13
Rin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and Rab22 Barbara Woller a , Susan Luiskandl a , Milica Popovic b , Barbara E.M. Prieler a , Gloria Ikonge a , Michaela Mutzl a , Holger Rehmann b , Ruth Herbst a, a Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austria b Department of Physiological Chemistry, UMC Utrecht, 3584 CG Utrecht, The Netherlands abstract article info Article history: Received 5 November 2010 Received in revised form 8 March 2011 Accepted 10 March 2011 Available online 17 March 2011 Keywords: Endocytosis Rab5 GEF MuSK Receptor tyrosine kinase RIN proteins serve as guanine nucleotide exchange factors for Rab5a. They are characterized by the presence of a RIN homology domain and a C-terminal Vps9 domain. Currently three family members have been described and analyzed. Here we report the identication of a novel RIN family member, Rin-like (Rinl), that represents a new interaction partner of the receptor tyrosine kinase MuSK, which is an essential key regulator of neuromuscular synapse development. Rinl is localized to neuromuscular synapses but shows the highest expression in thymus and spleen. Rinl preferentially binds to nucleotide-free Rab5a and catalyzes the exchange of GDP for GTP. Moreover, Rinl also binds GDP-bound Rab22 and increases the GDP/GTP exchange implicating Rinl in endocytotic processes regulated by Rab5a and Rab22. Interestingly, Rinl shows a higher catalytic rate for Rab22 compared to Rab5a. Rinl is closely associated with the cytoskeleton and thus contributes to the spatial control of Rab5a and Rab22 signaling at actin-positive compartments. Most importantly, overexpression of Rinl affects uid-phase as well as EGFR endocytosis. © 2011 Elsevier B.V. 1. Introduction Cycling of Rab GTPases between the inactive (GDP bound) and active (GTP bound) state depends on nucleotide exchange of GDP to GTP catalyzed by guanine nucleotide exchange factors (GEFs) and the stimulation of the intrinsically slow GTP-hydrolysis activity, which converts Rab bound GTP to GDP. The GDP to GTP exchange activates Rab proteins by inducing a conformation that enables them to interact with effector proteins. Ras-interaction/interference (RIN) proteins, also known as Ras and Rab interactors, belong to the family of Vps9 domain-containing GEFs [1]. The founding member of the RIN protein family, RIN1, was identied as an inhibitor of signaling mediated by the GTPase H-Ras. Via its Ras association (RA) domain RIN1 competes with binding of the Ras effector Raf1 [2]. Further studies revealed that RIN1 contains a functional Vps9 domain indicative for Rab GEFs. This domain was shown to be necessary for binding to Rab5a and for GDP/ GTP exchange activity, and indeed RIN1 was characterized as the Ras effector protein that connects Ras signaling to the control of endocytotic processes [3]. The Rab5a GEF activity is shared by all three known RIN proteins as well as the conserved domain structure consisting of an RA domain, a Vps9 domain, a RIN homology (RH) domain and an N-terminal SH2 domain [1]. The Vps9 domain was originally identied in the yeast protein Vps9p, which serves as GEF for Vps21p and its mammalian homologue Rab5a [4]. Nucleotide exchange by GEFs on Rab5a results in increased endocytosis [5]. The consensus Vps9p catalytic domain is present in a variety of multi-domain proteins including the RIN protein family [1]. The crystal structure of the Rabex-5 Vps9 domain showed that the core Vps9 domain is stabilized by an essential helical bundle N-terminal to the Vps9 region, in the case of the RIN proteins corresponding to the RH domain [6]. In addition, a high specicity for the Rab5 subfamily, which consists of Rab5a, Rab5b, Rab5c, Rab21, Rab22 and Rab31 (also termed Rab22B), was demonstrated. The proteins of the Rab5 subfamily are important regulators of early endocytosis. Rab5a is among the best-studied Rab proteins and is known as the key regulator of endocytotic processes like clathrin-coated vesicle formation, fusion between early endosomes, endosomal cargo recruit- ment and endosomal motility [7]. Rab21 and Rab22 are less well characterized. Both have been localized to early endosomes and implicated in early endocytosis [8,9]. However, Rab21 is also thought to be involved in transport from the trans Golgi network to endosomes whereas the predominant role of Rab22 has been attributed to recycling processes [10,11]. Biochimica et Biophysica Acta 1813 (2011) 11981210 Abbreviations: AChR, acetylcholine receptor; BGT, bungarotoxin; EGFR, epidermal growth factor receptor; GEF, guanine nucleotide exchange factor; GFP, green uorescent protein; GST, glutathione S-transferase; HRP, horseradish peroxidase; MuSK, muscle specic kinase; RT-PCR, reverse transcriptase polymerase chain reaction; RTK, receptor tyrosine kinase; SH2, Src homology 2 Corresponding author at: Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria. Tel.: +43 1 4016034350; fax: +43 1 40160934093. E-mail address: [email protected] (R. Herbst). 0167-4889 © 2011 Elsevier B.V. doi:10.1016/j.bbamcr.2011.03.005 Contents lists available at ScienceDirect Biochimica et Biophysica Acta journal homepage: www.elsevier.com/locate/bbamcr Open access under CC BY-NC-ND license. Open access under CC BY-NC-ND license.

Biochimica et Biophysica Acta - CORERin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and Rab22 Barbara Wollera, Susan Luiskandla, Milica

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Biochimica et Biophysica Acta - CORERin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and Rab22 Barbara Wollera, Susan Luiskandla, Milica

Biochimica et Biophysica Acta 1813 (2011) 1198–1210

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta

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

Rin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factorfor Rab5a and Rab22

Barbara Woller a, Susan Luiskandl a, Milica Popovic b, Barbara E.M. Prieler a, Gloria Ikonge a, Michaela Mutzl a,Holger Rehmann b, Ruth Herbst a,⁎a Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austriab Department of Physiological Chemistry, UMC Utrecht, 3584 CG Utrecht, The Netherlands

Abbreviations: AChR, acetylcholine receptor; BGT, bgrowth factor receptor; GEF, guanine nucleotide efluorescent protein; GST, glutathione S-transferase;MuSK, muscle specific kinase; RT-PCR, reverse transcriptRTK, receptor tyrosine kinase; SH2, Src homology 2⁎ Corresponding author at: Center for Brain Research

Spitalgasse 4, 1090 Vienna, Austria. Tel.: +43 1 4016034E-mail address: [email protected] (R. H

0167-4889 © 2011 Elsevier B.V.doi:10.1016/j.bbamcr.2011.03.005

Open access under CC BY

a b s t r a c t

a r t i c l e i n f o

Article history:Received 5 November 2010Received in revised form 8 March 2011Accepted 10 March 2011Available online 17 March 2011

Keywords:EndocytosisRab5GEFMuSKReceptor tyrosine kinase

RIN proteins serve as guanine nucleotide exchange factors for Rab5a. They are characterized by the presenceof a RIN homology domain and a C-terminal Vps9 domain. Currently three family members have beendescribed and analyzed. Here we report the identification of a novel RIN family member, Rin-like (Rinl), thatrepresents a new interaction partner of the receptor tyrosine kinase MuSK, which is an essential key regulatorof neuromuscular synapse development. Rinl is localized to neuromuscular synapses but shows the highestexpression in thymus and spleen. Rinl preferentially binds to nucleotide-free Rab5a and catalyzes theexchange of GDP for GTP. Moreover, Rinl also binds GDP-bound Rab22 and increases the GDP/GTP exchangeimplicating Rinl in endocytotic processes regulated by Rab5a and Rab22. Interestingly, Rinl shows a highercatalytic rate for Rab22 compared to Rab5a. Rinl is closely associated with the cytoskeleton and thuscontributes to the spatial control of Rab5a and Rab22 signaling at actin-positive compartments. Mostimportantly, overexpression of Rinl affects fluid-phase as well as EGFR endocytosis.

ungarotoxin; EGFR, epidermalxchange factor; GFP, greenHRP, horseradish peroxidase;ase polymerase chain reaction;

, Medical University of Vienna,350; fax: +43 1 40160934093.erbst).

-NC-ND license.

© 2011 Elsevier B.V. Open access under CC BY-NC-ND license.

1. Introduction

Cycling of Rab GTPases between the inactive (GDP bound) andactive (GTP bound) state depends on nucleotide exchange of GDP toGTP catalyzed by guanine nucleotide exchange factors (GEFs) and thestimulation of the intrinsically slow GTP-hydrolysis activity, whichconverts Rab bound GTP to GDP. The GDP to GTP exchange activatesRab proteins by inducing a conformation that enables them to interactwith effector proteins. Ras-interaction/interference (RIN) proteins,also known as Ras and Rab interactors, belong to the family of Vps9domain-containing GEFs [1]. The founding member of the RIN proteinfamily, RIN1, was identified as an inhibitor of signaling mediated bythe GTPase H-Ras. Via its Ras association (RA) domain RIN1 competeswith binding of the Ras effector Raf1 [2]. Further studies revealed thatRIN1 contains a functional Vps9 domain indicative for Rab GEFs. Thisdomain was shown to be necessary for binding to Rab5a and for GDP/GTP exchange activity, and indeed RIN1 was characterized as the Ras

effector protein that connects Ras signaling to the control ofendocytotic processes [3]. The Rab5a GEF activity is shared by allthree known RIN proteins as well as the conserved domain structureconsisting of an RA domain, a Vps9 domain, a RIN homology (RH)domain and an N-terminal SH2 domain [1]. The Vps9 domain wasoriginally identified in the yeast protein Vps9p, which serves as GEFfor Vps21p and its mammalian homologue Rab5a [4]. Nucleotideexchange by GEFs on Rab5a results in increased endocytosis [5].

The consensus Vps9p catalytic domain is present in a variety ofmulti-domain proteins including the RIN protein family [1]. Thecrystal structure of the Rabex-5 Vps9 domain showed that the coreVps9 domain is stabilized by an essential helical bundle N-terminal tothe Vps9 region, in the case of the RIN proteins corresponding to theRH domain [6]. In addition, a high specificity for the Rab5 subfamily,which consists of Rab5a, Rab5b, Rab5c, Rab21, Rab22 and Rab31 (alsotermed Rab22B), was demonstrated.

The proteins of the Rab5 subfamily are important regulators of earlyendocytosis. Rab5a is among thebest-studiedRabproteins and is knownas the key regulator of endocytotic processes like clathrin-coated vesicleformation, fusion between early endosomes, endosomal cargo recruit-ment and endosomal motility [7]. Rab21 and Rab22 are less wellcharacterized. Both have been localized to early endosomes andimplicated in early endocytosis [8,9]. However, Rab21 is also thoughtto be involved in transport from the trans Golgi network to endosomeswhereas the predominant role of Rab22 has been attributed to recyclingprocesses [10,11].

Page 2: Biochimica et Biophysica Acta - CORERin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and Rab22 Barbara Wollera, Susan Luiskandla, Milica

1199B. Woller et al. / Biochimica et Biophysica Acta 1813 (2011) 1198–1210

RIN proteins have been proposed as regulators of early Rab5a-dependent endocytosis due to their binding properties for Rab5a andtheir Rab5a-specific GEF activity. RIN1 stimulates Rab5a-dependentendosome fusion and ligand-mediated endocytosis of the EGF-receptor(EGFR), the insulin receptor and the EphA4 receptor [12–14]. Inaddition, RIN3 has been shown to interact with amphiphysin II, whichis involved in clathrin- and dynamin-dependent endocytosis [15]. Theseobservations are of special interest since increasing evidence supports aclose interplay between signaling and endocytosis of receptor tyrosinekinases (RTK). In particular, it was shown that receptors remain activewithin endosomes [16]. This led to the hypothesis that membranetrafficking could control signal transduction. Studies on different RTKshave now demonstrated that there are twomain types of functions thatreceptor trafficking has in regulating receptor signaling: it controls themagnitude of the response or it controls the specificity of the response[17].

Here we report the identification of a novel RIN family member,Rin-like (Rinl), which was isolated as interaction partner of themuscle-specific RTK MuSK. Signal transduction events induced byMuSK are crucially linked to the formation of the neuromuscularsynapse (NMS) [18]. MuSK is activated by the motor neuron-derivedheparansulfate proteoglycan agrin [19]. Agrin does not bind MuSKdirectly but interacts with Lrp4, a member of the LDL receptor family[20,21]. Upon binding, the Lrp4/MuSK complex presumably under-goes a structural rearrangement that results in a dimerization andsubsequent autophosphorylation of MuSK. The subsequent activationof the MuSK kinase induces a signaling cascade leading to theformation of the NMS including postsynaptic differentiation charac-terized by the accumulation of acetylcholine receptors (AChRs) atsynaptic sites and presynaptic differentiation as depicted by thedevelopment of active zones. Consistent with this model, agrin, MuSKand lrp4mutantmice fail to formNMSs, lacking all features of pre- andpostsynaptic specializations [22–24].

Rinl interacts with MuSK independent of its phosphorylation andlocalizes to NMSs. Further, we show that Rinl acts as GEF for Rab5a.Moreover, Rinl interactswith inactive Rab22 and catalyzes theGDP/GTPexchangeonRab22. Co-localizationof RinlwithRab5a orRab22 in actin-rich membrane ruffles implicates a novel mechanism for the recruit-ment of Rab5a and Rab22 to the cytoskeleton via binding to Rinl. Inaddition, increased fluid-phase uptake and EGFR endocytosis upon Rinloverexpression implicates Rinl as regulator of early endocytoticprocesses.

2. Materials and methods

2.1. Antibodies and reagents

The following antibodies were purchased from commercialsources: anti-phosphotyrosine PY99 (Santa Cruz) and PY-100 (CellSignaling), anti-myc 9E10 (Sigma-Aldrich), anti-GFP (Santa Cruz).Antibodies against the C-terminal sequence of MuSK were describedpreviously [25]. Alexa 594-conjugated α-bungarotoxin (BGT), Alexa555-conjugated EGF and Alexa 488-conjugated secondary antibodieswere obtained from Invitrogen. Horseradish-peroxidase-coupled andfluorophore-conjugated secondary antibodies were purchased fromJackson ImmunoResearch. TRITC-conjugated phalloidin and cytocha-lasin D were purchased from Sigma-Aldrich.

2.2. Generation of antibodies against Rinl

Rinl (aa 243–307, Mus musculus) was amplified by PCR (5′-TATGAATTCCACTTTTCCTGTCCT-3′ and 5′-TATAAGCTTTGGGCTG-TAGGGATTT-3′) and cloned into modified pMALc and pGEX-4T-3vectors, which encode for seven C-terminal histidines in addition toMBP or GST respectively [26]. Recombinant proteins were expressedin the Escherichia coli strain XL1 blue in LB medium supplemented

with 0.4% glucose by inductionwith 0.2 mM IPTG and purified via HIS-affinity chromatography. The MBP fusion protein was used to raisepolyclonal antisera in rabbits as described previously [26]. Reactiveserum was affinity purified on an Affi-Gel 10 (Bio-Rad) columncoupled with the GST fusion protein mentioned above. The columnwas sequentially washed with phosphate buffered saline (PBS), 0.1 MNaHCO3 and 0.1 M NaHCO3/0.5 M NaCl. The bound antibodies wereeluted with 7 ml acidic elution buffer (0.1 M glycine, pH 2.5) andfractions of 1 ml were collected. Purified antibodies were tested forimmunoblotting and immunoprecipitation as shown in Fig. S4.

2.3. Plasmids

The MuSK cytoplasmic domain carrying the mutations Y553F orY750, 754, 755F (KD)was subcloned into theyeast bait vector pBTM116.It was previously demonstrated that MuSK Y750, Y754, 755F, whichaffects the activation loop of the kinase domain, behaves as kinase-deficient mutant similar to a K608A mutant, which blocks ATP bindingwithin the autocatalytic loop [27,28]. The MMT bait construct containsthe TrkA cytoplasmic domain including the MuSK juxtamembraneregion in pBTM116 [27]. To generate BTM/tpr-met-JM, the juxtamem-brane region of MuSK (Rattus norvegicus, aa 528–570) was subclonedinto BTM/tpr-met using the primers 5′-AATGAATTCAGAGAGTCGG-CAGC-3′ and 5′-TAGTCGACTACGGATACTCCAGGCTGAG-3′. Kinase-dead(K608A) and kinase-active MuSK (LS745,746MT) carrying a myc-tagwere provided by Dr. Burden (NYU School ofMedicine, NewYork, [20]).

The Rinl 5′-end was amplified from mouse muscle cDNA using thefollowing primers: 5′-CTGAGCAGCCTTGAGTCTTGTCTT-3′ (located up-stream of the predicted start codon) and 5′-CCTGGCCAGAGCACG-GATGT-3′. Full length Rinl was then cloned into pcDNA3 (Invitrogen)with or without an N-terminal myc-tag and into pEGFP-C3 (Clontech).Truncations ΔSH2 (aa 200–563) and ΔVps9 (aa 1–394) with an N-terminal myc-tag were subcloned into pcDNA3. For yeast two-hybridand GST-pulldown experiments Rinl and truncation mutants werecloned into pACT2 (Clontech) and pGEX (GE Healthcare) vectorsrespectively: GST-Rinl (aa 1–563 in pGEX-5X-2), GST-Y2H (aa 200–563in pGEX-3), GST-RH/ΔVps9 (aa 200–479 in pGEX-2), GST-RH/Vps9 (aa248–563 in pGEX-2).

pLexA-Rab5a (wt, S34N, Q79L) plasmids were a gift from Dr. Zerial(Max Planck Institute of Molecular Cell Biology and Genetics,Dresden), for GFP-tagged constructs Rab5a wt and mutants werecloned into pEGFP-C2 (Clontech) using EcoRI. pEGFP-Rab22 (wt,S19N, Q64L) was provided by Dr. Donaldson (NIH, Bethesda). Rab22inserts were amplified by PCR (Rab22 MunI fw 5′-AAACAATT-GATGGCGCTGAGGGAGCTCAAA-3′ and Rab22 SalI rev 5′-AAAGTC-GACGCAGCAGCTTCGCTGCGGCTC-3′) and cloned into pLexAlinearized with EcoRI and SalI. GFP-Rab7a (wt, T22N, Q67L) constructswere a gift from Dr. Bucci (University of Salento, Lecce).

All constructs used in this study were checked by sequencing.

2.4. Cell culture

HEK 293T cells, COS7 and HeLa cells were maintained in DMEMsupplemented with glutamine, 4.5 mg/ml glucose, 10% fetal bovineserum (FBS) and 100 μg/ml penicillin/streptomycin. HEK 293T cellswere transfected using the protocol described by Chen and Okayama[29]. COS7 and HeLa cells were transfected using TurboFect(Fermentas). Briefly, COS7 and HeLa cells were plated on glasscoverslips and transfected the next day with 1 μg DNA and 1.5 μlTurboFect in 2 ml DMEM.

2.5. Yeast two-hybrid screen

The MuSK cytoplasmic domain (Rattus norvegicus, aa 517–868) wassubcloned into themodified pBTM116 vector, BTM/tpr-met (a gift fromDr. Birchmeier, Max-Delbrueck Center for Molecular Medicine, Berlin),

Page 3: Biochimica et Biophysica Acta - CORERin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and Rab22 Barbara Wollera, Susan Luiskandla, Milica

1200 B. Woller et al. / Biochimica et Biophysica Acta 1813 (2011) 1198–1210

which contains an active c-Met kinase lacking the multiple docking site[30]. Insertion ofMuSKproduces a fusion between c-Met andMuSK. TheMuSK bait was screened against a mouse muscle cDNA library (a giftfrom Dr. Chamberlain, University of Michigan Medical School, AnnArbor) cloned into the pACT2 prey vector [31]. The BTM/tpr-met-MuSKbait plasmid was introduced into the L40 yeast strain. Large scaletransformations with the prey library were performed according toVojtek and Hollenberg [32]. A total of 7.2×106 transformants werescreened for growth on –HIS. Forty-six positive clones were recovered.After retransformation of the recovered cDNA plasmids six clones grewon –HIS and produced a positive signal in a β-gal assay with MuSK butwere negative for control plasmids (BTM/tpr-met, BTM/cMet and BTM/lamin). To analyze the interaction between MuSK and Rinl or Rinl andRab5a/Rab22 respectively, the bait and prey plasmidswere transformedinto the yeast strain L40. Single colonieswere restreakedon–LEU/TRPor–LEU/TRP/HIS. A β-gal assay screening for lacZ expression wasperformed as described previously [32].

2.6. GST-pulldown experiments

All GST fusion proteins were expressed in the bacterial strainRosetta by induction with 0.5 mM IPTG. The harvested bacteria wereresuspended in ice-cold buffer R (PBS, 1% Triton X-100, 1 mg/mllysozyme, 0.2 mM phenylmethylsulfonyl fluoride (PMSF)). Cells werelysed on ice for 1 hour, 0.25% sarkosyl was added and samples weresonicated. Cleared lysates were incubated with glutathione agarosebeads (Sigma) for 1 hour under constant rotation at 4 °C, followed bythree washing steps with buffer R (without lysozyme). Proteinconcentration and quality were assayed by SDS-PAGE and subsequentCoomassie staining. Recombinant proteins were stored at −80 °Cuntil use. Cell lysates were incubated with immobilized GST fusionproteins for 4–18 hours at 4 °C. Beads were washed three times withNP-40 lysis buffer (1% Nonidet P-40, 5 mM EGTA, 50 mMNaCl, 30 mMtriethanolamine [pH 7.5], 50 mM NaF). Proteins bound to the beadswere subjected to SDS-PAGE and immunoblotting.

2.7. Lysate preparation, immunoprecipitation and immunoblotting

Lysates were prepared from cultured cells or adult mouse tissuesusing NP-40 lysis buffer supplemented with fresh proteinase inhibitors(1 μg/ml leupeptin, 1 μg/ml pepstatin, 1 μg/ml aprotinin, 0.2 mM PMSF,1 mMsodiumorthovanadate). Protein concentrationswere determinedusing Roti-Nanoquant (Roth) protein assay reagent.

For co-immunoprecipitation experiments, HEK 293T cells wereco-transfected with indicated constructs. Cleared lysates wereprecipitated with polyclonal affinity-purified antibodies directed toeither MuSK or Rinl overnight. The next day protein A agarose(Roche)was added for 1–3 hours, the beadswerewashed three timeswith NP-40 lysis buffer with increased (150 mM) NaCl concentrationand precipitated protein complexes were analyzed by SDS-PAGE andimmunoblotting.

2.8. RT-PCR and quantitative PCR

RNA was isolated from C57BL/6 mice using TRI reagent (Sigma)according to the manufacturer's protocol and reverse transcribed tocDNA. The following primers were used for subsequent PCR reactions:Rinl-1154fw (5′-GAAGATCTTGGCCCCGCTGT-3′), Rinl-1645rev (5′-CTGGTAGTGAGCAATGTGGT-3 ′) , act in1 (5 ′-TTCTACAAT-GAGCTGCGTGTGG-3′), actin2 (5′-CTCGGTCAGGATCTTCATGAGG-3′),Gapdh/fw (5′-TGCATCCTGCACCACCAACT-3′), Gapdh/rev (5′-ATGCCTGCTTCACCACCTTC-3′). Quantitative real-time PCR (qPCR)was performed with IQTM SYBR Green Supermix (Bio-Rad) using aBio-Rad iCycler. Rinl levels were normalized to Gapdh using theformula: Rinl/Gapdh=2(CTx−CTrel) /2(CTx−CTrel). Spleenwas used

as reference tissue (rel) with a value set to 1. PCR reactions wereperformed in duplicates or triplicates.

2.9. Immunofluorescence

Eighteen hours after transfection, COS7 cells were fixed with 4%paraformaldehyde (PFA)/PBS for 10 minutes at room temperature (RT),permeabilized with 0.1% Triton/PBS for 5 minutes at RT and incubatedwith blocking solution (PBS containing 10% FBS) followed by incubationwith appropriate primary antibodies for 1 hour at RT. Cellswerewashedwith PBS and incubated with fluorophore-conjugated secondaryantibodies with or without TRITC-labeled phalloidin. Cells weremounted in Mowiol and visualized using a Leica TCS SP5 spectralconfocalmicroscopewith a HCX PL APO CS 63×/1.4 oil objective. For thedisruption of the actin cytoskeleton, transfected cells were treated with1 μM cytochalasin D or the solvent DMSO in DMEM for 30 minutesbefore the cells were fixed and stainedwith antibodies. The distributionof Rab5a or Rab22 in Rinl positive cells was categorized into fourdifferent classes and plotted as the average±s.e.m. of three indepen-dent experiments (sum of all subcategories is 100%).

Mouse muscle cryosections were stained as previously described[33]. Stained muscle sections were viewed on a confocal microscopeas described above.

2.10. EGF uptake

Twenty to 24 hours post transfection cells were starved for 3 hoursin DMEM, labeled with Alexa 555-conjugated EGF for 30 minutes at4 °C and washed four times with ice-cold DMEM supplemented with1 mg/ml BSA. Then the cells were put back at 37 °C for endocytosis tooccur. At indicated time points cells were fixedwith 1% PFA in PBS andembedded in Mowiol. Cells were analyzed by confocal microscopy asdescribed above. Representative images are shown.

2.11. HRP uptake

HEK 293T cells were transfected with the following constructs:pEGFP, pEGFP-Rab5a wt, pEGFP-Rinl as stated. About 20 hours aftertransfection cells were sorted for GFP expression by FACS. GFP-positivecellswere seeded on polyornithine coated 12-well plates overnight. Thenext day HRP uptake was analyzed as previously described [34]. Briefly,cells were incubated in internalization medium (IM; DMEM, 1% FBS,24 mM HEPES [pH 7.5]) containing 4 mg/ml HRP for 1 hour at 37 °C.After internalization, cellswerewashed oncewithwarm IM followed bythree washes with ice-cold PBS supplemented with 1 mM CaCl2, 1 mMMgCl2, 2 mg/ml BSA. Finally, cells were washed once with cold PBS andextracted for 15 minutes on ice with lysis buffer (1% w/v Triton X-100,20 mMHEPES [pH 7.5], 0.2 mM PMSF). Total HRP activity of the lysateswas determined in duplicates of triplicate samples using the substrate3,3′,5,5′-tetramethylbenzidine (Sigma). HRP activity was normalized tothe protein concentration of individual lysates.

2.12. GEF activity assay

Rab5a (Cane lupus, aa 15–185), Rab7a (Cane lupus, aa 1–185),Rab22 (Cane lupus, aa 1–173) and Rinl (Mus musculus, aa 260–563)were cloned into pGEX-4T-3 and expressed in the bacterial strainCK600K in Standard I medium (Merck) by induction with 100 μMIPTG at an OD600 of 0.8 at 25 °C overnight. Bacteria were harvestedby centrifugation and washed with 0.9% NaCl. Rab proteins werepurified, cleaved from GST-tag and loaded with mGDP (2′-/3′-O-(N′-Methylanthraniloyl)guanosine-5′-O-diphosphate) essentially as de-scribed for Rap [35]. Rinl was purified the same way except that thebufferwasMgCl2-free and contained 5 mMEDTA, and the GST-tagwasnot cleaved. Fluorescencemeasurementswere performed as described[35]. In brief, 200 nM of the G-protein loaded with mGDP were

Page 4: Biochimica et Biophysica Acta - CORERin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and Rab22 Barbara Wollera, Susan Luiskandla, Milica

1201B. Woller et al. / Biochimica et Biophysica Acta 1813 (2011) 1198–1210

incubated in buffer G (50 mM Tris–HCl, pH 7.5, 50 mM NaCl, 5 mMMgCl2, 5 mM DTT, 5% glycerol) in the presence of 20 μM GDP andincreasing amounts of Rinl. For kinetic analysis the obtained curveswere fitted as single exponential decay to obtain the rate constant kobs.kobs were plotted against the concentration of Rinl and analyzed bylinear fitting, whereby the slope was defined as the catalytic efficiencyof the GEF reaction.

3. Results

3.1. Rinl: a novel interaction partner of MuSK

To identify proteins that bind to MuSK, a mouse muscle cDNAlibrary was screened with the MuSK cytoplasmic domain using theyeast two-hybrid system. Screening of 7.2×106 transformantsyielded six positive clones that interacted strongly with MuSK. Oneof these clones, termed CL-6, was identified independently threetimes. It consisted of a 2204 bp cDNA encoding 363 amino acids, anin-frame stop codon, the 3′ UTR and the poly(A) tail. The isolatedcDNA was identical to ENSMUST00000059857 lacking however thefirst 199 amino acids. We therefore isolated the missing 5′ sequenceby RT-PCR. Analysis of the total cDNA revealed an open reading framethat encoded 563 amino acids in which the original CL-6 sequencecorresponded to amino acids 200–563 (Fig. S1A).

Homology search revealed a sequence similarity and structuralhomology between the novel MuSK interacting protein and the familyof RIN proteins, which currently consists of RIN1, 2 and 3 (Fig. S1B).Therefore, this protein was designated Rin-like (Rinl) (gene locus:Rinl). Similar to the so-far known family members Rinl contains anSH2 domain, an RH domain and a Vps9 domain. In contrast, the RA

Fig. 1. Rinl is a specific binding partner of MuSK. (A) A scheme of Rinl and corresponding truInteraction was assayed by growth of yeast clones on –HIS. (C) Rinl GST-deletion construct293T cells. Proteins were detected by immunoblotting (IB) using anti-myc antibodies. GST pin the right panel. (D) MuSK-myc and Rinl were expressed in HEK 293T cells andImmunoprecipitated proteins were assayed by immunoblotting using anti-myc andphosphotyrosine (PY). wt, wild-type; KD, kinase-dead; KA, kinase-active. IP, immunopreciRinl (Y2H) prey construct. Interaction was followed by growth of yeast clones on –HIS.

domain and proline-rich regions found in RIN1, 2 and 3 are notpresent in Rinl.

To determine the region(s) within Rinl necessary for interactionwith MuSK, we generated several Rinl deletion mutants fused to theGal4 activation domain (Fig. 1A). Using the reporter genes HIS andlacZ, interactions with the full-lengthMuSK cytoplasmic domain wereassayed in yeast. As shown in Fig. 1B, deletion mutants lacking eitherthe SH2 domain (Y2H and RH/Vps9) or the Vps9 domain (RH/ΔVps9)were able to interact with MuSK. In contrast, no interaction wasdetected with a deletion construct that encodes the Vps9 domain only(Vps9). We confirmed these findings by GST pulldowns of MuSKexpressed in HEK 293T cells using the same Rinl deletion constructsfused to GST (Fig. 1C). These results suggest that the Rinl/MuSKinteraction depends on the RH domain of Rinl.

The MuSK cytoplasmic domain consists of a short juxtamembraneregion, a tyrosine kinasedomain and an eight aminoacid C-terminal tail.Upon agrin stimulationMuSK becomes tyrosine-phosphorylated,whichinduces recruitment of downstream factors and consequently down-stream signaling [25,27,36]. To test whether the interaction betweenMuSKandRinl is phosphorylationdependent,we expressedMuSKwild-type, MuSK kinase-active or MuSK kinase-dead together with Rinl inHEK 293T cells. Co-immunoprecipitation of Rinl with MuSK wasdetected independent of MuSK phosphorylation (Fig. 1D and Fig. S2).Tonarrowdown thesite of interactionwithinMuSKwegeneratedMuSKdeletion andpointmutants fused to the lexA bindingdomain. Binding ofRinl to these MuSK mutant proteins was assayed in yeast using thereporter genes HIS and lacZ (Fig. 1E). Mutation of tyrosines in theautoactivation loop of the kinase domain (MuSK KD) do not affect Rinl/MuSK interaction. Similarly, the juxtamembrane tyrosine Y553 isdispensable for Rinl binding to MuSK. We did not include any

ncation mutants. (B) Yeast was transformed with bait and prey constructs as indicated.s were purified from bacteria and used for a pulldown of MuSK-myc expressed in HEKrotein input was assayed by Ponceau staining. 5% of the total MuSK-myc input is shownco-immunoprecipitated either with anti-MuSK antibodies or anti-Rinl antibodies.anti-Rinl antibodies, respectively. Phosphorylated MuSK was detected with anti-pitation. (E) Yeast was transformed with the indicated MuSK bait constructs and the

Page 5: Biochimica et Biophysica Acta - CORERin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and Rab22 Barbara Wollera, Susan Luiskandla, Milica

1202 B. Woller et al. / Biochimica et Biophysica Acta 1813 (2011) 1198–1210

truncations into the kinase domain since deletions of C-terminal partswill destroy the correct folding of the protein and consequentlynegatively influence our binding studies. We used however constructscarrying only the MuSK juxtamembrane region (JM) or the MuSKjuxtamembrane NPXY motif inserted in the TrkA cytoplasmic domain(MMT). These fusion proteins are unable to interact with Rinl. Takentogether, we conclude that Rinl binds to the MuSK kinase domain andthat this binding is independent of MuSK phosphorylation.

3.2. Rinl is ubiquitously expressed with highest expression in lymphoidorgans and specifically accumulated at NMSs

Weperformed RT-PCR using RNA isolated from adult mouse tissue.Rinl is expressed in all tested tissues with the highest expression inlung, spleen and thymus (Fig. 2A). These datawere confirmed by qPCRshowing that Rinl is up to 40 times more expressed in thymus thanmuscle (Fig. 2B). We also studied the expression of Rinl duringdevelopment and found a slight increase in expression in spleen andthymus within the first weeks after birth but no profound develop-mental regulation of Rinl gene expression in brain and muscle (Fig.S3A and B). To further extend these expression studies weinvestigated Rinl protein expression in thymus, spleen and muscle.Analysis by immunoblotting revealed a similar expression profile asdetected by RT-PCR. High expression was found in thymus and spleencompared to a weak expression in muscle (Fig. S3C).

Proteins important for NMS development are usually enriched atsynaptic sites.We therefore testedwhetherRinl is localized atNMSs.Westained muscle sections with antibodies against Rinl and Alexa-594conjugated α-BGT (Fig. 2C and S4). As shown in Fig. 2C, a weak butspecific enrichment of Rinl at NMSs is detectable.

3.3. Rinl acts as GEF for Rab5a

The family of RIN proteins is characterized by its Vps9 domain andits binding affinity for Rab5a. All so far known RIN proteins are able to

Fig. 2. Rinl expression in different tissues and localization at the NMS. (A) RT-PCR was perfofrom cDNAs generated from different tissue RNAs was performed. Spleen was set to 1. s.e.m594-conjugated α-BGT. Images were taken on a confocal microscope. Scale bar, 20 μm.

catalyze the GDP to GTP exchange on Rab5a [3,15,37]. Firstexperiments were performed using the yeast two-hybrid system todetermine whether Rinl binds to Rab5a. The dominant-negative formRab5a S34N showed a strong interaction with Rinl. In contrast, Rinldid not interact with Rab5a wild-type and the constitutively-activeRab5a Q79L variant (Fig. 3A). Dominant-negative mutations areknown to reduce the nucleotide affinity of the G-protein, resulting inincreased concentrations of nucleotide free G-protein which bindswith high affinity to its GEFs. The observed interaction profile thussuggests that Rinl might be a Rab5a GEF. To map the site of interactionwe used the Rinl mutant constructs described in Fig. 1A and foundthat the N-terminal region is dispensable for Rinl/Rab5a interaction.However, deletion of the Vps9 domain or the RH domain abolishedthe binding of Rinl to Rab5a S34N suggesting that both the RH andVps9 domain are required for interaction (Fig. 3A and B). Thesefindings were confirmed by GST pulldowns of Rab5a expressed inCOS7 cells using the same Rinl deletion constructs fused to GST(Fig. 3C).We further tested the interaction between Rinl and Rab5a byco-immunoprecipitation from HEK 293T cells transfected with Rinland Rab5a wild-type, Rab5a S34N or Rab5a Q79L, respectively. Rab5aS34N efficiently co-immunoprecipitates with Rinl, whereas Rab5awild-type and Rab5a Q79L show a weak or no interaction (Fig. 3D). Incontrast, Rinl does not bind to the late endosomal marker Rab7a,independent of its activation status.

As Rinl contains a classical Vps9 domain and binds preferentially tonucleotide free Rab5a, we next asked whether Rinl indeed acts as GEFfor Rab5a. Recombinant proteins were expressed in bacteria and usedin an in vitro assay to measure nucleotide exchange. In brief, the RabGTPase was loaded with the fluorescent nucleotide analog mGDP. Thefluorescence intensity of mGDP is approximately twice as high ifbound to the hydrophobic environment of a protein as if exposedfreely to the buffer solution. The exchange of mGDP in the presence ofexcess unlabeled GDP can thus be measured in real time asfluorescence signal decay. As shown in Fig. 3E, the addition of Rinlaccelerates nucleotide exchange in a concentration dependent

rmed from different mouse tissue samples. Actin was used as quality control. (B) qPCR. is shown. (C) Mouse muscle sections were stained with anti-Rinl antibodies and Alexa

Page 6: Biochimica et Biophysica Acta - CORERin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and Rab22 Barbara Wollera, Susan Luiskandla, Milica

Fig. 3. Rinl preferentially interacts with nucleotide free Rab5a and acts as GEF for Rab5a. (A) Yeast was transformed with the indicated Rab5a bait constructs and Rinl full-length anddeletion prey constructs. Interaction was screened by growth on –HIS. (B) Table showing the activation of the reporter genes HIS and lacZ. (C) Rinl GST-deletion constructs werepurified from bacteria and used for a pulldown of GFP-Rab5a (wt, S34N, Q79L) expressed in COS7 cells. Proteins were detected by immunoblotting (IB) using anti-GFP antibodies. 5%of the total Rab5a lysates are shown as input. (D) Extracts of transiently transfected HEK 293T cells were used to immunoprecipitate Rinl with anti-Rinl antibodies. Co-immunoprecipitated Rab5a was detected by immunoblotting with anti-GFP. No Rab7a co-immunoprecipitation is detected. 10% of the input is shown (total lysate). IP,immunoprecipitation. (E) 200 nM Rab5a or Rab7a loaded with mGDP were incubated with increasing amounts of Rinl in the presence of 20 μM unlabeled GDP. The exchange ofmGDP for GDPwasmeasured as decay in fluorescence signal. In case of Rab7a proper nucleotide loadingwas demonstrated by the addition of EDTA at the indicated time point, whichinduces the release of nucleotides and a rapid decay in the fluorescence signal.

1203B. Woller et al. / Biochimica et Biophysica Acta 1813 (2011) 1198–1210

manner by several orders of magnitude. In contrast, no exchangeactivity of Rinl for Rab7a could be detected. To validate propernucleotide loading of Rab7a, the Mg2+ chelator EDTA was added tothe control reaction at the indicated time point. This induces therelease of nucleotides from G-proteins as their binding is Mg2+

dependent. These data demonstrate that Rinl similar to the othermembers of the RIN protein family acts as GEF for Rab5a.

3.4. Rinl colocalizes with dominant-negative Rab5a to cytoskeletal-richmembrane ruffles

To examine the subcellular distribution of Rinl COS7 cells weretransfectedwithmyc-tagged Rinl (Fig. 4A).We found Rinl localized to avariety of different compartments. To analyze the distribution in moredetail we divided Rinl localization into four categories and quantifiedthe distributionwithin these categories. As shown in Fig. 4A, around90%

of Rinl is localized either diffusely and/or in vesicles. The remaining 10%of Rinl show a ruffle-like distribution. This localization pattern is thesame for GFP-tagged Rinl or untagged Rinl (Fig. S5). Rinl shows no co-localization with early endosomes, recycling and late endosomes or theGolgi (Fig. S6). Interestingly, upon co-expression of Rinl and Rab5aS34N, Rinl and Rab5a are co-localized predominantly to membraneruffles (Fig. 4B and S7A). Similarly, a weak co-localization between Rinland Rab5a wt is detectable at membrane-ruffles. This co-localization isincreasedwhenRinl lacking the SH2domain is co-expressedwithRab5awt (Fig. S7B). Most importantly, Rab5a S34N becomes redistributedfrom a diffuse cytoplasmic localization to either ruffle-like or vesicular-like structures. Membrane ruffles have been implicated in endocytoticprocesses associatedwith cytoskeletal rearrangements. To testwhetheractin is concentrated within the Rinl/Rab5a-positive ruffles we stainedcells with phalloidin. Fig. 4C shows a co-localization of actin with Rinland Rab5a. Moreover, disruption of the cytoskeleton using cytochalasin

Page 7: Biochimica et Biophysica Acta - CORERin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and Rab22 Barbara Wollera, Susan Luiskandla, Milica

1204 B. Woller et al. / Biochimica et Biophysica Acta 1813 (2011) 1198–1210

Page 8: Biochimica et Biophysica Acta - CORERin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and Rab22 Barbara Wollera, Susan Luiskandla, Milica

Fig. 5. Rinl preferentially interacts with GDP-bound Rab22 and acts as GEF for Rab22. (A) Yeast was transformed with Rab22 bait constructs (wt, S19N, Q64L) and Rinl full-length ordeletion prey plasmids. Growth on –HIS is shown. (B) The interaction between Rab22 and Rinl was assayed by growth on –HIS and LacZ expression (β-gal assay). (C) Extracts oftransiently transfected HEK 293T cells were used to immunoprecipitate Rinl with anti-Rinl antibodies. Co-immunoprecipitated Rab22 was detected by immunoblotting (IB) withanti-GFP. Rab7a was used as negative control. 10% of the input is shown in the top panel (total lysate). (D) 200 nM ofmGDP loaded Rab22were incubated with increasing amounts ofRinl in the presence of 20 μM unlabeled GDP. The exchange of mGDP for GDP was measured in real time as decay in fluorescence signal. (E) The velocity of the nucleotide exchangereaction (kobs) toward Rab5a and Rab22 were determined and plotted against the concentration of Rinl.

1205B. Woller et al. / Biochimica et Biophysica Acta 1813 (2011) 1198–1210

D blocked the formation of membrane ruffles and caused anaccumulation of Rinl and Rab5a in actin-positive aggregates.

3.5. Rinl acts as GEF for Rab22 and co-localizes with Rab22 to actin-richdomains

Rab22 is the closest homologue of Rab5 with 52% sequenceidentity [38]. Like Rab5a, Rab22 has been localized to early endosomesbut its role during endocytosis is so far not well understood. SinceVps9 domain containing proteins have been reported to activateRab5a and its homologues Rab21 and Rab22, we tested the interactionof Rinl with Rab22 [6]. Using the yeast two-hybrid systemwe detect astrong interaction between Rinl and the dominant-negative form of

Fig. 4. Subcellular localization of Rinl and co-localization with Rab5a and actin. (A) myc-taggand myc. Representative confocal images are shown. Scale bar, 25 μm. Transfected cells wercategories. A quantification is shown (nN100). Error bars, s.e.m. (B) GFP-Rab5a S34N alonequantification of the Rab5a S34N distribution was performed as described in A (n=50). ErroDMSO for 30 minutes. Cells were stained with anti-myc antibodies to detect Rinl and withimages are shown. Scale bar, 25 μm.

Rab22. Moreover, the binding of Rinl to Rab22 is dependent on the RHand Vps9 domain but independent of the N-terminal region (Fig. 5Aand B). The Rinl/Rab22 interaction was confirmed by co-immunopre-cipitation experiments in transfected HEK 293T cells. Dominant-negative Rab22 S19N efficiently co-immunoprecipitates with Rinlupon co-expression, whereas Rab22 wild-type and constitutivelyactive Rab22 Q64L show a weak or no interaction (Fig. 5C). Todetermine whether Rinl acts indeed as GEF for Rab22, the GEF activityof Rinl toward Rab22 was measured in vitro (Fig. 5D). Rinl stronglyaccelerates nucleotide exchange of Rab22 and can therefore beclassified as a GEF of Rab22. For a more quantitative comparison ofthe Rinl activity toward Rab5a and Rab22, the rates of the nucleotideexchange reaction, kobs, were determined from measurements as

ed Rinl was transiently expressed in COS7 cells and stained with antibodies against Rinle assayed for their Rinl expression patterns. Expression patterns were divided into fouror together with myc-tagged Rinl were co-expressed in COS7 cells. Scale bar, 25 μm. Ar bars, s.e.m. (C) Transiently transfected COS7 cells were treated with cytochalasin D orTRITC-conjugated phalloidin to label the actin cytoskeleton. Representative confocal

Page 9: Biochimica et Biophysica Acta - CORERin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and Rab22 Barbara Wollera, Susan Luiskandla, Milica

1206 B. Woller et al. / Biochimica et Biophysica Acta 1813 (2011) 1198–1210

presented in Figs. 3E and 5D. The dependency of kobs on the Rinlconcentration is presented in Fig. 5E. As can be seen, nucleotideexchange toward Rab22 is more sensitive to the concentration of Rinl,which thus displays a higher catalytic rate for Rab22 than for Rab5a.Moreover, Rinl and Rab22 S19N co-localize to similar compartmentsupon co-expression in COS7 cells (Fig. S7). Like for Rab5a, Rinl onlyweakly co-localizes with Rab22 wt whereas Rinl lacking the SH2domain robustly co-localizes with Rab22 wt (Fig. S7B). Rinl induces aredistribution of diffusely localized Rab22 to ruffle-like structures(Fig. 6A). Similar to Rab5a and Rinl, also Rab22 and Rinl co-localize toactin-positive structures (Fig. 6B).

Fig. 6. Rab22 and Rinl co-localize to actin-positive membrane ruffles. (A) GFP-Rab22 S19Nconfocal images are shown. Scale bar, 25 μm. Rab22 expression, when transfected alone or twere quantified as described in Fig. 5. nN50; Error bars, s.e.m. (B) Transiently transfected Cwith anti-myc antibodies to detect Rinl and with TRITC-conjugated phalloidin to label the a

3.6. Rinl recruits Rab5a and Rab22 to the cytoskeleton via the Vps9 domain

Since we detected a significant redistribution of dominant-negativeRab5a and Rab22 upon co-expressionwith Rinl, we next asked how thislocalization is influenced by Rinl deletion mutants. For that we usedconstructs that either lack theN-terminal SH2domain or the C-terminalVps9 domain. These truncationswere co-expressedwith Rab5a S34N orRab22 S19N and the localization assayed by immunostaining. Rinllacking the SH2domain (ΔSH2) localizes to vesicles and ruffles togetherwith Rab5a and Rab22 (Fig. 7). In particular, Rinl ΔSH2 has a morepronounced localization to ruffles compared to full-lengthRinl (data not

alone or together with myc-tagged Rinl were expressed in COS7 cells. Representativeogether with Rinl, was assayed by confocal microscopy. Subcellular expression patternsOS7 cells were treated with cytochalasin D or DMSO for 30 minutes. Cells were stainedctin cytoskeleton. Scale bar, 25 μm.

Page 10: Biochimica et Biophysica Acta - CORERin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and Rab22 Barbara Wollera, Susan Luiskandla, Milica

Fig. 7. Rinl-dependent recruitment of Rab5a and Rab22 to the cytoskeleton. (A) myc-tagged Rinl (ΔSH2 and ΔVps9) and GDP-bound Rab5a or Rab22 were co-transfected into COS7cells. Cells were stained with anti-myc antibodies and visualized by confocal microscopy. Representative images are shown. Scale bar, 25 μm. (B) The distribution of Rab5a and Rab22was quantified as described in Fig. 5. nN35; Error bars, s.e.m.

1207B. Woller et al. / Biochimica et Biophysica Acta 1813 (2011) 1198–1210

Page 11: Biochimica et Biophysica Acta - CORERin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and Rab22 Barbara Wollera, Susan Luiskandla, Milica

1208 B. Woller et al. / Biochimica et Biophysica Acta 1813 (2011) 1198–1210

shown). Moreover, Rinl ΔSH2 shows a higher degree of co-localizationwith Rab5a or Rab22 to ruffles than full-length Rinl and Rab5a/Rab22(compare Figs. 4B and 6A to 7B). This suggests that the SH2 domainmight have an inhibitory effect on the interaction between Rinl and Rabproteins. In contrast, Rinl lacking theVps9 domain (ΔVps9) does not co-localizewith Rab5a S34N and Rab22 S19N (Fig. 7). Rab5a and Rab22 areunchanged upon co-expression with Rinl ΔVps9 and remain diffuselydistributed and/or in vesicles. Rinl ΔVps9 itself is localized mainlythroughout the cytoplasmor in vesicles but does not attach to actin-richmembrane ruffles. Therefore, the Vps9 domain is not only responsiblefor Rab5a/Rab22 activationbut alsomediates the localization of Rinl andRab proteins to the cytoskeleton.

3.7. Rinl regulates fluid-phase and receptor-mediated endocytosis

Since RIN family members have previously been implicated inendocytotic processes we set out to study the function of Rinl duringfluid-phase endocytosis. For that we examined the HRP uptake in HEK293T cells expressing Rinl, Rab5a wt, Rab5a wt together with Rinl, orGFP as a control. Expression of Rinl and Rab5a induced theinternalization of HRP (Fig. 8A). The co-expression of Rab5a wt andRinl induced a similar degree of HRP uptake than expression of Rinl orRab5a wt alone.

It has previously been demonstrated that RIN1 acts as regulator ofEGFR endocytosis [12,39]. We therefore asked whether Rinl also affectsEGFR internalization. We used Alexa 555-conjugated EGF to labelendogenous EGFR in HeLa cells expressing Rinl, Rab5a wt, Rab5a S34N,Rinl togetherwithRab5awt, orGFP as a control. EGFuptake at 37 °Cwasimaged at various times. As shown in Figs. 8B and S8, in control cellsEGFR endocytosis is rapidly induced within 5 minutes and perinuclear

Fig. 8. Rinl increases the internalization of HRP and EGFR. (A) HEK 293T cells expressing GFHRP uptake was quantified from two independent experiments done in triplicates. Error barGFP-Rab5a wt or GFP-Rab5a S34N. Cells were incubated with Alexa 555-conjugated EGF at 4images are shown. Scale bar, 25 μm. The GFP signals of the equivalent cells are shown in Fi

accumulations of EGF-positive vesicles is prominent by 15 minutes.Expression of Rab5a wt or Rinl leads to an acceleration of EGFRendocytosis and a decrease in EGF-positive vesicles. This increase inEGFR endocytosis is especially pronounced in cells co-expressing Rab5awt and Rinl. In contrast, cells expressing Rab5a S34N show a reducedEGFR endocytosis. These results support a functional role of Rinl duringearly endocytotic processes.

4. Discussion

The GTPase Rab5a is the key regulator during early endocytoticprocesses. Therefore, it is of particular interest to identify mechanismsand molecules that modulate Rab5a action. In this study we report theidentification and characterization of Rinl, a novel Rab5a GEF, whichshows a high homology to the family of RIN proteins. Rinl was isolatedvia its interactionwith theRTKMuSK. It specifically interactswithMuSKthrough the central portion of the protein containing the RH domain.Rinl interacts and co-localizes with Rab5a and catalyzes GDP/GTPexchange onRab5a. Similar biochemical and enzymatic propertiesweredemonstrated toward Rab22. Furthermore, we identified the Vps9domain of Rinl as a critical molecular determinant that controls therecruitment of Rab5a and Rab22 to cytoskeletal membrane compart-ments. Most importantly, Rinl stimulates fluid-phase uptake and EGFRendocytosis.

The formation of the NMS is crucially linked to signal transductionevents induced by the muscle-specific RTK MuSK [18]. MuSKactivation via agrin/Lrp4 induces a signaling cascade that leads topost- as well as presynaptic differentiation. Several MuSK bindingproteins have been identified which include adaptor proteins likeDok7, kinases like Abl and scaffolding proteins including Magi-1c and

P-Rinl, GFP-Rab5a wt, GFP-Rinl and GFP-Rab5a wt or GFP alone were loaded with HRP.s, s.e.m. (B) HeLa cells were transfected with GFP, GFP-Rinl, GFP-Rab5a wt, GFP-Rinl and°C and EGF uptake at 37 °C was imaged after 5 and 15 minutes. Representative confocalg. S8.

Page 12: Biochimica et Biophysica Acta - CORERin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and Rab22 Barbara Wollera, Susan Luiskandla, Milica

1209B. Woller et al. / Biochimica et Biophysica Acta 1813 (2011) 1198–1210

ColQ [18]. More recently the trafficking protein NSF and the E3ubiquitin ligases PDZRN3 and PAUL were isolated as MuSK interactionpartner, which are thought to regulate MuSK endocytosis anddegradation, respectively [40–42]. Here we identified Rinl as novelinteraction partner of MuSK. Binding of Rinl to MuSK requires theinternal RH domain of Rinl but is independent of MuSK phosphory-lation. This suggests that Rinl binding is not associated with MuSKactivation. However, a regulation of Rinl/MuSK interaction dependenton the subcellular localization appears likely since we show a specificlocalization of Rinl to vesicles andmembrane ruffles. In support of thishypothesis we detect a specific co-localization of MuSK and Rinl atactin-positive membrane ruffles (data not shown). The identificationof Rinl as binding partner ofMuSK appeared of distinct interest since itwas recently demonstrated that MuSK endocytosis regulates MuSKsignaling [42]. However so far, we have not been able to correlate Rinlaction and MuSK function. Rinl is very weakly expressed in musclecells questioning the importance of Rinl in cultured muscle cells.Nevertheless, this does not exclude the possibility that Rinl plays arole during MuSK-dependent signal transduction in vivo, either at theNMS or in the brain, where MuSK function during memory formationhas been implicated recently [43]. A gene targeting approach in micewill be necessary to answer these questions. Furthermore, a functionalcompensation by one of the other RIN family members cannot beruled out at the moment.

The RIN familymembers have been implicated in early endocytoticevents [1]. In particular, the role of RIN1 during receptor endocytosishas been demonstrated many-fold [12–14,39]. Here we are able toshow that Rinl overexpression accelerates fluid-phase endocytosis aswell as EGFR endocytosis. These data support our in vitro data andprovide first hints on the physiological role of Rinl. RIN1 has beenpostulated as crucial regulator of EGFR endocytosis and signaling[12,39,44]. Our findings raise the questions whether Rinl represents asimilarly important regulator of EGFR endocytosis and whether Rinland RIN1 play complementary roles during EGFR endocytosis due totheir differential expression patterns.

The currentmembers of theRINprotein family share anSH2domain,anRH region, a Vps9 domain and anRAdomain. RIN1was isolated by itsability to bind to H-Ras via the RA domain thereby competing with Raf1and inhibiting Ras action [2,45]. In addition, the RA domain has beenimplicated in the interaction with the EGFR via Ras, which leads to arecruitment of RIN1 to the activated receptor linking internalized EGFRto Rab5a-positive endosomes [3]. Rinl lacks an RA domain. Therefore itappears unlikely that Rinl acts as an effector for Ras proteins. However, arecruitment to activated RTKs can still occur through the N-terminalSH2domain,whichwould then bring the intracellular receptors to earlyendosomes. Such an SH2-dependent interaction has been shown forRIN1 and EGFR as well as RIN1 and EphA4 [12,13].

The RIN proteins belong to the family of Vps9 domain containingproteins. These proteins are characterized by their ability to bind Rab5aand to catalyze the GDP/GTP exchange on Rab5a [1]. Crystallizationstudies on Rabex-5 have shown that an N-terminal helical bundle inaddition to the Vps9 domain is required for GEF activity, the so-calledHB-Vps9 tandem [6]. The helical bundle conforms to the RH domain inthe RIN proteins. Consistent with the Rabex-5 data, it was shown that asplice-variant of RIN1 lacking part of the helical bundle is unable tointeract with dominant-negative Rab5a. Likewise, Rinl truncationsdeleting the RH domain/helical bundle do not bind dominant-negativeRab5a implicating similar biochemical properties for all Vps9 domaincontaining proteins. It was also reported that full-length Vps9 domaincontaining proteins including the RIN proteins have a reduced bindingactivity and/or GEF activity for Rab5a [6,46]. This suggests thatautoinhibition by regulatory elements in the N- and/or C-terminusplay a role. Similarly, we find a reduced binding activity for Rab5a andRab22 in full-length Rinl constructs compared to Rinl constructscarrying only the RH and Vps9 domain. Therefore, the N-terminal SH2domain might represent such an inhibitory element. Protein interac-

tions via the SH2 domain would then relief the autoinhibition andinduce exchange activity. So far however, it remains unclear whichproteins bind to the SH2 domain of Rinl.

Analysis of the Rabex-5 HB-Vps9 tandem revealed a high specificitytoward the Rab5 subfamily but also showed a selective exchangeactivity for the different Rab5 subfamily members: strong GEF activityfor Rab5 and Rab21, a weak activity for Rab22 [6]. This specificity andselectivity is achieved on one hand by conserved exchange determi-nants on a common surface of the Vps9 domain and on the other handby invariant aromatic residues in the switch regions of the Rab GTPases.Delprato and colleagues also reported a similar catalytic activity andRabspecificity for RIN1 [6]. In this studywe show that Rinl specifically bindsRab5a and Rab22, and there preferentially the nucleotide free forms.Rinl acts as GEF for Rab22 and Rab5a, surprisingly however, presents ahigher efficiency for Rab22. This distinguishes Rinl from RIN1 andsuggests that different Vps9 domains have different specificity profilesfor the Rab5 subfamily. This of course also raises the question whetherthis specificity is also represented at a physiological level wherebydifferent RIN proteins regulate different Rab5 subfamily dependentprocesses. Four amino acids (D313, P317, Y354 and T357) in the Vps9domain in Rabex-5 have been shown to be essential for exchangeactivity and these residues are highly conserved among different Vps9domains [6]. Likewise, the Rinl Vps9 domain contains these conservedamino acids (D456, P460, Y497 and T500). Other residues, which havebeen shown to lie within the binding site for the GTPase differ betweenRabex-5 and Rinl. These residues are expected to be responsible for thedifferences in the specificity profile.

When studying the subcellular distribution of Rinl protein wenoticed a characteristic localization in vesicles and membrane ruffles.Moreover, co-expression of Rinl and Rab5a or Rab22 mutants withlowered nucleotide affinity leads to a redistribution of diffuselyexpressed Rab5a andRab22 to actin-positivemembrane compartments.The localization of Rinl tomembrane ruffles aswell as the recruitment ofRab5a and Rab22 to the cytoskeleton are dependent on the Vps9domain. It is unclear at themomentwhether the Vps9 domain interactsdirectly or indirectly (via a so-far unknown actin binding protein) withthe cytoskeleton. Rab mutants with lowered nucleotide affinity areknown to bind GEFs with high affinity, thereby trapping the active GEFin an unproductive complex [47]. The ability of Rinl to recruit Rab5aS34N and Rab22 S19N indicates that Rinl interacts with the actincytoskeleton in an active state. Thus it is expected that Rinl activates Rabproteins locally at the cytoskeleton. Actin cytoskeleton remodeling hasbeen implicated in early endocytosis as well as recycling [48].Furthermore, membrane ruffling is a characteristic of actin remodelingand is closely associated with regions of active endocytosis [49]. Ourfindings therefore support a model whereby Rinl-dependent activationof Rab5a and Rab22 at the cytoskeleton regulates early endocytotic and/or recycling processes. Future experiments will have to show how actinremodeling, exchange activity by Rinl and Rab5a/Rab22-dependentendocytosis are connected.

Supplementarymaterials related to this article can be found onlineat doi:10.1016/j.bbamcr.2011.03.005.

Acknowledgments

We are grateful to Jeffrey Chamberlain for providing us with themouse muscle prey library. We would like to thank WalterBirchmeier, Cecilia Bucci, Steve Burden, Judy Donaldson, SaidHashemolhosseini and Marino Zerial for providing plasmids. Manythanks also go to Marije Rensen-De Leeuw for technical assistance.Wilfried Ellmeier provided useful comments on the manuscript. Thiswork was supported by the Austrian Science Fund (P19223-B09 to R.H.) and by the Chemical Sciences of the Netherlands Organization forScientific Research (NOW-CW to H.R.). B.W. was supported by theDOC-fFORTE Programme from the Austrian Academy of Sciences. M.P.was supported by the TI Pharma Project T3-106.

Page 13: Biochimica et Biophysica Acta - CORERin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and Rab22 Barbara Wollera, Susan Luiskandla, Milica

1210 B. Woller et al. / Biochimica et Biophysica Acta 1813 (2011) 1198–1210

References

[1] D.S. Carney, B.A. Davies, B.F. Horazdovsky, Vps9 domain-containing proteins:activators of Rab5 GTPases from yeast to neurons, Trends Cell Biol. 16 (2006)27–35.

[2] L. Han, J. Colicelli, A human protein selected for interference with Ras functioninteracts directly with Ras and competes with Raf1, Mol. Cell. Biol. 15 (1995)1318–1323.

[3] G.G. Tall, M.A. Barbieri, P.D. Stahl, B.F. Horazdovsky, Ras-activated endocytosis ismediated by the Rab5 guanine nucleotide exchange activity of RIN1, Dev. Cell 1(2001) 73–82.

[4] H. Hama, G.G. Tall, B.F. Horazdovsky, Vps9p is a guanine nucleotide exchangefactor involved in vesicle-mediated vacuolar protein transport, J. Biol. Chem. 274(1999) 15284–15291.

[5] G. Vitale, K. Alexandrov, O. Ullrich, H. Horiuchi, A. Giner, C. Dobson, O. Baykova, H.Gournier, H. Stenmark, M. Zerial, The GDP/GTP cycle of Rab5 in the regulation ofendocytotic membrane traffic, Cold Spring Harb. Symp. Quant. Biol. 60 (1995)211–220.

[6] A. Delprato, E. Merithew, D.G. Lambright, Structure, exchange determinants, andfamily-wide rab specificity of the tandem helical bundle and Vps9 domains ofRabex-5, Cell 118 (2004) 607–617.

[7] M. Zerial, H. McBride, Rab proteins as membrane organizers, Nat. reviews 2(2001) 107–117.

[8] M. Kauppi, A. Simonsen, B. Bremnes, A. Vieira, J. Callaghan, H. Stenmark, V.M.Olkkonen, The small GTPase Rab22 interacts with EEA1 and controls endosomalmembrane trafficking, J. Cell Sci. 115 (2002) 899–911.

[9] J.C. Simpson, G. Griffiths, M. Wessling-Resnick, J.A. Fransen, H. Bennett, A.T. Jones,A role for the small GTPase Rab21 in the early endocytic pathway, J. Cell Sci. 117(2004) 6297–6311.

[10] R. Weigert, A.C. Yeung, J. Li, J.G. Donaldson, Rab22a regulates the recycling ofmembrane proteins internalized independently of clathrin, Mol. Biol. Cell 15(2004) 3758–3770.

[11] F.J. Opdam, A. Echard, H.J. Croes, J.A. van den Hurk, R.A. van de Vorstenbosch, L.A.Ginsel, B. Goud, J.A. Fransen, The small GTPase Rab6B, a novel Rab6 subfamilymember, is cell-type specifically expressed and localised to the Golgi apparatus, J.Cell Sci. 113 (Pt 15) (2000) 2725–2735.

[12] M.A. Barbieri, C. Kong, P.I. Chen, B.F. Horazdovsky, P.D. Stahl, The SRC homology 2domain of Rin1 mediates its binding to the epidermal growth factor receptor andregulates receptor endocytosis, J. Biol. Chem. 278 (2003) 32027–32036.

[13] K. Deininger, M. Eder, E.R. Kramer, W. Zieglgansberger, H.U. Dodt, K. Dornmair, J.Colicelli, R. Klein, The Rab5 guanylate exchange factor Rin1 regulates endocytosisof the EphA4 receptor in mature excitatory neurons, Proc. Natl Acad. Sci. U.S.A.105 (2008) 12539–12544.

[14] C.M. Hunker, H. Giambini, A. Galvis, J. Hall, I. Kruk, M.L. Veisaga, M.A. Barbieri, Rin1regulates insulin receptor signal transduction pathways, Exp. Cell Res. 312 (2006)1106–1118.

[15] H. Kajiho, K. Saito, K. Tsujita, K. Kontani, Y. Araki, H. Kurosu, T. Katada, RIN3: anovel Rab5 GEF interacting with amphiphysin II involved in the early endocyticpathway, J. Cell Sci. 116 (2003) 4159–4168.

[16] W.H. Lai, P.H. Cameron, I. Wada, J.J. Doherty 2nd, D.G. Kay, B.I. Posner, J.J. Bergeron,Ligand-mediated internalization, recycling, and downregulation of the epidermalgrowth factor receptor in vivo, J. Cell Biol. 109 (1989) 2741–2749.

[17] A. Sorkin, M. von Zastrow, Endocytosis and signalling: intertwining molecularnetworks, Nat. reviews 10 (2009) 609–622.

[18] H.Wu,W. Xiong, M. Lin, To build a synapse: signaling pathways in neuromuscularjunction assembly, Dev. Camb. Engl. 137 (2010) 1017–1033.

[19] D.J. Glass, D.C. Bowen, T.N. Stitt, C. Radziejewski, J. Bruno, T.E. Ryan, D.R. Gies, S.Shah, K. Mattsson, S.J. Burden, P.S. DiStefano, D.M. Valenzuela, T.M. DeChiara, G.D.Yancopoulos, Agrin acts via a MuSK receptor complex, Cell 85 (1996) 513–523.

[20] N. Kim, A.L. Stiegler, T.O. Cameron, P.T. Hallock, A.M. Gomez, J.H. Huang, S.R.Hubbard, M.L. Dustin, S.J. Burden, Lrp4 is a receptor for Agrin and forms a complexwith MuSK, Cell 135 (2008) 334–342.

[21] B. Zhang, S. Luo, Q. Wang, T. Suzuki, W.C. Xiong, L. Mei, LRP4 serves as a coreceptorof agrin, Neuron 60 (2008) 285–297.

[22] T.M. DeChiara, D.C. Bowen, D.M. Valenzuela, M.V. Simmons, W.T. Poueymirou, S.Thomas, E. Kinetz, D.L. Compton, E. Rojas, J.S. Park, C. Smith, P.S. DiStefano, D.J.Glass, S.J. Burden, G.D. Yancopoulos, The receptor tyrosine kinase MuSK isrequired for neuromuscular junction formation in vivo, Cell 85 (1996) 501–512.

[23] M. Gautam, P.G. Noakes, L. Moscoso, F. Rupp, R.H. Scheller, J.P. Merlie, J.R. Sanes,Defective neuromuscular synaptogenesis in agrin-deficient mutant mice, Cell 85(1996) 525–535.

[24] S.D. Weatherbee, K.V. Anderson, L.A. Niswander, LDL-receptor-related protein 4 iscrucial for formation of the neuromuscular junction, Dev. Camb. Engl. 133 (2006)4993–5000.

[25] A. Watty, G. Neubauer, M. Dreger, M. Zimmer, M. Wilm, S.J. Burden, The in vitroand in vivo phosphotyrosine map of activated MuSK, Proc. Natl Acad. Sci. U.S.A. 97(2000) 4585–4590.

[26] B. Mossier, M. Togel, K. Fuchs,W. Sieghart, Immunoaffinity purification of gamma-aminobutyric acidA (GABAA) receptors containing gamma 1-subunits. Evidencefor the presence of a single type of gamma-subunit in GABAA receptors, J. Biol.Chem. 269 (1994) 25777–25782.

[27] R. Herbst, S.J. Burden, The juxtamembrane region of MuSK has a critical role inagrin-mediated signaling, EMBO J. 19 (2000) 67–77.

[28] D.J. Glass, E.D. Apel, S. Shah, D.C. Bowen, T.M. DeChiara, T.N. Stitt, J.R. Sanes, G.D.Yancopoulos, Kinase domain of the muscle-specific receptor tyrosine kinase(MuSK) is sufficient for phosphorylation but not clustering of acetylcholinereceptors: required role for the MuSK ectodomain? Proc. Natl Acad. Sci. U.S.A. 94(1997) 8848–8853.

[29] C. Chen, H. Okayama, High-efficiency transformation of mammalian cells byplasmid DNA, Mol. Cell. Biol. 7 (1987) 2745–2752.

[30] U. Schaeper, N.H. Gehring, K.P. Fuchs, M. Sachs, B. Kempkes, W. Birchmeier,Coupling of Gab1 to c-Met, Grb2, and Shp2 mediates biological responses, J. CellBiol. 149 (2000) 1419–1432.

[31] C. Lumeng, S. Phelps, G.E. Crawford, P.D. Walden, K. Barald, J.S. Chamberlain,Interactions between beta 2-syntrophin and a family of microtubule-associatedserine/threonine kinases, Nat. Neurosci. 2 (1999) 611–617.

[32] A.B. Vojtek, S.M. Hollenberg, Ras–Raf interaction: two-hybrid analysis, Meth.Enzymol. 255 (1995) 331–342.

[33] V. Nizhynska, R. Neumueller, R. Herbst, Phosphoinositide 3-kinase acts throughRAC and Cdc42 during agrin-induced acetylcholine receptor clustering, Dev.Neurobiol. 67 (2007) 1047–1058.

[34] C. Schnatwinkel, S. Christoforidis, M.R. Lindsay, S. Uttenweiler-Joseph,M.Wilm, R.G.Parton,M. Zerial, The Rab5 effector Rabankyrin-5 regulates and coordinates differentendocytic mechanisms, PLoS Biol. 2 (2004) E261.

[35] H. Rehmann, Characterization of the activation of the Rap-specific exchange factorEpac by cyclic nucleotides, Meth. Enzymol. 407 (2006) 159–173.

[36] H. Zhou, D.J. Glass, G.D. Yancopoulos, J.R. Sanes, Distinct domains ofMuSKmediateits abilities to induce and to associate with postsynaptic specializations, J. Cell Biol.146 (1999) 1133–1146.

[37] K. Saito, J. Murai, H. Kajiho, K. Kontani, H. Kurosu, T. Katada, A novel bindingprotein composed of homophilic tetramer exhibits unique properties for the smallGTPase Rab5, J. Biol. Chem. 277 (2002) 3412–3418.

[38] J.B. Pereira-Leal, M.C. Seabra, Evolution of the Rab family of small GTP-bindingproteins, J. Mol. Biol. 313 (2001) 889–901.

[39] C. Kong, X. Su, P.I. Chen, P.D. Stahl, Rin1 interacts with signal-transducing adaptormolecule (STAM) and mediates epidermal growth factor receptor trafficking anddegradation, J. Biol. Chem. 282 (2007) 15294–15301.

[40] P.A. Bromann, J.A. Weiner, E.D. Apel, R.M. Lewis, J.R. Sanes, A putative ariadne-likeE3 ubiquitin ligase (PAUL) that interacts with the muscle-specific kinase (MuSK),Gene Expr. Patterns 4 (2004) 77–84.

[41] Z. Lu, H.S. Je, P. Young, J. Gross, B. Lu, G. Feng, Regulation of synaptic growth andmaturation by a synapse-associated E3 ubiquitin ligase at the neuromuscularjunction, J. Cell Biol. 177 (2007) 1077–1089.

[42] D. Zhu, Z. Yang, Z. Luo, S. Luo, W.C. Xiong, L. Mei, Muscle-specific receptor tyrosinekinase endocytosis in acetylcholine receptor clustering in response to agrin, J.Neurosci. 28 (2008) 1688–1696.

[43] A. Garcia-Osta, P. Tsokas, G. Pollonini, E.M. Landau, R. Blitzer, C.M. Alberini, MuSKexpressed in the brain mediates cholinergic responses, synaptic plasticity, andmemory formation, J. Neurosci. 26 (2006) 7919–7932.

[44] M.A. Barbieri, S. Fernandez-Pol, C. Hunker, B.H. Horazdovsky, P.D. Stahl, Role ofrab5 in EGF receptor-mediated signal transduction, Eur. J. Cell Biol. 83 (2004)305–314.

[45] Y. Wang, R.T. Waldron, A. Dhaka, A. Patel, M.M. Riley, E. Rozengurt, J. Colicelli, TheRAS effector RIN1 directly competes with RAF and is regulated by 14-3-3 proteins,Mol. Cell. Biol. 22 (2002) 916–926.

[46] A. Delprato, D.G. Lambright, Structural basis for Rab GTPase activation by VPS9domain exchange factors, Nat. Struct. Mol. Biol. 14 (2007) 406–412.

[47] J.L. Bos, H. Rehmann, A. Wittinghofer, GEFs and GAPs: critical elements in thecontrol of small G proteins, Cell 129 (2007) 865–877.

[48] G.J. Doherty, H.T. McMahon, Mechanisms of endocytosis, Annu. Rev. Biochem. 78(2009) 857–902.

[49] J.D. Orth, M.A. McNiven, Get off my back! Rapid receptor internalization throughcircular dorsal ruffles, Cancer Res. 66 (2006) 11094–11096.