellular and

subcellular localization of the Nramp2 iron transporter in the intestinal brush border andregulation by dietary iron. Blood 93, 4406-4417.

Canonne-Hergaux, F., Zhang, A. S., Ponka, P. and Gros, P. (2001). Characterization ofthe iron transporter DMT1 (NRAMP2/DCT1) in red blood cells of normal and anemicmk/mk mice. Blood 98, 3823-3830.

Carlton, J., Bujny, M., Peter, B. J., Oorschot, V. M., Rutherford, A., Mellor, H.,Klumperman, J., McMahon, H. T. and Cullen, P. J. (2004). Sorting nexin-1 mediatestubular endosome-to-TGN transport through coincidence sensing of high-curvaturemembranes and 3-phosphoinositides. Curr. Biol. 14, 1791-1800.

Collins, B. M. (2008). The structure and function of the retromer protein complex. Traffic9, 1811-1822.

Eaton, S. (2008). Retromer retrieves wntless. Dev. Cell 14, 4-6.Fleming, M. D., Trenor, C. C., 3rd, Su, M. A., Foernzler, D., Beier, D. R., Dietrich, W.

F. and Andrews, N. C. (1997). Microcytic anaemia mice have a mutation in Nramp2,a candidate iron transporter gene. Nat. Genet. 16, 383-386.

Jour

nal o

f Cel

l Sci

ence

Page 11: Retromer-mediated direct sorting is required for proper ... · Divalent metal transporter 1 (DMT1 or SLC11A2, formerly called NRAMP2/DCT1) is an integral membrane protein consisting

766 Journal of Cell Science 123 (5)

Fleming, M. D., Romano, M. A., Su, M. A., Garrick, L. M., Garrick, M. D. andAndrews, N. C. (1998). Nramp2 is mutated in the anemic Belgrade (b) rat: evidenceof a role for Nramp2 in endosomal iron transport. Proc. Natl. Acad. Sci. USA 95, 1148-1153.

Ghosh, P., Dahms, N. M. and Kornfeld, S. (2003). Mannose 6-phosphate receptors: newtwists in the tale. Nat. Rev. Mol. Cell. Biol. 4, 202-212.

Gokool, S., Tattersall, D., Reddy, J. V. and Seaman, M. N. (2007). Identification of aconserved motif required for Vps35p/Vps26p interaction and assembly of the retromercomplex. Biochem. J. 408, 287-295.

Gruenheid, S., Canonne-Hergaux, F., Gauthier, S., Hackam, D. J., Grinstein, S. andGros, P. (1999). The iron transport protein NRAMP2 is an integral membraneglycoprotein that colocalizes with transferrin in recycling endosomes. J. Exp. Med. 189,831-841.

Gunshin, H., Mackenzie, B., Berger, U. V., Gunshin, Y., Romero, M. F., Boron, W. F.,Nussberger, S., Gollan, J. L. and Hediger, M. A. (1997). Cloning and characterizationof a mammalian proton-coupled metal-ion transporter. Nature 388, 482-488.

Hubert, N. and Hentze, M. W. (2002). Previously uncharacterized isoforms of divalentmetal transporter (DMT)-1: implications for regulation and cellular function. Proc. Natl.Acad. Sci. USA 99, 12345-12350.

Imai, Y., Matsushima, Y., Sugimura, T. and Terada, M. (1991). A simple and rapidmethod for generating a deletion by PCR. Nucleic Acids Res. 19, 2785.

Katzmann, D. J., Odorizzi, G. and Emr, S. D. (2002). Receptor downregulation andmultivesicular-body sorting. Nat. Rev. Mol. Cell. Biol. 3, 893-905.

Lam-Yuk-Tseung, S. and Gros, P. (2006). Distinct targeting and recycling properties oftwo isoforms of the iron transporter DMT1 (NRAMP2, Slc11A2). Biochemistry 45, 2294-2301.

Lam-Yuk-Tseung, S., Touret, N., Grinstein, S. and Gros, P. (2005). Carboxyl-terminusdeterminants of the iron transporter DMT1/SLC11A2 isoform II (-IRE/1B) mediateinternalization from the plasma membrane into recycling endosomes. Biochemistry 44,12149-12159.

Lee, P. L., Gelbart, T., West, C., Halloran, C. and Beutler, E. (1998). The human Nramp2gene: characterization of the gene structure, alternative splicing, promoter region andpolymorphisms. Blood Cells Mol. Dis. 24, 199-215.

Luton, F., Hexham, M. J., Zhang, M. and Mostov, K. E. (2009). Identification of acytoplasmic signal for apical transcytosis. Traffic 10, 1128-1142.

McMahon, H. T. and Gallop, J. L. (2005). Membrane curvature and mechanisms ofdynamic cell membrane remodelling. Nature 438, 590-596.

Nothwehr, S. F., Ha, S. A. and Bruinsma, P. (2000). Sorting of yeast membrane proteinsinto an endosome-to-Golgi pathway involves direct interaction of their cytosolicdomains with Vps35p. J. Cell Biol. 151, 297-310.

Puertollano, R., Aguilar, R. C., Gorshkova, I., Crouch, R. J. and Bonifacino, J. S.(2001). Sorting of mannose 6-phosphate receptors mediated by the GGAs. Science 292,1712-1716.

Rink, J., Ghigo, E., Kalaidzidis, Y. and Zerial, M. (2005). Rab conversion as a mechanismof progression from early to late endosomes. Cell 122, 735-749.

Rojas, R., van Vlijmen, T., Mardones, G. A., Prabhu, Y., Rojas, A. L., Mohammed,S., Heck, A. J., Raposo, G., van der Sluijs, P. and Bonifacino, J. S. (2008). Regulationof retromer recruitment to endosomes by sequential action of Rab5 and Rab7. J. CellBiol. 183, 513-526.

Seaman, M. N. (2004). Cargo-selective endosomal sorting for retrieval to the Golgi requiresretromer. J. Cell Biol. 165, 111-122.

Seaman, M. N. (2005). Recycle your receptors with retromer. Trends Cell Biol. 15, 68-75.

Seaman, M. N. (2007). Identification of a novel conserved sorting motif required forretromer-mediated endosome-to-TGN retrieval. J. Cell Sci. 120, 2378-2389.

Seaman, M. N. (2008). Endosome protein sorting: motifs and machinery. Cell Mol. LifeSci. 65, 2842-2858.

Seaman, M. N., McCaffery, J. M. and Emr, S. D. (1998). A membrane coat complexessential for endosome-to-Golgi retrograde transport in yeast. J. Cell Biol. 142, 665-681.

Seaman, M. N., Harbour, M. E., Tattersall, D., Read, E. and Bright, N. (2009).Membrane recruitment of the cargo-selective retromer subcomplex is catalysed by thesmall GTPase Rab7 and inhibited by the Rab-GAP TBC1D5. J. Cell Sci. 122, 2371-2382.

Shah, Y. M., Matsubara, T., Ito, S., Yim, S. H. and Gonzalez, F. J. (2009). Intestinalhypoxia-inducible transcription factors are essential for iron absorption following irondeficiency. Cell Metab. 9, 152-164.

Strochlic, T. I., Setty, T. G., Sitaram, A. and Burd, C. G. (2007). Grd19/Snx3p functionsas a cargo-specific adapter for retromer-dependent endocytic recycling. J. Cell Biol. 177,115-125.

Su, M. A., Trenor, C. C., Fleming, J. C., Fleming, M. D. and Andrews, N. C. (1998).The G185R mutation disrupts function of the iron transporter Nramp2. Blood 92, 2157-2163.

Tabuchi, M., Yoshimori, T., Yamaguchi, K., Yoshida, T. and Kishi, F. (2000). HumanNRAMP2/DMT1, which mediates iron transport across endosomal membranes, is localizedto late endosomes and lysosomes in HEp-2 cells. J. Biol. Chem. 275, 22220-22228.

Tabuchi, M., Tanaka, N., Nishida-Kitayama, J., Ohno, H. and Kishi, F. (2002).Alternative splicing regulates the subcellular localization of divalent metal transporter1 isoforms. Mol. Biol. Cell 13, 4371-4387.

Takatsu, H., Katoh, Y., Shiba, Y. and Nakayama, K. (2001). Golgi-localizing, gamma-adaptin ear homology domain, ADP-ribosylation factor-binding (GGA) proteins interactwith acidic dileucine sequences within the cytoplasmic domains of sorting receptorsthrough their Vps27p/Hrs/STAM (VHS) domains. J. Biol. Chem. 276, 28541-28545.

Touret, N., Furuya, W., Forbes, J., Gros, P. and Grinstein, S. (2003). Dynamic trafficthrough the recycling compartment couples the metal transporter Nramp2 (DMT1) withthe transferrin receptor. J. Biol. Chem. 278, 25548-25557.

Traer, C. J., Rutherford, A. C., Palmer, K. J., Wassmer, T., Oakley, J., Attar, N., Carlton,J. G., Kremerskothen, J., Stephens, D. J. and Cullen, P. J. (2007). SNX4 coordinatesendosomal sorting of TfnR with dynein-mediated transport into the endocytic recyclingcompartment. Nat. Cell Biol. 9, 1370-1380.

Verges, M., Luton, F., Gruber, C., Tiemann, F., Reinders, L. G., Huang, L., Burlingame,A. L., Haft, C. R. and Mostov, K. E. (2004). The mammalian retromer regulatestranscytosis of the polymeric immunoglobulin receptor. Nat. Cell Biol. 6, 763-769.

Wassmer, T., Attar, N., Harterink, M., van Weering, J. R., Traer, C. J., Oakley, J.,Goud, B., Stephens, D. J., Verkade, P., Korswagen, H. C. et al. (2009). The retromercoat complex coordinates endosomal sorting and dynein-mediated transport, with carrierrecognition by the trans-Golgi network. Dev. Cell 17, 110-122.

Yang, P. T., Lorenowicz, M. J., Silhankova, M., Coudreuse, D. Y., Betist, M. C. andKorswagen, H. C. (2008). Wnt signaling requires retromer-dependent recycling of MIG-14/Wntless in Wnt-producing cells. Dev. Cell 14, 140-147.

Yoshimori, T., Shimonishi, Y. and Uchida, T. (1988). Binding properties of monoclonalantibody to the cytoplasmic domain of transferrin receptor. Cell Struct. Funct. 13, 311-324.

Zhu, Y., Doray, B., Poussu, A., Lehto, V. P. and Kornfeld, S. (2001). Binding of GGA2to the lysosomal enzyme sorting motif of the mannose 6-phosphate receptor. Science292, 1716-1718.

Jour

nal o

f Cel

l Sci

ence


Recommended