Rin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and...

23
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 , and 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 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 identification 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 fluid-phase as well as EGFR endocytosis. Research highlights Rinl is a novel member of the RIN protein family and specifically binds MuSK. Rinl catalyzes the nucleotide exchange on Rab5a and Rab22. Rinl shows a higher catalytic activity for Rab22 than Rab5a. Rinl regulates the localization of Rab5a and Rab22 to actin-positive membrane ruffles. Overexpression of Rinl increases both fluid-phase and receptor-mediated endocytosis. Abbreviations AChR, acetylcholine receptor; BGT, bungarotoxin; EGFR, epidermal growth factor receptor; GEF, guanine nucleotide exchange factor; GFP, green fluorescent protein; GST, glutathione S- transferase; HRP, horseradish peroxidase; MuSK, muscle specific kinase; RT-PCR, reverse transcriptase polymerase chain reaction; RTK, receptor tyrosine kinase; SH2, Src homology 2 © 2011 Elsevier B.V. Corresponding author at: Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria. Tel.: +43 1 4016034350; fax: +43 1 40160934093. [email protected]. This document was posted here by permission of the publisher. At the time of deposit, it included all changes made during peer review, copyediting, and publishing. The U.S. National Library of Medicine is responsible for all links within the document and for incorporating any publisher-supplied amendments or retractions issued subsequently. The published journal article, guaranteed to be such by Elsevier, is available for free, on ScienceDirect. Sponsored document from Biochimica et Biophysica Acta Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210. Sponsored Document Sponsored Document Sponsored Document

Transcript of Rin-like, a novel regulator of endocytosis, acts as guanine nucleotide exchange factor for Rab5a and...

Rin-like, a novel regulator of endocytosis, acts as guaninenucleotide exchange factor for Rab5a and Rab22

Barbara Wollera, Susan Luiskandla, Milica Popovicb, Barbara E.M. Prielera, Gloria Ikongea,Michaela Mutzla, Holger Rehmannb, and Ruth Herbsta,⁎

aCenter for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, AustriabDepartment of Physiological Chemistry, UMC Utrecht, 3584 CG Utrecht, The Netherlands

AbstractRIN proteins serve as guanine nucleotide exchange factors for Rab5a. They are characterized bythe presence of a RIN homology domain and a C-terminal Vps9 domain. Currently three familymembers have been described and analyzed. Here we report the identification of a novel RINfamily member, Rin-like (Rinl), that represents a new interaction partner of the receptor tyrosinekinase MuSK, which is an essential key regulator of neuromuscular synapse development. Rinl islocalized to neuromuscular synapses but shows the highest expression in thymus and spleen. Rinlpreferentially 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 implicatingRinl 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 andthus contributes to the spatial control of Rab5a and Rab22 signaling at actin-positivecompartments. Most importantly, overexpression of Rinl affects fluid-phase as well as EGFRendocytosis.

Research highlights► Rinl is a novel member of the RIN protein family and specifically binds MuSK. ► Rinlcatalyzes the nucleotide exchange on Rab5a and Rab22. ► Rinl shows a higher catalytic activityfor Rab22 than Rab5a. ► Rinl regulates the localization of Rab5a and Rab22 to actin-positivemembrane ruffles. ► Overexpression of Rinl increases both fluid-phase and receptor-mediatedendocytosis.

AbbreviationsAChR, acetylcholine receptor; BGT, bungarotoxin; EGFR, epidermal growth factor receptor;GEF, guanine nucleotide exchange factor; GFP, green fluorescent protein; GST, glutathione S-transferase; HRP, horseradish peroxidase; MuSK, muscle specific kinase; RT-PCR, reversetranscriptase polymerase chain reaction; RTK, receptor tyrosine kinase; SH2, Src homology 2

© 2011 Elsevier B.V.⁎Corresponding author at: Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria. Tel.: +43 14016034350; fax: +43 1 40160934093. [email protected] document was posted here by permission of the publisher. At the time of deposit, it included all changes made during peerreview, copyediting, and publishing. The U.S. National Library of Medicine is responsible for all links within the document and forincorporating any publisher-supplied amendments or retractions issued subsequently. The published journal article, guaranteed to besuch by Elsevier, is available for free, on ScienceDirect.

Sponsored document fromBiochimica et Biophysica Acta

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

KeywordsEndocytosis; Rab5; GEF; MuSK; Receptor tyrosine kinase

1 IntroductionCycling of Rab GTPases between the inactive (GDP bound) and active (GTP bound) statedepends on nucleotide exchange of GDP to GTP catalyzed by guanine nucleotide exchangefactors (GEFs) and the stimulation of the intrinsically slow GTP-hydrolysis activity, whichconverts Rab bound GTP to GDP. The GDP to GTP exchange activates Rab proteins byinducing 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 ofVps9 domain-containing GEFs [1]. The founding member of the RIN protein family, RIN1,was identified as an inhibitor of signaling mediated by the GTPase H-Ras. Via its Rasassociation (RA) domain RIN1 competes with binding of the Ras effector Raf1 [2]. Furtherstudies revealed that RIN1 contains a functional Vps9 domain indicative for Rab GEFs. Thisdomain was shown to be necessary for binding to Rab5a and for GDP/GTP exchangeactivity, and indeed RIN1 was characterized as the Ras effector protein that connects Rassignaling to the control of endocytotic processes [3]. The Rab5a GEF activity is shared byall three known RIN proteins as well as the conserved domain structure consisting of an RAdomain, a Vps9 domain, a RIN homology (RH) domain and an N-terminal SH2 domain [1].The Vps9 domain was originally identified in the yeast protein Vps9p, which serves as GEFfor Vps21p and its mammalian homologue Rab5a [4]. Nucleotide exchange by GEFs onRab5a results in increased endocytosis [5].

The consensus Vps9p catalytic domain is present in a variety of multi-domain proteinsincluding the RIN protein family [1]. The crystal structure of the Rabex-5 Vps9 domainshowed that the core Vps9 domain is stabilized by an essential helical bundle N-terminal tothe Vps9 region, in the case of the RIN proteins corresponding to the RH domain [6]. Inaddition, a high specificity 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 isamong the best-studied Rab proteins and is known as the key regulator of endocytoticprocesses like clathrin-coated vesicle formation, fusion between early endosomes,endosomal cargo recruitment and endosomal motility [7]. Rab21 and Rab22 are less wellcharacterized. Both have been localized to early endosomes and implicated in earlyendocytosis [8,9]. However, Rab21 is also thought to be involved in transport from the transGolgi network to endosomes whereas the predominant role of Rab22 has been attributed torecycling processes [10,11].

RIN proteins have been proposed as regulators of early Rab5a-dependent endocytosis due totheir binding properties for Rab5a and their Rab5a-specific GEF activity. RIN1 stimulatesRab5a-dependent endosome fusion and ligand-mediated endocytosis of the EGF-receptor(EGFR), the insulin receptor and the EphA4 receptor [12–14]. In addition, RIN3 has beenshown to interact with amphiphysin II, which is involved in clathrin- and dynamin-dependent endocytosis [15]. These observations are of special interest since increasingevidence supports a close interplay between signaling and endocytosis of receptor tyrosinekinases (RTK). In particular, it was shown that receptors remain active within endosomes[16]. This led to the hypothesis that membrane trafficking could control signal transduction.Studies on different RTKs have now demonstrated that there are two main types of functions

Woller et al. Page 2

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

that receptor trafficking has in regulating receptor signaling: it controls the magnitude of theresponse or it controls the specificity of the response [17].

Here we report the identification of a novel RIN family member, Rin-like (Rinl), which wasisolated as interaction partner of the muscle-specific RTK MuSK. Signal transduction eventsinduced by MuSK are crucially linked to the formation of the neuromuscular synapse(NMS) [18]. MuSK is activated by the motor neuron-derived heparansulfate proteoglycanagrin [19]. Agrin does not bind MuSK directly but interacts with Lrp4, a member of theLDL receptor family [20,21]. Upon binding, the Lrp4/MuSK complex presumablyundergoes a structural rearrangement that results in a dimerization and subsequentautophosphorylation of MuSK. The subsequent activation of the MuSK kinase induces asignaling cascade leading to the formation of the NMS including postsynaptic differentiationcharacterized by the accumulation of acetylcholine receptors (AChRs) at synaptic sites andpresynaptic differentiation as depicted by the development of active zones. Consistent withthis model, agrin, MuSK and lrp4 mutant mice fail to form NMSs, lacking all features ofpre- and postsynaptic specializations [22–24].

Rinl interacts with MuSK independent of its phosphorylation and localizes to NMSs.Further, we show that Rinl acts as GEF for Rab5a. Moreover, Rinl interacts with inactiveRab22 and catalyzes the GDP/GTP exchange on Rab22. Co-localization of Rinl with Rab5aor Rab22 in actin-rich membrane ruffles implicates a novel mechanism for the recruitmentof Rab5a and Rab22 to the cytoskeleton via binding to Rinl. In addition, increased fluid-phase uptake and EGFR endocytosis upon Rinl overexpression implicates Rinl as regulatorof early endocytotic processes.

2 Materials and methods2.1 Antibodies and reagents

The following antibodies were purchased from commercial sources: anti-phosphotyrosinePY99 (Santa Cruz) and PY-100 (Cell Signaling), 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), Alexa 555-conjugated EGFand Alexa 488-conjugated secondary antibodies were obtained from Invitrogen.Horseradish-peroxidase-coupled and fluorophore-conjugated secondary antibodies werepurchased from Jackson ImmunoResearch. TRITC-conjugated phalloidin and cytochalasinD were purchased from Sigma-Aldrich.

2.2 Generation of antibodies against RinlRinl (aa 243–307, Mus musculus) was amplified by PCR (5′-TATGAATTCCACTTTTCCTGTCCT-3′ and 5′-TATAAGCTTTGGGCTGTAGGGATTT-3′) and cloned into modified pMALc andpGEX-4T-3 vectors, which encode for seven C-terminal histidines in addition to MBP orGST respectively [26]. Recombinant proteins were expressed in the Escherichia coli strainXL1 blue in LB medium supplemented with 0.4% glucose by induction with 0.2 mM IPTGand purified via HIS-affinity chromatography. The MBP fusion protein was used to raisepolyclonal antisera in rabbits as described previously [26]. Reactive serum was affinitypurified on an Affi-Gel 10 (Bio-Rad) column coupled with the GST fusion proteinmentioned above. The column was sequentially washed with phosphate buffered saline(PBS), 0.1 M NaHCO3 and 0.1 M NaHCO3/0.5 M NaCl. The bound antibodies were elutedwith 7 ml acidic elution buffer (0.1 M glycine, pH 2.5) and fractions of 1 ml were collected.Purified antibodies were tested for immunoblotting and immunoprecipitation as shown inFig. S4.

Woller et al. Page 3

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

2.3 PlasmidsThe MuSK cytoplasmic domain carrying the mutations Y553F or Y750, 754, 755F (KD)was subcloned into the yeast bait vector pBTM116. It was previously demonstrated thatMuSK Y750, Y754, 755F, which affects the activation loop of the kinase domain, behavesas kinase-deficient mutant similar to a K608A mutant, which blocks ATP binding within theautocatalytic loop [27,28]. The MMT bait construct contains the TrkA cytoplasmic domainincluding the MuSK juxtamembrane region in pBTM116 [27]. To generate BTM/tpr-met-JM, the juxtamembrane region of MuSK (Rattus norvegicus, aa 528–570) was subclonedinto BTM/tpr-met using the primers 5′-AATGAATTCAGAGAGTCGGCAGC-3′ and 5′-TAGTCGACTACGGATACTCCAGGCTGAG-3′. Kinase-dead (K608A) and kinase-activeMuSK (LS745,746MT) carrying a myc-tag were provided by Dr. Burden (NYU School ofMedicine, New York, [20]).

The Rinl 5′-end was amplified from mouse muscle cDNA using the following primers: 5′-CTGAGCAGCCTTGAGTCTTGTCTT-3′ (located upstream of the predicted start codon)and 5′-CCTGGCCAGAGCACGGATGT-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 weresubcloned into pcDNA3. For yeast two-hybrid and GST-pulldown experiments Rinl andtruncation mutants were cloned into pACT2 (Clontech) and pGEX (GE Healthcare) vectorsrespectively: GST-Rinl (aa 1–563 in pGEX-5X-2), GST-Y2H (aa 200–563 in pGEX-3),GST-RH/ΔVps9 (aa 200–479 in pGEX-2), GST-RH/Vps9 (aa 248–563 in pGEX-2).

pLexA-Rab5a (wt, S34N, Q79L) plasmids were a gift from Dr. Zerial (Max Planck Instituteof Molecular Cell Biology and Genetics, Dresden), for GFP-tagged constructs Rab5a wt andmutants were cloned into pEGFP-C2 (Clontech) using EcoRI. pEGFP-Rab22 (wt, S19N,Q64L) was provided by Dr. Donaldson (NIH, Bethesda). Rab22 inserts were amplified byPCR (Rab22 MunI fw 5′-AAACAATTGATGGCGCTGAGGGAGCTCAAA-3′ and Rab22SalI rev 5′-AAAGTCGACGCAGCAGCTTCGCTGCGGCTC-3′) and cloned into pLexAlinearized with EcoRI and SalI. GFP-Rab7a (wt, T22N, Q67L) constructs were a gift fromDr. Bucci (University of Salento, Lecce).

All constructs used in this study were checked by sequencing.

2.4 Cell cultureHEK 293T cells, COS7 and HeLa cells were maintained in DMEM supplemented withglutamine, 4.5 mg/ml glucose, 10% fetal bovine serum (FBS) and 100 μg/ml penicillin/streptomycin. HEK 293T cells were transfected using the protocol described by Chen andOkayama [29]. COS7 and HeLa cells were transfected using TurboFect (Fermentas).Briefly, COS7 and HeLa cells were plated on glass coverslips and transfected the next daywith 1 μg DNA and 1.5 μl TurboFect in 2 ml DMEM.

2.5 Yeast two-hybrid screenThe MuSK cytoplasmic domain (Rattus norvegicus, aa 517–868) was subcloned into themodified pBTM116 vector, BTM/tpr-met (a gift from Dr. Birchmeier, Max-DelbrueckCenter for Molecular Medicine, Berlin), which contains an active c-Met kinase lacking themultiple docking site [30]. Insertion of MuSK produces a fusion between c-Met and MuSK.The MuSK bait was screened against a mouse muscle cDNA library (a gift from Dr.Chamberlain, University of Michigan Medical School, Ann Arbor) cloned into the pACT2prey vector [31]. The BTM/tpr-met-MuSK bait plasmid was introduced into the L40 yeaststrain. Large scale transformations with the prey library were performed according to Vojtekand Hollenberg [32]. A total of 7.2 × 106 transformants were screened for growth on –HIS.

Woller et al. Page 4

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

Forty-six positive clones were recovered. After retransformation of the recovered cDNAplasmids six clones grew on –HIS and produced a positive signal in a β-gal assay withMuSK but were negative for control plasmids (BTM/tpr-met, BTM/cMet and BTM/lamin).To analyze the interaction between MuSK and Rinl or Rinl and Rab5a/Rab22 respectively,the bait and prey plasmids were transformed into the yeast strain L40. Single colonies wererestreaked on –LEU/TRP or –LEU/TRP/HIS. A β-gal assay screening for lacZ expressionwas performed as described previously [32].

2.6 GST-pulldown experimentsAll GST fusion proteins were expressed in the bacterial strain Rosetta by induction with0.5 mM IPTG. The harvested bacteria were resuspended in ice-cold buffer R (PBS, 1%Triton X-100, 1 mg/ml lysozyme, 0.2 mM phenylmethylsulfonyl fluoride (PMSF)). Cellswere lysed on ice for 1 hour, 0.25% sarkosyl was added and samples were sonicated.Cleared lysates were incubated with glutathione agarose beads (Sigma) for 1 hour underconstant rotation at 4 °C, followed by three washing steps with buffer R (without lysozyme).Protein concentration and quality were assayed by SDS-PAGE and subsequent Coomassiestaining. Recombinant proteins were stored at − 80 °C until use. Cell lysates were incubatedwith immobilized GST fusion proteins for 4–18 hours at 4 °C. Beads were washed threetimes with NP-40 lysis buffer (1% Nonidet P-40, 5 mM EGTA, 50 mM NaCl, 30 mMtriethanolamine [pH 7.5], 50 mM NaF). Proteins bound to the beads were subjected to SDS-PAGE and immunoblotting.

2.7 Lysate preparation, immunoprecipitation and immunoblottingLysates were prepared from cultured cells or adult mouse tissues using NP-40 lysis buffersupplemented with fresh proteinase inhibitors (1 μg/ml leupeptin, 1 μg/ml pepstatin, 1 μg/mlaprotinin, 0.2 mM PMSF, 1 mM sodium orthovanadate). Protein concentrations weredetermined using Roti-Nanoquant (Roth) protein assay reagent.

For co-immunoprecipitation experiments, HEK 293T cells were co-transfected withindicated constructs. Cleared lysates were precipitated with polyclonal affinity-purifiedantibodies directed to either MuSK or Rinl overnight. The next day protein A agarose(Roche) was added for 1–3 hours, the beads were washed three times with NP-40 lysisbuffer with increased (150 mM) NaCl concentration and precipitated protein complexeswere analyzed by SDS-PAGE and immunoblotting.

2.8 RT-PCR and quantitative PCRRNA was isolated from C57BL/6 mice using TRI reagent (Sigma) according to themanufacturer's protocol and reverse transcribed to cDNA. The following primers were usedfor subsequent PCR reactions: Rinl-1154fw (5′-GAAGATCTTGGCCCCGCTGT-3′),Rinl-1645rev (5′-CTGGTAGTGAGCAATGTGGT-3′), actin1 (5′-TTCTACAATGAGCTGCGTGTGG-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 a Bio-Rad iCycler. Rinl levels were normalized to Gapdh using the formula:Rinl/Gapdh = 2(CTx − CTrel) / 2(CTx − CTrel). Spleen was used as reference tissue (rel)with a value set to 1. PCR reactions were performed in duplicates or triplicates.

2.9 ImmunofluorescenceEighteen 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

Woller et al. Page 5

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

5 minutes at RT and incubated with blocking solution (PBS containing 10% FBS) followedby incubation with appropriate primary antibodies for 1 hour at RT. Cells were washed withPBS and incubated with fluorophore-conjugated secondary antibodies with or withoutTRITC-labeled phalloidin. Cells were mounted in Mowiol and visualized using a Leica TCSSP5 spectral confocal microscope with a HCX PL APO CS 63×/1.4 oil objective. For thedisruption of the actin cytoskeleton, transfected cells were treated with 1 μM cytochalasin Dor the solvent DMSO in DMEM for 30 minutes before the cells were fixed and stained withantibodies. The distribution of Rab5a or Rab22 in Rinl positive cells was categorized intofour different classes and plotted as the average ± s.e.m. of three independent experiments(sum of all subcategories is 100%).

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

2.10 EGF uptakeTwenty to 24 hours post transfection cells were starved for 3 hours in DMEM, labeled withAlexa 555-conjugated EGF for 30 minutes at 4 °C and washed four times with ice-coldDMEM supplemented with 1 mg/ml BSA. Then the cells were put back at 37 °C forendocytosis to occur. At indicated time points cells were fixed with 1% PFA in PBS andembedded in Mowiol. Cells were analyzed by confocal microscopy as described above.Representative images are shown.

2.11 HRP uptakeHEK 293T cells were transfected with the following constructs: pEGFP, pEGFP-Rab5a wt,pEGFP-Rinl as stated. About 20 hours after transfection cells were sorted for GFPexpression by FACS. GFP-positive cells were seeded on polyornithine coated 12-well platesovernight. The next day HRP uptake was analyzed as previously described [34]. Briefly,cells were incubated in internalization medium (IM; DMEM, 1% FBS, 24 mM HEPES [pH7.5]) containing 4 mg/ml HRP for 1 hour at 37 °C. After internalization, cells were washedonce with warm IM followed by three washes with ice-cold PBS supplemented with 1 mMCaCl2, 1 mM MgCl2, 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 mM HEPES [pH7.5], 0.2 mM PMSF). Total HRP activity of the lysates was determined in duplicates oftriplicate samples using the substrate 3,3′,5,5′-tetramethylbenzidine (Sigma). HRP activitywas normalized to the protein concentration of individual lysates.

2.12 GEF activity assayRab5a (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 inthe bacterial strain CK600K in Standard I medium (Merck) by induction with 100 μM IPTGat an OD600 of 0.8 at 25 °C overnight. Bacteria were harvested by centrifugation and washedwith 0.9% NaCl. Rab proteins were purified, cleaved from GST-tag and loaded with mGDP(2′-/3′-O-(N′-Methylanthraniloyl)guanosine-5′-O-diphosphate) essentially as described forRap [35]. Rinl was purified the same way except that the buffer was MgCl2-free andcontained 5 mM EDTA, and the GST-tag was not cleaved. Fluorescence measurements wereperformed as described [35]. In brief, 200 nM of the G-protein loaded with mGDP wereincubated in buffer G (50 mM Tris–HCl, pH 7.5, 50 mM NaCl, 5 mM MgCl2, 5 mM DTT,5% glycerol) in the presence of 20 μM GDP and increasing amounts of Rinl. For kineticanalysis the obtained curves were fitted as single exponential decay to obtain the rateconstant kobs. kobs were plotted against the concentration of Rinl and analyzed by linearfitting, whereby the slope was defined as the catalytic efficiency of the GEF reaction.

Woller et al. Page 6

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

3 Results3.1 Rinl: a novel interaction partner of MuSK

To identify proteins that bind to MuSK, a mouse muscle cDNA library was screened withthe MuSK cytoplasmic domain using the yeast two-hybrid system. Screening of 7.2 × 106

transformants yielded six positive clones that interacted strongly with MuSK. One of theseclones, termed CL-6, was identified independently three times. It consisted of a 2204 bpcDNA encoding 363 amino acids, an in-frame stop codon, the 3′ UTR and the poly(A) tail.The isolated cDNA was identical to ENSMUST00000059857 lacking however the first 199amino acids. We therefore isolated the missing 5′ sequence by RT-PCR. Analysis of thetotal cDNA revealed an open reading frame that encoded 563 amino acids in which theoriginal CL-6 sequence corresponded to amino acids 200–563 (Fig. S1A).

Homology search revealed a sequence similarity and structural homology between the novelMuSK interacting protein and the family of 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 an SH2 domain, an RH domainand a Vps9 domain. In contrast, the RA domain and proline-rich regions found in RIN1, 2and 3 are not present in Rinl.

To determine the region(s) within Rinl necessary for interaction with MuSK, we generatedseveral Rinl deletion mutants fused to the Gal4 activation domain (Fig. 1A). Using thereporter genes HIS and lacZ, interactions with the full-length MuSK cytoplasmic domainwere assayed in yeast. As shown in Fig. 1B, deletion mutants lacking either the SH2 domain(Y2H and RH/Vps9) or the Vps9 domain (RH/ΔVps9) were able to interact with MuSK. Incontrast, no interaction was detected with a deletion construct that encodes the Vps9 domainonly (Vps9). We confirmed these findings by GST pulldowns of MuSK expressed in HEK293T cells using the same Rinl deletion constructs fused to GST (Fig. 1C). These resultssuggest that the Rinl/MuSK interaction depends on the RH domain of Rinl.

The MuSK cytoplasmic domain consists of a short juxtamembrane region, a tyrosine kinasedomain and an eight amino acid C-terminal tail. Upon agrin stimulation MuSK becomestyrosine-phosphorylated, which induces recruitment of downstream factors andconsequently downstream signaling [25,27,36]. To test whether the interaction betweenMuSK and Rinl is phosphorylation dependent, we expressed MuSK wild-type, MuSKkinase-active or MuSK kinase-dead together with Rinl in HEK 293T cells. Co-immunoprecipitation of Rinl with MuSK was detected independent of MuSKphosphorylation (Fig. 1D and Fig. S2). To narrow down the site of interaction within MuSKwe generated MuSK deletion and point mutants fused to the lexA binding domain. Bindingof Rinl to these MuSK mutant proteins was assayed in yeast using the reporter genes HISand lacZ (Fig. 1E). Mutation of tyrosines in the autoactivation loop of the kinase domain(MuSK KD) do not affect Rinl/MuSK interaction. Similarly, the juxtamembrane tyrosineY553 is dispensable for Rinl binding to MuSK. We did not include any truncations into thekinase domain since deletions of C-terminal parts will destroy the correct folding of theprotein and consequently negatively influence our binding studies. We used howeverconstructs carrying only the MuSK juxtamembrane region (JM) or the MuSKjuxtamembrane NPXY motif inserted in the TrkA cytoplasmic domain (MMT). Thesefusion proteins are unable to interact with Rinl. Taken together, we conclude that Rinl bindsto the MuSK kinase domain and that this binding is independent of MuSK phosphorylation.

Woller et al. Page 7

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

3.2 Rinl is ubiquitously expressed with highest expression in lymphoid organs andspecifically accumulated at NMSs

We performed RT-PCR using RNA isolated from adult mouse tissue. Rinl is expressed in alltested tissues with the highest expression in lung, spleen and thymus (Fig. 2A). These datawere confirmed by qPCR showing that Rinl is up to 40 times more expressed in thymus thanmuscle (Fig. 2B). We also studied the expression of Rinl during development and found aslight increase in expression in spleen and thymus within the first weeks after birth but noprofound developmental regulation of Rinl gene expression in brain and muscle (Fig. S3Aand B). To further extend these expression studies we investigated Rinl protein expression inthymus, spleen and muscle. Analysis by immunoblotting revealed a similar expressionprofile as detected by RT-PCR. High expression was found in thymus and spleen comparedto a weak expression in muscle (Fig. S3C).

Proteins important for NMS development are usually enriched at synaptic sites. Wetherefore tested whether Rinl is localized at NMSs. We stained muscle sections withantibodies against Rinl and Alexa-594 conjugated α-BGT (Fig. 2C and S4). As shown inFig. 2C, a weak but specific enrichment of Rinl at NMSs is detectable.

3.3 Rinl acts as GEF for Rab5aThe family of RIN proteins is characterized by its Vps9 domain and its binding affinity forRab5a. All so far known RIN proteins are able to catalyze the GDP to GTP exchange onRab5a [3,15,37]. First experiments were performed using the yeast two-hybrid system todetermine whether Rinl binds to Rab5a. The dominant-negative form Rab5a S34N showed astrong interaction with Rinl. In contrast, Rinl did not interact with Rab5a wild-type and theconstitutively-active Rab5a Q79L variant (Fig. 3A). Dominant-negative mutations areknown to reduce the nucleotide affinity of the G-protein, resulting in increasedconcentrations of nucleotide free G-protein which binds with high affinity to its GEFs. Theobserved interaction profile thus suggests that Rinl might be a Rab5a GEF. To map the siteof interaction we used the Rinl mutant constructs described in Fig. 1A and found that the N-terminal region is dispensable for Rinl/Rab5a interaction. However, deletion of the Vps9domain or the RH domain abolished the binding of Rinl to Rab5a S34N suggesting that boththe RH and Vps9 domain are required for interaction (Fig. 3A and B). These findings wereconfirmed by GST pulldowns of Rab5a expressed in COS7 cells using the same Rinldeletion constructs fused to GST (Fig. 3C). We further tested the interaction between Rinland Rab5a by co-immunoprecipitation from HEK 293T cells transfected with Rinl andRab5a wild-type, Rab5a S34N or Rab5a Q79L, respectively. Rab5a S34N efficiently co-immunoprecipitates with Rinl, whereas Rab5a wild-type and Rab5a Q79L show a weak orno interaction (Fig. 3D). In contrast, 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 to nucleotide free Rab5a,we next asked whether Rinl indeed acts as GEF for Rab5a. Recombinant proteins wereexpressed in bacteria and used in an in vitro assay to measure nucleotide exchange. In brief,the Rab GTPase was loaded with the fluorescent nucleotide analog mGDP. The fluorescenceintensity of mGDP is approximately twice as high if bound to the hydrophobic environmentof a protein as if exposed freely to the buffer solution. The exchange of mGDP in thepresence of excess unlabeled GDP can thus be measured in real time as fluorescence signaldecay. As shown in Fig. 3E, the addition of Rinl accelerates nucleotide exchange in aconcentration dependent manner by several orders of magnitude. In contrast, no exchangeactivity of Rinl for Rab7a could be detected. To validate proper nucleotide loading ofRab7a, the Mg2+ chelator EDTA was added to the control reaction at the indicated timepoint. This induces the release of nucleotides from G-proteins as their binding is Mg2+

Woller et al. Page 8

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

dependent. These data demonstrate that Rinl similar to the other members of the RIN proteinfamily acts as GEF for Rab5a.

3.4 Rinl colocalizes with dominant-negative Rab5a to cytoskeletal-rich membrane rufflesTo examine the subcellular distribution of Rinl COS7 cells were transfected with myc-tagged Rinl (Fig. 4A). We found Rinl localized to a variety of different compartments. Toanalyze the distribution in more detail we divided Rinl localization into four categories andquantified the distribution within these categories. As shown in Fig. 4A, around 90% of Rinlis localized either diffusely and/or in vesicles. The remaining 10% of Rinl show a ruffle-likedistribution. This localization pattern is the same for GFP-tagged Rinl or untagged Rinl (Fig.S5). Rinl shows no co-localization with early endosomes, recycling and late endosomes orthe Golgi (Fig. S6). Interestingly, upon co-expression of Rinl and Rab5a S34N, Rinl andRab5a are co-localized predominantly to membrane ruffles (Fig. 4B and S7A). Similarly, aweak co-localization between Rinl and Rab5a wt is detectable at membrane-ruffles. This co-localization is increased when Rinl lacking the SH2 domain is co-expressed with Rab5a wt(Fig. S7B). Most importantly, Rab5a S34N becomes redistributed from a diffusecytoplasmic localization to either ruffle-like or vesicular-like structures. Membrane ruffleshave been implicated in endocytotic processes associated with cytoskeletal rearrangements.To test whether actin is concentrated within the Rinl/Rab5a-positive ruffles we stained cellswith phalloidin. Fig. 4C shows a co-localization of actin with Rinl and Rab5a. Moreover,disruption of the cytoskeleton using cytochalasin D blocked the formation of membraneruffles and caused an accumulation of Rinl and Rab5a in actin-positive aggregates.

3.5 Rinl acts as GEF for Rab22 and co-localizes with Rab22 to actin-rich domainsRab22 is the closest homologue of Rab5 with 52% sequence identity [38]. Like Rab5a,Rab22 has been localized to early endosomes but its role during endocytosis is so far notwell understood. Since Vps9 domain containing proteins have been reported to activateRab5a and its homologues Rab21 and Rab22, we tested the interaction of Rinl with Rab22[6]. Using the yeast two-hybrid system we detect a strong interaction between Rinl and thedominant-negative form of Rab22. Moreover, the binding of Rinl to Rab22 is dependent onthe RH and Vps9 domain but independent of the N-terminal region (Fig. 5A and B). TheRinl/Rab22 interaction was confirmed by co-immunoprecipitation experiments intransfected HEK 293T cells. Dominant-negative Rab22 S19N efficiently co-immunoprecipitates with Rinl upon co-expression, whereas Rab22 wild-type andconstitutively active Rab22 Q64L show a weak or no interaction (Fig. 5C). To determinewhether Rinl acts indeed as GEF for Rab22, the GEF activity of Rinl toward Rab22 wasmeasured in vitro (Fig. 5D). Rinl strongly accelerates nucleotide exchange of Rab22 and cantherefore be classified as a GEF of Rab22. For a more quantitative comparison of the Rinlactivity toward Rab5a and Rab22, the rates of the nucleotide exchange reaction, kobs, weredetermined from measurements as presented in Figs. 3E and 5D. The dependency of kobs onthe Rinl concentration is presented in Fig. 5E. As can be seen, nucleotide exchange towardRab22 is more sensitive to the concentration of Rinl, which thus displays a higher catalyticrate for Rab22 than for Rab5a. Moreover, Rinl and Rab22 S19N co-localize to similarcompartments upon co-expression in COS7 cells (Fig. S7). Like for Rab5a, Rinl onlyweakly co-localizes with Rab22 wt whereas Rinl lacking the SH2 domain robustly co-localizes with Rab22 wt (Fig. S7B). Rinl induces a redistribution of diffusely localizedRab22 to ruffle-like structures (Fig. 6A). Similar to Rab5a and Rinl, also Rab22 and Rinl co-localize to actin-positive structures (Fig. 6B).

3.6 Rinl recruits Rab5a and Rab22 to the cytoskeleton via the Vps9 domainSince we detected a significant redistribution of dominant-negative Rab5a and Rab22 uponco-expression with Rinl, we next asked how this localization is influenced by Rinl deletion

Woller et al. Page 9

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

mutants. For that we used constructs that either lack the N-terminal SH2 domain or the C-terminal Vps9 domain. These truncations were co-expressed with Rab5a S34N or Rab22S19N and the localization assayed by immunostaining. Rinl lacking the SH2 domain(ΔSH2) localizes to vesicles and ruffles together with Rab5a and Rab22 (Fig. 7). Inparticular, Rinl ΔSH2 has a more pronounced localization to ruffles compared to full-lengthRinl (data not shown). Moreover, Rinl ΔSH2 shows a higher degree of co-localization withRab5a or Rab22 to ruffles than full-length Rinl and Rab5a/Rab22 (compare Figs. 4B and 6Ato 7B). This suggests that the SH2 domain might have an inhibitory effect on the interactionbetween Rinl and Rab proteins. In contrast, Rinl lacking the Vps9 domain (ΔVps9) does notco-localize with Rab5a S34N and Rab22 S19N (Fig. 7). Rab5a and Rab22 are unchangedupon co-expression with Rinl ΔVps9 and remain diffusely distributed and/or in vesicles.Rinl ΔVps9 itself is localized mainly throughout the cytoplasm or in vesicles but does notattach to actin-rich membrane ruffles. Therefore, the Vps9 domain is not only responsiblefor Rab5a/Rab22 activation but also mediates the localization of Rinl and Rab proteins tothe cytoskeleton.

3.7 Rinl regulates fluid-phase and receptor-mediated endocytosisSince RIN family members have previously been implicated in endocytotic processes we setout to study the function of Rinl during fluid-phase endocytosis. For that we examined theHRP uptake in HEK 293T cells expressing Rinl, Rab5a wt, Rab5a wt together with Rinl, orGFP as a control. Expression of Rinl and Rab5a induced the internalization of HRP(Fig. 8A). The co-expression of Rab5a wt and Rinl induced a similar degree of HRP uptakethan expression of Rinl or Rab5a wt alone.

It has previously been demonstrated that RIN1 acts as regulator of EGFR endocytosis[12,39]. We therefore asked whether Rinl also affects EGFR internalization. We used Alexa555-conjugated EGF to label endogenous EGFR in HeLa cells expressing Rinl, Rab5a wt,Rab5a S34N, Rinl together with Rab5a wt, or GFP as a control. EGF uptake at 37 °C wasimaged at various times. As shown in Figs. 8B and S8, in control cells EGFR endocytosis israpidly induced within 5 minutes and perinuclear accumulations of EGF-positive vesicles isprominent by 15 minutes. Expression of Rab5a wt or Rinl leads to an acceleration of EGFRendocytosis and a decrease in EGF-positive vesicles. This increase in EGFR endocytosis isespecially pronounced in cells co-expressing Rab5a wt and Rinl. In contrast, cellsexpressing Rab5a S34N show a reduced EGFR endocytosis. These results support afunctional role of Rinl during early endocytotic processes.

4 DiscussionThe GTPase Rab5a is the key regulator during early endocytotic processes. Therefore, it isof particular interest to identify mechanisms and molecules that modulate Rab5a action. Inthis study we report the identification and characterization of Rinl, a novel Rab5a GEF,which shows a high homology to the family of RIN proteins. Rinl was isolated via itsinteraction with the RTK MuSK. It specifically interacts with MuSK through the centralportion of the protein containing the RH domain. Rinl interacts and co-localizes with Rab5aand catalyzes GDP/GTP exchange on Rab5a. Similar biochemical and enzymatic propertieswere demonstrated toward Rab22. Furthermore, we identified the Vps9 domain of Rinl as acritical molecular determinant that controls the recruitment of Rab5a and Rab22 tocytoskeletal membrane compartments. Most importantly, Rinl stimulates fluid-phase uptakeand EGFR endocytosis.

The formation of the NMS is crucially linked to signal transduction events induced by themuscle-specific RTK MuSK [18]. MuSK activation via agrin/Lrp4 induces a signalingcascade that leads to post- as well as presynaptic differentiation. Several MuSK binding

Woller et al. Page 10

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

proteins have been identified which include adaptor proteins like Dok7, kinases like Abl andscaffolding proteins including Magi-1c and ColQ [18]. More recently the trafficking proteinNSF and the E3 ubiquitin ligases PDZRN3 and PAUL were isolated as MuSK interactionpartner, which are thought to regulate MuSK endocytosis and degradation, respectively [40–42]. Here we identified Rinl as novel interaction partner of MuSK. Binding of Rinl to MuSKrequires the internal RH domain of Rinl but is independent of MuSK phosphorylation. Thissuggests that Rinl binding is not associated with MuSK activation. However, a regulation ofRinl/MuSK interaction dependent on the subcellular localization appears likely since weshow a specific localization of Rinl to vesicles and membrane ruffles. In support of thishypothesis we detect a specific co-localization of MuSK and Rinl at actin-positivemembrane ruffles (data not shown). The identification of Rinl as binding partner of MuSKappeared of distinct interest since it was recently demonstrated that MuSK endocytosisregulates MuSK signaling [42]. However so far, we have not been able to correlate Rinlaction and MuSK function. Rinl is very weakly expressed in muscle cells questioning theimportance of Rinl in cultured muscle cells. Nevertheless, this does not exclude thepossibility that Rinl plays a role during MuSK-dependent signal transduction in vivo, eitherat the NMS or in the brain, where MuSK function during memory formation has beenimplicated recently [43]. A gene targeting approach in mice will be necessary to answerthese questions. Furthermore, a functional compensation by one of the other RIN familymembers cannot be ruled out at the moment.

The RIN family members have been implicated in early endocytotic events [1]. In particular,the role of RIN1 during receptor endocytosis has been demonstrated many-fold [12–14,39].Here we are able to show that Rinl overexpression accelerates fluid-phase endocytosis aswell as EGFR endocytosis. These data support our in vitro data and provide first hints on thephysiological role of Rinl. RIN1 has been postulated as crucial regulator of EGFRendocytosis and signaling [12,39,44]. Our findings raise the questions whether Rinlrepresents a similarly important regulator of EGFR endocytosis and whether Rinl and RIN1play complementary roles during EGFR endocytosis due to their differential expressionpatterns.

The current members of the RIN protein family share an SH2 domain, an RH region, a Vps9domain and an RA domain. RIN1 was isolated by its ability to bind to H-Ras via the RAdomain thereby competing with Raf1 and inhibiting Ras action [2,45]. In addition, the RAdomain has been implicated in the interaction with the EGFR via Ras, which leads to arecruitment of RIN1 to the activated receptor linking internalized EGFR to Rab5a-positiveendosomes [3]. Rinl lacks an RA domain. Therefore it appears unlikely that Rinl acts as aneffector for Ras proteins. However, a recruitment to activated RTKs can still occur throughthe N-terminal SH2 domain, which would then bring the intracellular receptors to earlyendosomes. Such an SH2-dependent interaction has been shown for RIN1 and EGFR as wellas RIN1 and EphA4 [12,13].

The RIN proteins belong to the family of Vps9 domain containing proteins. These proteinsare characterized by their ability to bind Rab5a and to catalyze the GDP/GTP exchange onRab5a [1]. Crystallization studies on Rabex-5 have shown that an N-terminal helical bundlein addition to the Vps9 domain is required for GEF activity, the so-called HB-Vps9 tandem[6]. The helical bundle conforms to the RH domain in the RIN proteins. Consistent with theRabex-5 data, it was shown that a splice-variant of RIN1 lacking part of the helical bundle isunable to interact with dominant-negative Rab5a. Likewise, Rinl truncations deleting the RHdomain/helical bundle do not bind dominant-negative Rab5a implicating similarbiochemical properties for all Vps9 domain containing proteins. It was also reported thatfull-length Vps9 domain containing proteins including the RIN proteins have a reducedbinding activity and/or GEF activity for Rab5a [6,46]. This suggests that autoinhibition by

Woller et al. Page 11

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

regulatory elements in the N- and/or C-terminus play a role. Similarly, we find a reducedbinding activity for Rab5a and Rab22 in full-length Rinl constructs compared to Rinlconstructs carrying only the RH and Vps9 domain. Therefore, the N-terminal SH2 domainmight represent such an inhibitory element. Protein interactions via the SH2 domain wouldthen relief the autoinhibition and induce exchange activity. So far however, it remainsunclear which proteins bind to the SH2 domain of Rinl.

Analysis of the Rabex-5 HB-Vps9 tandem revealed a high specificity toward the Rab5subfamily but also showed a selective exchange activity for the different Rab5 subfamilymembers: strong GEF activity for Rab5 and Rab21, a weak activity for Rab22 [6]. Thisspecificity and selectivity is achieved on one hand by conserved exchange determinants on acommon surface of the Vps9 domain and on the other hand by invariant aromatic residues inthe switch regions of the Rab GTPases. Delprato and colleagues also reported a similarcatalytic activity and Rab specificity for RIN1 [6]. In this study we show that Rinlspecifically binds Rab5a and Rab22, and there preferentially the nucleotide free forms. Rinlacts as GEF for Rab22 and Rab5a, surprisingly however, presents a higher efficiency forRab22. This distinguishes Rinl from RIN1 and suggests that different Vps9 domains havedifferent specificity profiles for the Rab5 subfamily. This of course also raises the questionwhether this specificity is also represented at a physiological level whereby different RINproteins regulate different Rab5 subfamily dependent processes. Four amino acids (D313,P317, Y354 and T357) in the Vps9 domain in Rabex-5 have been shown to be essential forexchange activity and these residues are highly conserved among different Vps9 domains[6]. Likewise, the Rinl Vps9 domain contains these conserved amino acids (D456, P460,Y497 and T500). Other residues, which have been shown to lie within the binding site forthe GTPase differ between Rabex-5 and Rinl. These residues are expected to be responsiblefor the differences in the specificity profile.

When studying the subcellular distribution of Rinl protein we noticed a characteristiclocalization in vesicles and membrane ruffles. Moreover, co-expression of Rinl and Rab5aor Rab22 mutants with lowered nucleotide affinity leads to a redistribution of diffuselyexpressed Rab5a and Rab22 to actin-positive membrane compartments. The localization ofRinl to membrane ruffles as well as the recruitment of Rab5a and Rab22 to the cytoskeletonare dependent on the Vps9 domain. It is unclear at the moment whether the Vps9 domaininteracts directly or indirectly (via a so-far unknown actin binding protein) with thecytoskeleton. Rab mutants with lowered nucleotide affinity are known to bind GEFs withhigh affinity, thereby trapping the active GEF in an unproductive complex [47]. The abilityof Rinl to recruit Rab5a S34N and Rab22 S19N indicates that Rinl interacts with the actincytoskeleton in an active state. Thus it is expected that Rinl activates Rab proteins locally atthe cytoskeleton. Actin cytoskeleton remodeling has been implicated in early endocytosis aswell as recycling [48]. Furthermore, membrane ruffling is a characteristic of actinremodeling and is closely associated with regions of active endocytosis [49]. Our findingstherefore support a model whereby Rinl-dependent activation of Rab5a and Rab22 at thecytoskeleton regulates early endocytotic and/or recycling processes. Future experiments willhave to show how actin remodeling, exchange activity by Rinl and Rab5a/Rab22-dependentendocytosis are connected.

The following are the supplementary materials related to this article.

Supplementary Material1.Supplementary material

Supplementary materials related to this article can be found online atdoi:10.1016/j.bbamcr.2011.03.005.

Woller et al. Page 12

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

References1. Carney D.S. Davies B.A. Horazdovsky B.F. Vps9 domain-containing proteins: activators of Rab5

GTPases from yeast to neurons. Trends Cell Biol.. 2006; 16:27–35. [PubMed: 16330212]2. Han L. Colicelli J. A human protein selected for interference with Ras function interacts directly

with Ras and competes with Raf1. Mol. Cell. Biol.. 1995; 15:1318–1323. [PubMed: 7862125]3. Tall G.G. Barbieri M.A. Stahl P.D. Horazdovsky B.F. Ras-activated endocytosis is mediated by the

Rab5 guanine nucleotide exchange activity of RIN1. Dev. Cell. 2001; 1:73–82. [PubMed:11703925]

4. Hama H. Tall G.G. Horazdovsky B.F. Vps9p is a guanine nucleotide exchange factor involved invesicle-mediated vacuolar protein transport. J. Biol. Chem.. 1999; 274:15284–15291. [PubMed:10329739]

5. Vitale G. Alexandrov K. Ullrich O. Horiuchi H. Giner A. Dobson C. Baykova O. Gournier H.Stenmark H. Zerial M. The GDP/GTP cycle of Rab5 in the regulation of endocytotic membranetraffic. Cold Spring Harb. Symp. Quant. Biol.. 1995; 60:211–220. [PubMed: 8824393]

6. Delprato A. Merithew E. Lambright D.G. Structure, exchange determinants, and family-wide rabspecificity of the tandem helical bundle and Vps9 domains of Rabex-5. Cell. 2004; 118:607–617.[PubMed: 15339665]

7. Zerial M. McBride H. Rab proteins as membrane organizers. Nat. reviews. 2001; 2:107–117.8. Kauppi M. Simonsen A. Bremnes B. Vieira A. Callaghan J. Stenmark H. Olkkonen V.M. The small

GTPase Rab22 interacts with EEA1 and controls endosomal membrane trafficking. J. Cell Sci..2002; 115:899–911. [PubMed: 11870209]

9. Simpson J.C. Griffiths G. Wessling-Resnick M. Fransen J.A. Bennett H. Jones A.T. A role for thesmall GTPase Rab21 in the early endocytic pathway. J. Cell Sci.. 2004; 117:6297–6311. [PubMed:15561770]

10. Weigert R. Yeung A.C. Li J. Donaldson J.G. Rab22a regulates the recycling of membrane proteinsinternalized independently of clathrin. Mol. Biol. Cell. 2004; 15:3758–3770. [PubMed: 15181155]

11. Opdam F.J. Echard A. Croes H.J. van den Hurk J.A. van de Vorstenbosch R.A. Ginsel L.A. GoudB. Fransen J.A. The small GTPase Rab6B, a novel Rab6 subfamily member, is cell-typespecifically expressed and localised to the Golgi apparatus. J. Cell Sci.. 2000; 113(Pt 15):2725–2735. [PubMed: 10893188]

12. Barbieri M.A. Kong C. Chen P.I. Horazdovsky B.F. Stahl P.D. The SRC homology 2 domain ofRin1 mediates its binding to the epidermal growth factor receptor and regulates receptorendocytosis. J. Biol. Chem.. 2003; 278:32027–32036. [PubMed: 12783862]

13. Deininger K. Eder M. Kramer E.R. Zieglgansberger W. Dodt H.U. Dornmair K. Colicelli J. KleinR. The Rab5 guanylate exchange factor Rin1 regulates endocytosis of the EphA4 receptor inmature excitatory neurons. Proc. Natl Acad. Sci. U.S.A.. 2008; 105:12539–12544. [PubMed:18723684]

14. Hunker C.M. Giambini H. Galvis A. Hall J. Kruk I. Veisaga M.L. Barbieri M.A. Rin1 regulatesinsulin receptor signal transduction pathways. Exp. Cell Res.. 2006; 312:1106–1118. [PubMed:16457816]

15. Kajiho H. Saito K. Tsujita K. Kontani K. Araki Y. Kurosu H. Katada T. RIN3: a novel Rab5 GEFinteracting with amphiphysin II involved in the early endocytic pathway. J. Cell Sci.. 2003;116:4159–4168. [PubMed: 12972505]

16. Lai W.H. Cameron P.H. Wada I. Doherty J.J. 2nd, Kay D.G. Posner B.I. Bergeron J.J. Ligand-mediated internalization, recycling, and downregulation of the epidermal growth factor receptor invivo. J. Cell Biol.. 1989; 109:2741–2749. [PubMed: 2592403]

17. Sorkin A. von Zastrow M. Endocytosis and signalling: intertwining molecular networks. Nat.reviews. 2009; 10:609–622.

18. Wu H. Xiong W. Lin M. To build a synapse: signaling pathways in neuromuscular junctionassembly. Dev. Camb. Engl.. 2010; 137:1017–1033.

19. Glass D.J. Bowen D.C. Stitt T.N. Radziejewski C. Bruno J. Ryan T.E. Gies D.R. Shah S. MattssonK. Burden S.J. DiStefano P.S. Valenzuela D.M. DeChiara T.M. Yancopoulos G.D. Agrin acts viaa MuSK receptor complex. Cell. 1996; 85:513–523. [PubMed: 8653787]

Woller et al. Page 13

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

20. Kim N. Stiegler A.L. Cameron T.O. Hallock P.T. Gomez A.M. Huang J.H. Hubbard S.R. DustinM.L. Burden S.J. Lrp4 is a receptor for Agrin and forms a complex with MuSK. Cell. 2008;135:334–342. [PubMed: 18848351]

21. Zhang B. Luo S. Wang Q. Suzuki T. Xiong W.C. Mei L. LRP4 serves as a coreceptor of agrin.Neuron. 2008; 60:285–297. [PubMed: 18957220]

22. DeChiara T.M. Bowen D.C. Valenzuela D.M. Simmons M.V. Poueymirou W.T. Thomas S. KinetzE. Compton D.L. Rojas E. Park J.S. Smith C. DiStefano P.S. Glass D.J. Burden S.J. YancopoulosG.D. The receptor tyrosine kinase MuSK is required for neuromuscular junction formation in vivo.Cell. 1996; 85:501–512. [PubMed: 8653786]

23. Gautam M. Noakes P.G. Moscoso L. Rupp F. Scheller R.H. Merlie J.P. Sanes J.R. Defectiveneuromuscular synaptogenesis in agrin-deficient mutant mice. Cell. 1996; 85:525–535. [PubMed:8653788]

24. Weatherbee S.D. Anderson K.V. Niswander L.A. LDL-receptor-related protein 4 is crucial forformation of the neuromuscular junction. Dev. Camb. Engl.. 2006; 133:4993–5000.

25. Watty A. Neubauer G. Dreger M. Zimmer M. Wilm M. Burden S.J. The in vitro and in vivophosphotyrosine map of activated MuSK. Proc. Natl Acad. Sci. U.S.A.. 2000; 97:4585–4590.[PubMed: 10781064]

26. Mossier B. Togel M. Fuchs K. Sieghart W. Immunoaffinity purification of gamma-aminobutyricacidA (GABAA) receptors containing gamma 1-subunits. Evidence for the presence of a singletype of gamma-subunit in GABAA receptors. J. Biol. Chem.. 1994; 269:25777–25782. [PubMed:7929282]

27. Herbst R. Burden S.J. The juxtamembrane region of MuSK has a critical role in agrin-mediatedsignaling. EMBO J.. 2000; 19:67–77. [PubMed: 10619845]

28. Glass D.J. Apel E.D. Shah S. Bowen D.C. DeChiara T.M. Stitt T.N. Sanes J.R. Yancopoulos G.D.Kinase domain of the muscle-specific receptor tyrosine kinase (MuSK) is sufficient forphosphorylation but not clustering of acetylcholine receptors: required role for the MuSKectodomain? Proc. Natl Acad. Sci. U.S.A.. 1997; 94:8848–8853. [PubMed: 9238066]

29. Chen C. Okayama H. High-efficiency transformation of mammalian cells by plasmid DNA. Mol.Cell. Biol.. 1987; 7:2745–2752. [PubMed: 3670292]

30. Schaeper U. Gehring N.H. Fuchs K.P. Sachs M. Kempkes B. Birchmeier W. Coupling of Gab1 toc-Met, Grb2, and Shp2 mediates biological responses. J. Cell Biol.. 2000; 149:1419–1432.[PubMed: 10871282]

31. Lumeng C. Phelps S. Crawford G.E. Walden P.D. Barald K. Chamberlain J.S. Interactions betweenbeta 2-syntrophin and a family of microtubule-associated serine/threonine kinases. Nat. Neurosci..1999; 2:611–617. [PubMed: 10404183]

32. Vojtek A.B. Hollenberg S.M. Ras–Raf interaction: two-hybrid analysis. Meth. Enzymol.. 1995;255:331–342. [PubMed: 8524119]

33. Nizhynska V. Neumueller R. Herbst R. Phosphoinositide 3-kinase acts through RAC and Cdc42during agrin-induced acetylcholine receptor clustering. Dev. Neurobiol.. 2007; 67:1047–1058.[PubMed: 17565704]

34. Schnatwinkel C. Christoforidis S. Lindsay M.R. Uttenweiler-Joseph S. Wilm M. Parton R.G.Zerial M. The Rab5 effector Rabankyrin-5 regulates and coordinates different endocyticmechanisms. PLoS Biol.. 2004; 2:E261. [PubMed: 15328530]

35. Rehmann H. Characterization of the activation of the Rap-specific exchange factor Epac by cyclicnucleotides. Meth. Enzymol.. 2006; 407:159–173. [PubMed: 16757322]

36. Zhou H. Glass D.J. Yancopoulos G.D. Sanes J.R. Distinct domains of MuSK mediate its abilitiesto induce and to associate with postsynaptic specializations. J. Cell Biol.. 1999; 146:1133–1146.[PubMed: 10477765]

37. Saito K. Murai J. Kajiho H. Kontani K. Kurosu H. Katada T. A novel binding protein composed ofhomophilic tetramer exhibits unique properties for the small GTPase Rab5. J. Biol. Chem.. 2002;277:3412–3418. [PubMed: 11733506]

38. Pereira-Leal J.B. Seabra M.C. Evolution of the Rab family of small GTP-binding proteins. J. Mol.Biol.. 2001; 313:889–901. [PubMed: 11697911]

Woller et al. Page 14

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

39. Kong C. Su X. Chen P.I. Stahl P.D. Rin1 interacts with signal-transducing adaptor molecule(STAM) and mediates epidermal growth factor receptor trafficking and degradation. J. Biol.Chem.. 2007; 282:15294–15301. [PubMed: 17403676]

40. Bromann P.A. Weiner J.A. Apel E.D. Lewis R.M. Sanes J.R. A putative ariadne-like E3 ubiquitinligase (PAUL) that interacts with the muscle-specific kinase (MuSK). Gene Expr. Patterns. 2004;4:77–84. [PubMed: 14678832]

41. Lu Z. Je H.S. Young P. Gross J. Lu B. Feng G. Regulation of synaptic growth and maturation by asynapse-associated E3 ubiquitin ligase at the neuromuscular junction. J. Cell Biol.. 2007;177:1077–1089. [PubMed: 17576800]

42. Zhu D. Yang Z. Luo Z. Luo S. Xiong W.C. Mei L. Muscle-specific receptor tyrosine kinaseendocytosis in acetylcholine receptor clustering in response to agrin. J. Neurosci.. 2008; 28:1688–1696. [PubMed: 18272689]

43. Garcia-Osta A. Tsokas P. Pollonini G. Landau E.M. Blitzer R. Alberini C.M. MuSK expressed inthe brain mediates cholinergic responses, synaptic plasticity, and memory formation. J. Neurosci..2006; 26:7919–7932. [PubMed: 16870737]

44. Barbieri M.A. Fernandez-Pol S. Hunker C. Horazdovsky B.H. Stahl P.D. Role of rab5 in EGFreceptor-mediated signal transduction. Eur. J. Cell Biol.. 2004; 83:305–314. [PubMed: 15511088]

45. Wang Y. Waldron R.T. Dhaka A. Patel A. Riley M.M. Rozengurt E. Colicelli J. The RAS effectorRIN1 directly competes with RAF and is regulated by 14-3-3 proteins. Mol. Cell. Biol.. 2002;22:916–926. [PubMed: 11784866]

46. Delprato A. Lambright D.G. Structural basis for Rab GTPase activation by VPS9 domain exchangefactors. Nat. Struct. Mol. Biol.. 2007; 14:406–412. [PubMed: 17450153]

47. Bos J.L. Rehmann H. Wittinghofer A. GEFs and GAPs: critical elements in the control of small Gproteins. Cell. 2007; 129:865–877. [PubMed: 17540168]

48. Doherty G.J. McMahon H.T. Mechanisms of endocytosis. Annu. Rev. Biochem.. 2009; 78:857–902. [PubMed: 19317650]

49. Orth J.D. McNiven M.A. Get off my back! Rapid receptor internalization through circular dorsalruffles. Cancer Res.. 2006; 66:11094–11096. [PubMed: 17145849]

AcknowledgmentsWe are grateful to Jeffrey Chamberlain for providing us with the mouse muscle prey library. We would like tothank Walter Birchmeier, Cecilia Bucci, Steve Burden, Judy Donaldson, Said Hashemolhosseini and Marino Zerialfor providing plasmids. Many thanks also go to Marije Rensen-De Leeuw for technical assistance. WilfriedEllmeier provided useful comments on the manuscript. This work was supported by the Austrian Science Fund(P19223-B09 to R.H.) and by the Chemical Sciences of the Netherlands Organization for Scientific Research(NOW-CW to H.R.). B.W. was supported by the DOC-fFORTE Programme from the Austrian Academy ofSciences. M.P. was supported by the TI Pharma Project T3-106.

Woller et al. Page 15

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

Fig. 1.Rinl is a specific binding partner of MuSK. (A) A scheme of Rinl and correspondingtruncation mutants. (B) Yeast was transformed with bait and prey constructs as indicated.Interaction was assayed by growth of yeast clones on –HIS. (C) Rinl GST-deletionconstructs were purified from bacteria and used for a pulldown of MuSK-myc expressed inHEK 293T cells. Proteins were detected by immunoblotting (IB) using anti-myc antibodies.GST protein input was assayed by Ponceau staining. 5% of the total MuSK-myc input isshown in the right panel. (D) MuSK-myc and Rinl were expressed in HEK 293T cells andco-immunoprecipitated either with anti-MuSK antibodies or anti-Rinl antibodies.Immunoprecipitated proteins were assayed by immunoblotting using anti-myc and anti-Rinlantibodies, respectively. Phosphorylated MuSK was detected with anti-phosphotyrosine(PY). wt, wild-type; KD, kinase-dead; KA, kinase-active. IP, immunoprecipitation. (E)Yeast was transformed with the indicated MuSK bait constructs and the Rinl (Y2H) preyconstruct. Interaction was followed by growth of yeast clones on –HIS.

Woller et al. Page 16

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

Fig. 2.Rinl expression in different tissues and localization at the NMS. (A) RT-PCR was performedfrom different mouse tissue samples. Actin was used as quality control. (B) qPCR fromcDNAs generated from different tissue RNAs was performed. Spleen was set to 1. s.e.m. isshown. (C) Mouse muscle sections were stained with anti-Rinl antibodies and Alexa 594-conjugated α-BGT. Images were taken on a confocal microscope. Scale bar, 20 μm.

Woller et al. Page 17

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

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 showingthe 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) expressedin 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 transfectedHEK 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 withincreasing amounts of Rinl in the presence of 20 μM unlabeled GDP. The exchange ofmGDP for GDP was measured as decay in fluorescence signal. In case of Rab7a propernucleotide loading was demonstrated by the addition of EDTA at the indicated time point,which induces the release of nucleotides and a rapid decay in the fluorescence signal.

Woller et al. Page 18

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

Fig. 4.Subcellular localization of Rinl and co-localization with Rab5a and actin. (A) myc-taggedRinl was transiently expressed in COS7 cells and stained with antibodies against Rinl andmyc. Representative confocal images are shown. Scale bar, 25 μm. Transfected cells wereassayed for their Rinl expression patterns. Expression patterns were divided into fourcategories. A quantification is shown (n > 100). Error bars, s.e.m. (B) GFP-Rab5a S34Nalone or together with myc-tagged Rinl were co-expressed in COS7 cells. Scale bar, 25 μm.A quantification of the Rab5a S34N distribution was performed as described in A (n = 50).Error bars, s.e.m. (C) Transiently transfected COS7 cells were treated with cytochalasin D orDMSO for 30 minutes. Cells were stained with anti-myc antibodies to detect Rinl and withTRITC-conjugated phalloidin to label the actin cytoskeleton. Representative confocalimages are shown. Scale bar, 25 μm.

Woller et al. Page 19

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

Fig. 5.Rinl preferentially interacts with GDP-bound Rab22 and acts as GEF for Rab22. (A) Yeastwas 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 andRinl 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-Rinlantibodies. Co-immunoprecipitated Rab22 was detected by immunoblotting (IB) with anti-GFP. Rab7a was used as negative control. 10% of the input is shown in the top panel (totallysate). (D) 200 nM of mGDP loaded Rab22 were incubated with increasing amounts ofRinl in the presence of 20 μM unlabeled GDP. The exchange of mGDP for GDP wasmeasured in real time as decay in fluorescence signal. (E) The velocity of the nucleotideexchange reaction (kobs) toward Rab5a and Rab22 were determined and plotted against theconcentration of Rinl.

Woller et al. Page 20

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

Fig. 6.Rab22 and Rinl co-localize to actin-positive membrane ruffles. (A) GFP-Rab22 S19N aloneor together with myc-tagged Rinl were expressed in COS7 cells. Representative confocalimages are shown. Scale bar, 25 μm. Rab22 expression, when transfected alone or togetherwith Rinl, was assayed by confocal microscopy. Subcellular expression patterns werequantified as described in Fig. 5. n > 50; Error bars, s.e.m. (B) Transiently transfected COS7cells were treated with cytochalasin D or DMSO for 30 minutes. Cells were stained withanti-myc antibodies to detect Rinl and with TRITC-conjugated phalloidin to label the actincytoskeleton. Scale bar, 25 μm.

Woller et al. Page 21

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

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 COS7 cells.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 andRab22 was quantified as described in Fig. 5. n > 35; Error bars, s.e.m.

Woller et al. Page 22

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent

Fig. 8.Rinl increases the internalization of HRP and EGFR. (A) HEK 293T cells expressing GFP-Rinl, GFP-Rab5a wt, GFP-Rinl and GFP-Rab5a wt or GFP alone were loaded with HRP.HRP uptake was quantified from two independent experiments done in triplicates. Errorbars, s.e.m. (B) HeLa cells were transfected with GFP, GFP-Rinl, GFP-Rab5a wt, GFP-Rinland GFP-Rab5a wt or GFP-Rab5a S34N. Cells were incubated with Alexa 555-conjugatedEGF at 4 °C and EGF uptake at 37 °C was imaged after 5 and 15 minutes. Representativeconfocal images are shown. Scale bar, 25 μm. The GFP signals of the equivalent cells areshown in Fig. S8.

Woller et al. Page 23

Published as: Biochim Biophys Acta. 2011 June ; 1813(6): 1198–1210.

Sponsored Docum

ent Sponsored D

ocument

Sponsored Docum

ent