Cyclic AMP-dependent and Epac-mediated Activation of R-Ras by G Protein-coupled Receptors Leads to...

11
Cyclic AMP-dependent and Epac-mediated Activation of R-Ras by G Protein-coupled Receptors Leads to Phospholipase D Stimulation * Received for publication, May 1, 2006 Published, JBC Papers in Press, June 5, 2006, DOI 10.1074/jbc.M604156200 Maider Lo ´ pez De Jesu ´s ‡1,2 , Matthias B. Stope ‡1,3 , Paschal A. Oude Weernink , Yvonne Mahlke , Christof Bo ¨ rgermann , Viktoria N. Ananaba , Christian Rimmbach ‡4 , Dieter Rosskopf ‡4 , Martin C. Michel § , Karl H. Jakobs , and Martina Schmidt ‡§5 From the Institut fu ¨r Pharmakologie, Universita ¨tsklinikum Essen, 45122 Essen, Germany and the § Department of Pharmacology and Pharmacotherapy, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands The activation of the Ras-related GTPase R-Ras, which has been implicated in the regulation of various cellular functions, by G protein-coupled receptors (GPCRs) was studied in HEK- 293 cells stably expressing the M 3 muscarinic acetylcholine receptor (mAChR), which can couple to several types of hetero- trimeric G proteins. Activation of the receptor induced a very rapid and transient activation of R-Ras. Studies with inhibitors and activators of various signaling pathways indicated that R-Ras activation by the M 3 mAChR is dependent on cyclic AMP formation but is independent of protein kinase A. Similar to the rather promiscuous M 3 mAChR, two typical G s -coupled recep- tors also induced R-Ras activation. The receptor actions were mimicked by an Epac-specific cyclic AMP analog and sup- pressed by depletion of endogenous Epac1 by small interfering RNAs, as well as expression of a cyclic AMP binding-deficient Epac1 mutant, but not by expression of dominant negative Rap GTPases. In vitro studies demonstrated that Epac1 directly interacts with R-Ras and catalyzes GDP/GTP exchange at this GTPase. Finally, it is shown that the cyclic AMP- and Epac- activated R-Ras plays a major role in the M 3 mAChR-mediated stimulation of phospholipase D but not phospholipase C. Col- lectively, our data indicate that GPCRs rapidly activate R-Ras, that R-Ras activation by the GPCRs is apparently directly induced by cyclic AMP-regulated Epac proteins, and that acti- vated R-Ras specifically controls GPCR-mediated phospho- lipase D stimulation. R-Ras, a member of the Ras superfamily of small GTPases, originally cloned through its homology to prototypic H-Ras, has been shown to interact in vitro with the three H-Ras down- stream effectors, Raf-1, phosphatidylinositol (PI) 6 3-kinase, and RalGDS (1– 4). In addition, in vitro interaction of R-Ras with several Ras regulatory proteins, including various guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs), has been reported (5– 8). Despite these simi- larities, studies in different cell types demonstrated that R-Ras has biological functions distinct from classic H-Ras. Notably, although H-Ras inhibits cell adhesion in fibroblasts by reducing the affinity of integrins for their ligands, R-Ras has been shown to promote integrin activation (7, 9 –11). Recently, it has been proposed that R-Ras might enhance integrin-mediated cell attachment and spreading through alterations in the cellular Ca 2 handling, by decreasing the Ca 2 content of the endoplas- mic reticulum (12). The capacity of R-Ras to modulate cell adhesion by maintaining integrin activity can be regulated by phosphorylation of a tyrosine residue in its effector domain by an Eph receptor kinase (13). Meanwhile, it has been reported that Eph receptor signaling to R-Ras is controlled by tyrosine phosphorylation of R-Ras by the Src kinase on tyrosine 66, and by binding of R-Ras to the Src homology 2 domain-containing Eph receptor-binding protein SHEP1, which binds in addition to the Ras-related GTPase Rap1A (14, 15). Notably, Rap1 has been shown to play a key role in the regulation of several aspects of cell adhesion, in particular in integrin-mediated cell adhe- sion (16), and thus R-Ras might modulate integrin signaling in concert with Rap GTPases. Other studies have shown that the semaphorin 4D receptor, plexin-B1, can down-regulate R-Ras activity by acting as an R-Ras-specific GAP in the presence of the Rho family member Rnd1, and thereby suppresses integrin activation and cell adhesion (17). Furthermore, recent studies in different cell types demonstrated that R-Ras is involved in * This work was supported in part by the Deutsche Forschungsgemeinschaft, the Interne Forschungsfo ¨ rderung Essen, and the Fonds der Chemischen Industrie. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. 1 Both authors contributed equally to this work. 2 Recipient of a Basque Government Fellowship. 3 Present address: Institut fu ¨ r Molekulare Entwicklungs- und Zellphysiologie, Universita ¨ t Karlsruhe, 76128 Karlsruhe, Germany. 4 Present address: Institut fu ¨ r Pharmakologie, Ernst-Moritz-Arndt-Universita ¨t Greifswald, 17487 Greifswald, Germany. 5 To whom correspondence should be addressed: Dept. of Molecular Phar- macology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands. Tel.: 31-50-363-3322; Fax: 31-50-363-6908; E-mail: [email protected]. 6 The abbreviations used are: PI, phosphatidylinositol; GEF, guanine nucle- otide exchange factor; GAP, GTPase-activating protein; GPCR, G pro- tein-coupled receptor; mAChR, muscarinic acetylcholine receptor; PLC, phospholipase C; PLD, phospholipase D; PGE 1 , prostaglandin E 1 ; 8-Br- cAMP, 8-bromo-cAMP; 8-pCPT-2Me-cAMP, 8-(4-chlorophenylthio)-2- O-methyl-cAMP; Rp-CPT-cAMPS, 8-pCPT-adenosine-3,5-cyclic mono- phosphorothioate; VASP, vasodilator-stimulated phosphoprotein; PtdEtOH, phosphatidylethanol; IP 3 , inositol 1,4,5-trisphosphate; GST, glutathione S-transferase; siRNA, small interfering RNA; PKA, protein kinase A; GTPS, guanosine 5-[-thio]triphosphate; ddAdo, 2, 5-dideoxyadenosine; BAPTA/AM, 1,2-bis(2-aminophenoxy)ethane- N,N,N,N-tetraacetic acid tetrakis(acetoxymethyl ester); AMP-PNP, adenosine 5-(,-imido)triphosphate. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 281, NO. 31, pp. 21837–21847, August 4, 2006 © 2006 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A. AUGUST 4, 2006 • VOLUME 281 • NUMBER 31 JOURNAL OF BIOLOGICAL CHEMISTRY 21837 by on January 14, 2007 www.jbc.org Downloaded from

Transcript of Cyclic AMP-dependent and Epac-mediated Activation of R-Ras by G Protein-coupled Receptors Leads to...

Cyclic AMP-dependent and Epac-mediated Activationof R-Ras by G Protein-coupled Receptors Leadsto Phospholipase D Stimulation*

Received for publication, May 1, 2006 Published, JBC Papers in Press, June 5, 2006, DOI 10.1074/jbc.M604156200

Maider Lopez De Jesus‡1,2, Matthias B. Stope‡1,3, Paschal A. Oude Weernink‡, Yvonne Mahlke‡,Christof Borgermann‡, Viktoria N. Ananaba‡, Christian Rimmbach‡4, Dieter Rosskopf ‡4,Martin C. Michel§, Karl H. Jakobs‡, and Martina Schmidt‡§5

From the ‡Institut fur Pharmakologie, Universitatsklinikum Essen, 45122 Essen, Germany and the §Department of Pharmacologyand Pharmacotherapy, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands

The activation of the Ras-related GTPase R-Ras, which hasbeen implicated in the regulation of various cellular functions,by G protein-coupled receptors (GPCRs) was studied in HEK-293 cells stably expressing the M3 muscarinic acetylcholinereceptor (mAChR), which can couple to several types of hetero-trimeric G proteins. Activation of the receptor induced a veryrapid and transient activation of R-Ras. Studies with inhibitorsand activators of various signaling pathways indicated thatR-Ras activation by theM3mAChR is dependent on cyclic AMPformation but is independent of protein kinase A. Similar to therather promiscuous M3 mAChR, two typical Gs-coupled recep-tors also induced R-Ras activation. The receptor actions weremimicked by an Epac-specific cyclic AMP analog and sup-pressed by depletion of endogenous Epac1 by small interferingRNAs, as well as expression of a cyclic AMP binding-deficientEpac1 mutant, but not by expression of dominant negative RapGTPases. In vitro studies demonstrated that Epac1 directlyinteracts with R-Ras and catalyzes GDP/GTP exchange at thisGTPase. Finally, it is shown that the cyclic AMP- and Epac-activated R-Ras plays a major role in the M3 mAChR-mediatedstimulation of phospholipase D but not phospholipase C. Col-lectively, our data indicate that GPCRs rapidly activate R-Ras,that R-Ras activation by the GPCRs is apparently directlyinduced by cyclic AMP-regulated Epac proteins, and that acti-vated R-Ras specifically controls GPCR-mediated phospho-lipase D stimulation.

R-Ras, a member of the Ras superfamily of small GTPases,originally cloned through its homology to prototypic H-Ras,

has been shown to interact in vitrowith the three H-Ras down-streameffectors, Raf-1, phosphatidylinositol (PI)6 3-kinase, andRalGDS (1–4). In addition, in vitro interaction of R-Ras withseveral Ras regulatory proteins, including various guaninenucleotide exchange factors (GEFs) and GTPase-activatingproteins (GAPs), has been reported (5–8). Despite these simi-larities, studies in different cell types demonstrated that R-Rashas biological functions distinct from classic H-Ras. Notably,althoughH-Ras inhibits cell adhesion in fibroblasts by reducingthe affinity of integrins for their ligands, R-Ras has been shownto promote integrin activation (7, 9–11). Recently, it has beenproposed that R-Ras might enhance integrin-mediated cellattachment and spreading through alterations in the cellularCa2�handling, by decreasing theCa2� content of the endoplas-mic reticulum (12). The capacity of R-Ras to modulate celladhesion by maintaining integrin activity can be regulated byphosphorylation of a tyrosine residue in its effector domain byan Eph receptor kinase (13). Meanwhile, it has been reportedthat Eph receptor signaling to R-Ras is controlled by tyrosinephosphorylation of R-Ras by the Src kinase on tyrosine 66, andby binding of R-Ras to the Src homology 2 domain-containingEph receptor-binding protein SHEP1, which binds in additionto the Ras-related GTPase Rap1A (14, 15). Notably, Rap1 hasbeen shown to play a key role in the regulation of several aspectsof cell adhesion, in particular in integrin-mediated cell adhe-sion (16), and thus R-Ras might modulate integrin signaling inconcert with Rap GTPases. Other studies have shown that thesemaphorin 4D receptor, plexin-B1, can down-regulate R-Rasactivity by acting as an R-Ras-specific GAP in the presence ofthe Rho family member Rnd1, and thereby suppresses integrinactivation and cell adhesion (17). Furthermore, recent studiesin different cell types demonstrated that R-Ras is involved in* This work was supported in part by the Deutsche Forschungsgemeinschaft,

the Interne Forschungsforderung Essen, and the Fonds der ChemischenIndustrie. The costs of publication of this article were defrayed in part bythe payment of page charges. This article must therefore be herebymarked “advertisement” in accordance with 18 U.S.C. Section 1734 solely toindicate this fact.

1 Both authors contributed equally to this work.2 Recipient of a Basque Government Fellowship.3 Present address: Institut fur Molekulare Entwicklungs- und Zellphysiologie,

Universitat Karlsruhe, 76128 Karlsruhe, Germany.4 Present address: Institut fur Pharmakologie, Ernst-Moritz-Arndt-Universitat

Greifswald, 17487 Greifswald, Germany.5 To whom correspondence should be addressed: Dept. of Molecular Phar-

macology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen,The Netherlands. Tel.: 31-50-363-3322; Fax: 31-50-363-6908; E-mail:[email protected].

6 The abbreviations used are: PI, phosphatidylinositol; GEF, guanine nucle-otide exchange factor; GAP, GTPase-activating protein; GPCR, G pro-tein-coupled receptor; mAChR, muscarinic acetylcholine receptor; PLC,phospholipase C; PLD, phospholipase D; PGE1, prostaglandin E1; 8-Br-cAMP, 8-bromo-cAMP; 8-pCPT-2Me-cAMP, 8-(4-chlorophenylthio)-2�-O-methyl-cAMP; Rp-CPT-cAMPS, 8-pCPT-adenosine-3�,5�-cyclic mono-phosphorothioate; VASP, vasodilator-stimulated phosphoprotein;PtdEtOH, phosphatidylethanol; IP3, inositol 1,4,5-trisphosphate; GST,glutathione S-transferase; siRNA, small interfering RNA; PKA, proteinkinase A; GTP�S, guanosine 5�-[�-thio]triphosphate; ddAdo, 2�,5�-dideoxyadenosine; BAPTA/AM, 1,2-bis(2-aminophenoxy)ethane-N,N,N�,N�-tetraacetic acid tetrakis(acetoxymethyl ester); AMP-PNP,adenosine 5�-(�,�-imido)triphosphate.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 281, NO. 31, pp. 21837–21847, August 4, 2006© 2006 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.

AUGUST 4, 2006 • VOLUME 281 • NUMBER 31 JOURNAL OF BIOLOGICAL CHEMISTRY 21837

by on January 14, 2007 w

ww

.jbc.orgD

ownloaded from

control of cell migration, apparently through spatio-temporalregulation of Rho and Rac activities (18–20). In a recently gen-erated R-Ras-null mouse, increased proliferation of vascularsmooth muscle cells and enhanced angiogenesis was observed(21).Despite the increasing evidence that R-Ras is obviously a key

regulator of several cellular processes, our knowledge about themechanisms leading to activation of R-Ras is very limited. Inparticular, it should be mentioned that in most studies the bio-logical function of R-Raswas analyzed by ectopically expressingconstitutively active or dominant negativeR-Rasmutants. Acti-vation of R-Ras, i.e.GTP loading, is catalyzed by several distinctGEFs, comprising about 20 distinct proteins (5–7). Of note,there is no report on a GEF that is specific for R-Ras. For exam-ple, the Ca2�/diacylglycerol-regulated GEFs, RasGRP1–3, havebeen reported to activate R-Ras, H-Ras, and Rap1 (22, 23),although C3G and AND-34 have been demonstrated to pro-mote GTP loading on R-Ras and Rap1 (22, 24, 25). It should beemphasized, however, that none of these studies addressed thequestionwhether activation of R-Ras by these GEFs is triggeredby specific membrane receptors.The aim of this report was to study whether and by which

mechanisms G protein-coupled receptors (GPCRs) activateR-Ras and to define a biological function of R-Ras in the GPCRactions. For this, we used the M3 muscarinic acetylcholinereceptor (mAChR) stably expressed in HEK-293 cells. Numer-ous studies have demonstrated that this prototypical GPCR cancouple to all major subtypes of heterotrimeric G proteins, notonly to Gq proteins, as initially assumed (26–28), but also toG12, Gi, andGs proteins. By this distinct G protein coupling, theM3 mAChR can lead to the regulation of various effectorenzymes, including phospholipase C (PLC), phospholipase D(PLD), and adenylyl cyclase, and activation of small GTPasesfrom distinct families (26, 28, 29). Here we report that the M3mAChR strongly activates R-Ras, that this activation requiresendogenously expressed cyclic AMP-activated Epac1 proteinsas shown by silencing of cellular Epac1 using siRNAs, and thatthis cellular response is mimicked by typical Gs-coupled recep-tors. Evidence is provided that Epac-activated R-Ras controlsthe M3 mAChR-mediated stimulation of PLD.

EXPERIMENTAL PROCEDURES

Materials, Expression Plasmids, and Transfection—Forskolin,prostaglandinE1, 2�,5�-dideoxyadenosine (dd-Ado), BAPTA/AM,and H-89 were from Calbiochem,Merck Biosciences, and adren-aline and wortmannin were from Sigma. 8-Br-cAMP, LY294002,and tyrphostin 23were fromBiomol, and 8-(4-chlorophenylthio)-2�-O-methyl cyclic AMP (8-pCPT-2Me-cAMP) and 8-pCPTad-enosine-3�,5�-cyclic monophosphorothioate (Rp-CPT-cAMPS)were from BIOLOG Life Science Institute (Bremen, Germany).GTP�S was from Roche Applied Science. Antibodies against R-Ras, RhoA, and Epac1 were from Santa Cruz Biotechnology; theanti-phosphovasodilator-stimulated phosphoprotein (VASP, Ser-157) antibodywas fromCell SignalingTechnology; the anti-VASPantibody was from Calbiochem,Merck Biosciences, and the anti-cofilin antibody was from tebu-bio. The anti-GAP1 IP4BP ant-ibody was kindly provided by D. Bouyoucef and P. Cullen and amouse monoclonal antibody against Epac1 by J. Bos (30). [3H]-

Oleic acid (5 Ci/mmol) was from PerkinElmer Life Sciences,and [3H]GDP (14.2 Ci/mmol) was fromGEHealthcare. cDNAsencoding wild-type R-Ras, S43N R-Ras, G38V R-Ras, Epac1(each subcloned into pMT2-HA), S17N Rap2B (subcloned inpRK5), the �2-adrenergic receptor (subcloned into pCDNA3),and GAP1 IP4BP (subcloned into pCI-neo) were kindlyprovided by Drs. M. Spaargaren, H. Rehmann, J. de Gunzb-urg, R. Jockers, D. Bouyoucef, and P. Cullen. M3 mAChR-expressing HEK-293 cells and N1E-115 neuroblastoma cellsgrown to near confluence on 145-mm culture dishes weretransfected with an efficiency of at least 50% as reported be-fore (31, 32), typically with 25 �g of DNA from each of the�2-adrenergic receptors andwild-type Epac1, 50�g each ofwil-d-type R-Ras and G38V R-Ras, 100 �g each of S17N Rap2B,S43N R-Ras, and R279K Epac1, or with the indicated amountsof plasmid DNA. For transfection of HEK-293 cells with siR-NAs, cells on 35-mm dishes were transfected with 20 �l ofLipofectamine 2000 in 2 ml of Opti-MEM and 200 pmol ofsiRNA according to the manufacturer’s instructions (Invitrog-en). If vector DNA was co-transfected with siRNA, 4 �g of theindividual vectorDNAwas used in a total amount of 12�g. Thespecific siRNA (Eurogentec) was si-Epac1: 5�-CCAUCAUCC-UGCGAGAAGA99-3� (effective against human Epac1). Toevaluate transfection efficiency, fluorescence-labeled siRNA(Alexa Fluor 488,Qiagen)was used, which also served as unspe-cific control siRNA. Expression of the encoded proteins wasverified by immunoblotting of cell lysates with specific antibod-ies. Assays were performed 48 h after transfection.Measurement of PLD and PLC Activities—PLD activity was

measured in HEK-293 cells prelabeled with [3H]oleic acid asformation of the specific PLD product, [3H]phosphatidyletha-nol ([3H]PtdEtOH), for 30 min at 37 °C in the presence of eth-anol as reported previously (28). Formation of [3H]PtdEtOH isexpressed as percentage of total labeled phospholipids. PLCactivity was measured for 1 min at 37 °C as accumulation ofinositol 1,4,5-trisphosphate (IP3) as described before (29).Activation of R-Ras and VASP Phosphorylation—Serum-

starved HEK-293 cells andN1E-115 neuroblastoma cells trans-fected with wild-type R-Ras were incubated for the indicatedperiods of time at 37 °C with the indicated agents. After celllysis, activatedR-Raswas extractedwith glutathione S-transfer-ase (GST)-tagged Raf1-RBD (Ras-binding domain of Raf-1)bound to glutathione-Sepharose beads and immunoblottingwith the anti-R-Ras antibody as described before (29, 33, 34).Densitometric analysis of the bandswas performedwith Image-Quant software (Amersham Biosciences). For measurement ofVASP phosphorylation, serum-starved HEK-293 cells wereincubated for 5 min at 37 °C with 30 �M forskolin, followed bycell lysis in a buffer containing 1% SDS and 10mMTris/HCl, pH7.4, and five passages through a 25-gauge needle. Thereafter,the lysates were clarified by centrifugation, followed by deter-mination of protein concentration and incubation in Laemmlibuffer for 10 min at 95 °C. After SDS-PAGE and transfer tonitrocellulose membranes, phosphorylated VASP (Ser-157)was detected with the anti-phospho-VASP antibody.Purification of Proteins—GST-tagged R-Ras, Rap2B, and

�DEP Epac1 (Epac1-(149–881)) and His6-tagged C3G-(830–1078) (cDNAs kindly provided by Drs. M. Spaargaren, J. de

R-Ras Activation by Epac

21838 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 31 • AUGUST 4, 2006

by on January 14, 2007 w

ww

.jbc.orgD

ownloaded from

Gunzburg, andH. Rehmann) were expressed in Escherichia coliand purified with glutathione-Sepharose or nickel-nitrilotri-acetic acid superflow agarose beads as described before (31, 33,35). To obtain the soluble and untagged proteins, the immobi-lized proteins were proteolytically digested by thrombin(Rap2B,�DEP Epac1), factor Xa (R-Ras) or eluted with 400mMimidazole (C3G). For protease digestion, the washed beadswere resuspended in thrombin buffer (50mMTris/HCl, pH 8.6,150mMNaCl, 2.5mMCaCl2, 0.1%2-mercaptoethanol) or factorXa buffer (137mMNaCl, 2.7mMKCl, 6.5mMNa2HPO4, 1.5mMKH2PO4, 8.8mMCaCl2, pH7.2), respectively, and incubated for2 h at room temperature with thrombin or factor Xa (followedby overnight incubation at 4 °C). Purification of the proteinswas analyzed by SDS-PAGE. Expression and purification ofGST-tagged ARF1 in Spodoptera frugiperda cells were per-formed as described before (36), except that a different buffer(50mMTris/HCl, pH 7.5, 2mMEDTA, 1mMdithiothreitol, 250mM sucrose, 10 �M phenylmethylsulfonyl fluoride, and 0.5�g/ml leupeptin) was used.Measurement of the GEFActivities of Epac1 and C3G—Bind-

ing ofGDP to R-Ras, Rap2B, andARF1was determined at roomtemperature by the filter bindingmethod (37). In brief, for GDPbinding the recombinant GTPases were first made nucleotide-free by incubation for 5 min in an EDTA-containing loadingbuffer (20 mM Tris/HCl, pH 7.5, 100 mM NaCl, 2 mM EDTA, 1mM AMP-PNP, and 1 mM dithiothreitol), supplemented with 3�M [3H]GDP. Thereafter, MgCl2 was added to a final concen-tration of 5 mM, and the incubation was continued for a further20 min. Finally, purified �DEP Epac1 or C3G-(830–1078)equilibrated for 15 min in exchange buffer, containing 20 mMTris/HCl, pH 7.5, 80 mMNaCl, 5 mMMgCl2, 0.4 mg/ml bovineserum albumin, 1 mM dithiothreitol, 10 mM AMP-PNP, and 10mMGTP, was added in the absence (C3G-(830–1078)) or pres-ence of 1 mM 8-Br-cAMP(�DEP Epac1), and the reaction wascontinued for the indicated periods of time. Bound and free[3H]GDP were separated by adding 1.5 ml of washing buffer(20 mM Tris/HCl, pH 7.5, 8 mM NaCl, 10 mM MgCl2, 0.05%2-mercaptoethanol) and filtration through nitrocellulose fil-ters. The ratio of GEF protein to the GTPases in the exchangeassays typically was 1:1, as assessed from the protein bands ofCoomassie-stained gels.Protein Binding Assays—Serum-starved HEK-293 cells

transfectedwithwild-type Epac1were lysed in a buffer contain-ing 50 mM Tris/HCl, pH 7.5, 200 mM NaCl, 2 mM MgCl2, 1%Nonidet P-40, 10% glycerol, 1 mM phenylmethylsulfonyl fluo-ride, 10 �g/ml benzamidine, 10 �g/ml soybean trypsin inhibi-tor, 0.5 �g/ml aprotinin, and 0.5 �g/ml leupeptin. Nucleotide-free, GDP-bound, and GTP�S-bound forms of recombinantGST-taggedR-Ras, Rap2B, andARF1, immobilized on glutathi-one-Sepharose beads, were prepared exactly as described (38).Thereafter, the beads (5–10 or 50�g of protein) were incubatedwith lysates of the transfected cells (3–4mg of protein) or puri-fied �DEP Epac1 (50 �g of protein) overnight at 4 °C. Finally,after three washes, the beads were resolved in Laemmli buffer,subjected to SDS-PAGE, and transferred to nitrocellulose forWestern blotting, using anti-Epac1 antibodies as indicated.Immunoblot Analysis—For detection of R-Ras, phospho-

VASP,VASP (each at a dilution of 1:500), andEpac1 (dilution of

1:1000–1:10,000), equal amounts of protein were separated bySDS-PAGE on 10% acrylamide gels. After transfer to nitrocel-lulose membranes and a 1-h incubation with the antibodies atthe above given dilution factors, the proteins were visualized byenhanced chemiluminescence.Generation of HEK-293 Cells Stably Expressing Retroviral

Encoded siRNA Epac1—siRNA molecules targeted againstEpac1 were delivered to HEK-293 cells by retroviral transferas described (39) with the following modifications. First, thehuman H1 promoter, suitable for expression of short RNAmolecules, was cloned by PCR from genomic DNA using theprimers 5�-CGCGGTCGACATGGAAATCGAACGCTGAC-GTCATCAACCCGCTC-3� (forward) and 5�-CGCGGTCGA-CCTCGAGGCGCTTCGAAGTGGTCTCATACAGAACTT-ATAAGATTCC-3� (reverse). This construct contains twoartificial restriction sites for XhoI and Bsp119I for site-directedinsertion of the respective siRNA constructs, including suitabletranscription stop signals. The entire construct was sequencedand cloned into the SalI site within the 3�-long terminal repeatof the pQCXIH vector (Clontech). This vector contains ahygromycin resistance gene under the control of a cytomegalo-virus promoter followed by an internal ribosome entry site aswell as enhanced green fluorescent protein sequence. siRNAsequences targeted against human Epac1 were designedaccording to current algorithms and consisted of the cDNA ofthe target transcript followed by nine nucleotides hairpinsequences and the reverse sequence. For human Epac1(GenBankTM accession number NM_006105), the entiresequences were 5�-CGAACCCGGCACTTCGTGGTACATT-ATTCAAGAGATAATGTACCACGAAGTGCCTTTTTGG-AAC-3� (forward) and 5�-TCGAGTTCCAAAAAGGCACTT-CGTGGTACATTATCTCTTGAATAATGTACCACGAAG-TGCCGGGTT-3� (reverse). Two oligonucleotides (0.06 �g/�l,final concentration) encoding these sequences were annealedin a stepwise process for 4 min at 90 °C, 10 min at 70 °C, 5 minat 62 °C, 5min at 37 °C, 5min at 25 °C, and 5min at 10 °C in 100mM NaCl, 50 mM Hepes buffer, pH 7.4. After hybridization theconstruct was ligated into the XhoI/Bsp119I site of the modi-fied pQCXIHvector. All vector constructs usedwere sequence-verified. Packaging GP2–293 cells (Clontech) were co-trans-fected with the respective pQCXIH-siRNA vector and thepVSV-G plasmid (encoding the envelope protein of the vesicu-lar stomatitis virus) by calcium-phosphate transfection. Super-natants were harvested and ultracentrifuged for enrichment ofrecombinant retroviruses. M3 mAChR-expressing HEK-293cells were infectedwith the recombinant retroviruses for 24 h at37 °C, and infection was monitored by fluorescent microscopydetection of enhanced green fluorescent protein. Stablyinfected cells were further selected by incubation with 200�g/ml hygromycin. Epac1 transcripts were measured by quan-titative real time PCR using the fluorescent dye SYBR Green.Total RNA samples were obtained using the NucleoSpin RNAII kit, including a DNA digestion step (Macherey-Nagel)according to the manufacturer’s instructions. As control, cellsinfected with retroviruses lacking a siRNA insert were used.Reverse transcription was done with an oligo(dT)15 primer and1�g of RNAusing SuperScript II reverse transcriptase (Invitro-gen). Quantitative PCR was performed by monitoring the fluo-

R-Ras Activation by Epac

AUGUST 4, 2006 • VOLUME 281 • NUMBER 31 JOURNAL OF BIOLOGICAL CHEMISTRY 21839

by on January 14, 2007 w

ww

.jbc.orgD

ownloaded from

rescence of the SYBR Green dye (Platinum SYBR Green qPCRSuperMix UDG; Invitrogen) on a 7500 real time PCR system(Applied Biosystems). Applied primer pairs were specific forthe sequences of Epac1 (5�-AGTTTCCCACCTCCACGAG-GAC-3�; 5�-ACATAAGCCCAGGTGCTGGCTG-3�) and thehousekeeping gene glyceraldehyde-3-phosphate dehydrogen-ase (5�-GGCGATGCTGGCGCTGAGT-3�; 5�-CATGGTTC-ACACCCATGACGA-3�), which was used for normalization.The relative transcription level of the EPAC1 gene in siRNAexpressing cells compared with vector controls is expressed as�CT values (40).Data Presentation—Data shown in the figures are either rep-

resentative experiments or the mean � S.E. of n independentexperiments, each performed in triplicate. Comparisonsbetween means were either with the Student’s paired t test orone-way analysis of variance test, and a difference was regardedsignificant at p � 0.05.

RESULTS

Activation of R-Ras by theM3mAChR—TheM3mAChR sta-bly expressed in HEK-293 cells can couple to several types ofheterotrimeric G proteins, thereby leading to stimulation ofvarious effector enzymes and activation of small GTPases fromdifferent families (26–29). Thus, we used the M3 mAChR-ex-pressing HEK-293 cells to study whether and by which mecha-nisms GPCRs activate R-Ras. For this, wild-type R-Ras wasoverexpressed in the cells, and upon stimulation GTP-loadedR-Raswas extracted fromcell lysateswith the immobilized Ras-binding domain of Raf-1. As shown in Fig. 1, agonist (carbachol)activation of the receptor induced a very rapid but transientactivation of R-Ras. The stimulatory effect of carbachol reachedits maximum at�1min and declined thereafter (Fig. 1A). Half-maximal and maximal activation was observed at about 5 and100 �M carbachol, respectively (Fig. 1B). To elucidate themechanisms of R-Ras activation, we interfered with the knownsignaling pathways of this receptor. The M3 mAChR potentlystimulates PLC and increases [Ca2�]i in HEK-293 cells, a proc-essmainlymediated byGq-activated PLC-�1 (29). On the otherhand, PLD stimulation by the M3 mAChR is mediated by G12types of G proteins and the small GTPases, RhoA and ARF1,which are activated by a tyrosine kinase- and a PI 3-kinase-de-pendent mechanism, respectively (28, 36, 41). As shown in Fig.2A, chelation of intracellular Ca2� with BAPTA/AM (20 �M)had no effect on the carbachol-induced activation of R-Ras.Similarly, treatment of HEK-293 cells with the general tyrosinekinase inhibitor, tyrphostin 23 (100 �M), which prevented M3mAChR-mediated RhoA activation and PLD stimulation (41),had no effect on activation of R-Ras (Fig. 2A). Activation ofR-Ras was also not affected by the PI 3-kinase inhibitors,LY294002 (10 �M) and wortmannin (200 nM) (Fig. 2B), whichon the other hand prevented activation of ARF1 and stimula-tion of PLD by theM3mAChR (36). Furthermore, treatment ofHEK-293 cells with pertussis toxin and expression of the Cterminus of the �-adrenergic receptor kinase, a G�� scavenger,had no effects on the carbachol-induced R-Ras activation (datanot shown). These data suggested that signaling of the M3mAChR to R-Ras does not require Gq-mediated and PLC-de-pendent [Ca2�]i increase, G12-mediated PLD stimulation,

including activation of PI 3-kinase and tyrosine kinases, andthe small GTPases, RhoA and ARF1, and also not Gi and G��proteins.Cyclic AMP-dependent and Epac-mediated Activation of

R-Ras by the M3 mAChR and Gs-coupled Receptors—We havereported recently that the M3 mAChR activates the RapGTPase Rap2B via Gs-dependent cyclic AMP formation (29).Activation of R-Ras by the M3 mAChR apparently involves

FIGURE 1. Activation of R-Ras by the M3 mAChR. HEK-293 cells transfectedwith wild-type R-Ras were stimulated for the indicated periods of time with-out (�) and with (�) 100 �M carbachol (A) or for 1 min with the indicatedconcentrations of carbachol (B). Thereafter, the levels of R-Ras-GTP and totalR-Ras (Lysate) were determined as described under “Experimental Proce-dures.” Representative immunoblots of R-Ras-GTP and total R-Ras are shown.The lower panels in A and B show mean � S.E. (n � 3– 4).

R-Ras Activation by Epac

21840 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 31 • AUGUST 4, 2006

by on January 14, 2007 w

ww

.jbc.orgD

ownloaded from

cyclic AMP as well. Treatment of HEK-293 cells with the P-siteadenylyl cyclase inhibitor, dd-Ado (10�M), fully suppressed thestimulatory effect of carbachol on activation of R-Ras (Fig. 3A).Furthermore, direct activation of adenylyl cyclase with forsko-lin (30 �M) and treatment of the cells with the membrane-per-

meable cyclic AMP analog, 8-Br-cAMP(1 mM), stronglyenhanced, by 2–2.5-fold, GTP loading of R-Ras (Fig. 3C). How-ever, the activation of R-Ras by cyclic AMP was apparently notmediated by protein kinase A (PKA), the major cellular cyclicAMP target. Treatment of HEK-293 cells with the PKA inhibi-tor, H-89 (10 �M), did not alter activation of R-Ras by eithercarbachol, forskolin, or 8-Br-cAMP (Fig. 3, B and C). Similar toH-89, the PKA inhibitor, Rp-CPT-cAMPS (10 �M), did notaffect GTP loading of R-Ras by the stimulatory agents (data notshown). On the other hand, both PKA inhibitors, H-89 andRp-CPT-cAMPS (not shown), suppressed the forskolin (30�M)-induced and PKA-mediated phosphorylation of endoge-nous VASP proteins (Fig. 3D). Thus, theM3mAChR-mediatedactivation of R-Ras is apparently cyclic AMP-dependent butindependent of PKA.Activation of Rap2B by the M3 mAChR in HEK-293 cells is

mediated by cyclic AMP-regulated Epac proteins (29, 42).Again, the cyclic AMP-dependent and PKA-independent acti-vation of R-Ras is apparently also mediated by Epac proteins,endogenously expressed in HEK-293 cells (34). Treatment ofHEK-293 cells with the membrane-permeable Epac-specificcyclic AMP analog, 8-pCPT-2Me-cAMP (100 �M) (43),induced a robust and rather long lasting activation R-Ras, witha maximum at 5–10 min (Fig. 4A). Half-maximal and maximalactivation was observed at about 10 and 100 �M 8-pCPT-2Me-cAMP, respectively (Fig. 4B). R-Ras activation by 8-pCPT-2Me-cAMPwas not affected by the PKA inhibitorH-89 (10�M)(Fig. 4C).

FIGURE 2. R-Ras activation by the M3 mAChR is independent of tyrosinekinases, Ca2�, and PI 3-kinase. HEK-293 cells transfected with wild-typeR-Ras were first treated for 15 min without (Control) and with 100 �M tyrphos-tin 23, 20 �M BAPTA/AM (A), 10 �M LY294002, or 200 nM wortmannin (B) andthen stimulated for 1 min without (�) and with (�) 100 �M carbachol. Blotsare representative for 3–5 experiments.

FIGURE 3. Activation of R-Ras by the M3 mAChR is cyclic AMP-dependentbut PKA-independent. HEK-293 cells transfected with wild-type R-Ras werefirst treated for 15 min without (Control) and with 10 �M dd-Ado (A) or 10 �M

H-89 (B–D). Then the cells were incubated for 1 min without (�) and with (�)100 �M carbachol (A and B) or for 5 min without (Basal) and with 30 �M fors-kolin or 1 mM 8-Br-cAMP (C ). D, cells were stimulated for 5 min without (�)and with (�) 30 �M forskolin, followed by measurement of phosphorylatedVASP (P-VASP) and the total cellular content of VASP in cell lysates with ananti-phospho-VASP (Ser-157) and an anti-VASP antibody, respectively. Blotsare representative for 3– 6 experiments.

FIGURE 4. Activation of R-Ras by the Epac-specific cyclic AMP analog. HEK-293 cells transfected with wild-type R-Ras were stimulated for the indicatedperiods of time without (�) and with (�) 100 �M 8-pCPT-2Me-cAMP (A) or for5 min with the indicated concentrations of 8-pCPT-2Me-cAMP (B). C, HEK-293cells transfected with wild-type R-Ras were first treated for 15 min without(Control) and with 10 �M H-89 and then incubated for 5 min without (�) andwith (�) 100 �M 8-pCPT-2Me-cAMP. Thereafter, the levels of R-Ras-GTP andtotal R-Ras (Lysate) were determined. Blots are representative for 3–5experiments.

R-Ras Activation by Epac

AUGUST 4, 2006 • VOLUME 281 • NUMBER 31 JOURNAL OF BIOLOGICAL CHEMISTRY 21841

by on January 14, 2007 w

ww

.jbc.orgD

ownloaded from

To substantiate the involvement of Epac proteins in R-Rasactivation, HEK-293 cells were transfected with R279K Epac1,an Epac1 mutant deficient in cyclic AMP binding, which inter-fered with cyclic AMP-dependent Rap2B activation (34). Asshown in Fig. 5A, expression of R279K Epac1 suppressed theactivation of R-Ras by both the M3 mAChR agonist, carbachol,and the Epac-specific cyclic AMP analog, 8-pCPT-2Me-cAMP.Epac proteins act as GEFs for Rap GTPases (35, 42). Therefore,we studied whether Rap GTPases are involved in the activationof R-Ras. However, co-expression of dominant negative S17NRap2B and dominant negative S17NRap1A (not shown), whichprevented activation of H-Ras and extracellular signal-regu-lated kinases byGs-coupled receptors inHEK-293 cells (34), didnot interfere with activation of R-Ras by either carbachol or8-pCPT-2Me-cAMP (Fig. 5A). These data suggested that thecyclicAMP-dependent activation of R-Ras by theM3mAChR ismediated by Epac proteins but apparently independent of RapGTPases. The data presented thus far suggested that the M3mAChR, which can couple to several heterotrimeric G pro-teins, induces R-Ras activation specifically by enhancing cyclicAMP formation. Therefore, it was of interest to know whether

typical Gs- and adenylyl cyclase-coupled receptors may induce acti-vation of R-Ras as well. Stimulationof the �2-adrenergic receptor tran-siently expressed in HEK-293 cellsby adrenaline (10 �M) induced GTPloading of R-Ras, and this R-Rasactivation was fully suppressed byco-expression of R279K Epac1 (Fig.5B). Furthermore, agonist activa-tion of the Gs-coupled receptor forPGE1 endogenously expressed inN1E-115 neuroblastoma cells in-duced activation of R-Ras by about2-fold, similar to treatmentof thecellswith 8-pCPT-2Me-cAMP (Fig. 5B).Again, expression of R279K Epac1suppressed activation of R-Ras byPGE1 and the Epac-specific cyclicAMP analog. To assign Epac1 as anessential regulator of R-Ras activa-tion, its endogenous expression inHEK-293 cells was suppressed byspecific siRNA. As shown in Fig. 5C,this maneuver abolished cellularexpression of Epac1 leaving theexpression of RhoA and cofilinunaffected. Most important, silenc-ing of cellular Epac1 severelyimpaired activation of R-Ras by car-bachol and the Epac-specific cyclicAMP analog (Fig. 5C).Interaction of Epac1 with R-Ras—

The activation of R-Ras by Epacproteins observed in intact cells sug-gested that Epac proteins may binddirectly to R-Ras and catalyze GDP/

GTP exchange at this GTPase. Binding of Epac proteins toR-Ras was studied in comparison to Rap2B and ARF1. For this,GST-tagged Rap2B, R-Ras, and ARF1, each either in the nucle-otide-free, GDP-bound, or GTP�S-bound state, and immobi-lized on glutathione-Sepharose beads, were examined for bind-ing of full-length Epac1 from lysates of HEK-293 cellsoverexpressing the protein or purified �DEP Epac1, Epac1-(149–881), an Epac1 mutant lacking the N-terminal DEPdomain (35). As expected, both full-length Epac1 and �DEPEpac1 specifically bound to Rap2B in its nucleotide-free state(Fig. 6A, upper panel). Most strikingly, a similar efficient bind-ing of full-length Epac1 and �DEP Epac1 to nucleotide-freeR-Raswas observed (Fig. 6A,middle panel), whereas no bindingat all was observed to ARF1, even when this GTPase was pre-sented at a 10-fold excess (Fig. 6A, lower panel).

The GEF activity of Epac1 was determined by measuringrelease of [3H]GDP bound to purified Rap2B, R-Ras, andARF1, in the absence and presence of 8-Br-cAMP and puri-fied �DEP Epac1. As expected, �DEP Epac1 stimulatedrelease of [3H]GDP from purified Rap2B, but only in thepresence of 8-Br-cAMP (Fig. 6B). In contrast, �DEP Epac1

FIGURE 5. Epac-mediated but Rap-independent activation of R-Ras by the M3 mAChR and Gs-coupledreceptors. HEK-293 (A and B, upper panel, and C ) and N1E-115 neuroblastoma cells (B, lower panel) weretransfected with wild-type R-Ras alone (Control) together with the �2-adrenergic receptor (B, upper panel) andwith R279K Epac1, S17N Rap2B, unspecific siRNA (Alexa Fluor 488), or Epac1-specific-siRNA as indicated. At 48 hafter transfection, the cells were incubated for 1 min without (Basal) and with 100 �M carbachol, for 5 min with100 �M 8-pCPT-2Me-cAMP, for 2 min with 10 �M adrenaline or 1 �M PGE1 as indicated, followed by determi-nation of R-Ras-GTP and total R-Ras (Lysate) levels. Blots are representative for 3–5 experiments. C, left panel,immunoblot detection of Epac1, RhoA, and cofilin in lysates of control cells (siRNA Control) and Epac1-specificsiRNA transfected cells (siRNA Epac1).

R-Ras Activation by Epac

21842 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 31 • AUGUST 4, 2006

by on January 14, 2007 w

ww

.jbc.orgD

ownloaded from

exhibited no GEF activity toward ARF1. Most important,�DEP Epac1 induced release of [3H]GDP from R-Ras (Fig.6B). As observed with Rap2B, significant GEF activity of�DEP Epac1 toward R-Ras, although less efficient, was onlyobserved in the presence of 8-Br-cAMP. The release of[3H]GDP from R-Ras induced by 8-Br-cAMP-activated�DEP Epac1 was of a similar magnitude as the releaseinduced by purified C3G-(830–1078), containing the cata-lytic GEF domain of the protein (44).

Role of R-Ras in M3 mAChRSignaling—To study whether R-Rasplays a role inM3mAChR signaling,we first studied the effects of theexpression of the dominant negativeand constitutively active R-Rasmutants, S43N R-Ras and G38VR-Ras, respectively (2, 3), on M3mAChR-mediated PLC and PLDstimulation. As shown in Fig. 7A,neither expression of S43N R-Rasnor that of G38V R-Ras had anyeffect on M3 mAChR-mediated IP3formation. Expression of theseR-Ras mutants had also no effect onthe cellular PLC substrate (phos-phatidylinositol 4,5-bisphosphate)levels (data not shown). In contrast,expression of S43N R-Ras stronglydecreased (by up to 70%) andexpression of G38V R-Ras stronglyincreased (by up to 2-fold) the M3mAChR agonist-induced PLD stim-ulation (Fig. 7B). Inhibition of R-Rassignaling by expression of theGTPase-activating protein GAP1IP4BP, known to stimulate GTPhydrolysis on R-Ras in vitro (45),mimicked the inhibitory effect ofdominant negative R-Ras on PLDstimulation by the M3 mAChR (Fig.7C). These data suggested thatR-Ras plays amajor and specific rolein the M3 mAChR-mediated PLDstimulation.R-Ras activation by the M3

mAChR was dependent on cyclicAMP formation (see Fig. 3A). Inagreement with these data, weobserved that treatment of HEK-293cells with the adenylyl cyclase in-hibitor, dd-Ado (10 �M), stronglyreduced, by about 50%, the M3mAChR-mediated PLD stimulation(Fig. 8A). Furthermore, in agreementwith the involvement of Epac pro-teins, but not PKA, in the M3mAChR-mediated R-Ras activation,it was found that the PKA inhibitor,

H-89 (10 �M), had no effect (Fig. 8A), whereas expression ofR279K Epac1 reduced the M3 mAChR-mediated PLD stimula-tion (Fig. 8B). Conversely, direct stimulation of Epac proteinswith 8-pCPT-2Me-cAMP induced PLD stimulation (Fig. 8C).Compared with the M3 mAChR action, PLD stimulationinduced by 8-pCPT-2Me-cAMP was much less efficient, onlyabout 30% (see ordinates in Fig. 8, A and C), but on the otherhand was more sensitive to inhibition by R279K Epac1. Deple-tion of cellular Epac1 by transient transfection with Epac1-spe-

FIGURE 6. Epac1 binds to and catalyzes GDP/GTP exchange at R-Ras. A, binding of wild-type Epac1 (lysatesof HEK-293 cells transfected with wild-type Epac1; left panels) or purified recombinant �DEP Epac1 (rightpanels) to GST-tagged Rap2B and R-Ras (5–10 �g), and ARF1 (50 �g of protein), each either in the nucleotide-free (NF), GDP-bound, or GTP�S-bound state, and immobilized to glutathione-Sepharose beads, was deter-mined as described under “Experimental Procedures.” Bound Epac1 proteins were resolved by SDS-PAGE andidentified with anti-Epac1 antibodies. B, release of [3H]GDP bound to Rap2B, R-Ras, and ARF1 was determinedin the absence and presence of 1 mM 8-Br-cAMP, �DEP Epac1, or both for the indicated periods of time. Incomparison, release of [3H]GDP bound to R-Ras was measured in the absence (Control) and presence of C3G-(830 –1078). In the exchange assays the ratio of �DEP Epac1 and C3G-(830 –1078) to the GTPases was adjusted1:1. Bound [3H]GDP is expressed as percent of initial binding. Data shown are means � S.E. (n � 3– 6).

R-Ras Activation by Epac

AUGUST 4, 2006 • VOLUME 281 • NUMBER 31 JOURNAL OF BIOLOGICAL CHEMISTRY 21843

by on January 14, 2007 w

ww

.jbc.orgD

ownloaded from

cific siRNA diminished activation of R-Ras by the M3 mAChRand the Epac-specific cyclicAMPanalog (see Fig. 5E). As shownin Fig. 9A, silencing of cellular Epac1 also reduced the PLDresponse by the M3 mAChR. To corroborate these data, HEK-293 cells stably expressing retroviruses encoding siRNA tar-geted against human Epac1 were generated. Expression ofendogenous Epac1 was clearly reduced in these cells both

on the mRNA and protein level,whereas expression of R-Ras wasnot altered (Fig. 9B, upper panel).The specific abrogation of Epac1 ex-pression resulted in a reduced acti-vation of PLD by the M3 mAChR.

DISCUSSION

Despite its similarities to H-Ras,it is now generally accepted thatR-Ras exhibits unique biologicalfunctions. In particular, R-Ras pro-motes integrin-mediated cell adhe-sion, in part through Rap-depend-ent signaling, and cell migration bymodulating the spatio-temporalregulation of Rho and Rac activities(9–11, 18–20). Activation of R-Ras,i.e.GTP loading, is catalyzed by sev-eral distinct GEFs, e.g. RasGRP1–3,C3G, and AND-34 (5–7, 22–25).However, none of these GEFsappears to be specific for R-Ras, andeven more important, activation ofR-Ras by membrane receptors hasnot been studied at all. Therefore,we examined whether and by whichmechanisms GPCRs activate R-Ras.Here we show for three GPCRs, theM3 mAChR and the �2-adrenergicreceptor stably and transiently ex-pressed in HEK-293 cells, respec-tively, and the PGE1 receptor endo-genously expressed in N1E-115neuroblastoma cells, strong andrapid activation of R-Ras. The acti-vation of R-Ras by these threeGPCRs is apparently because oftheir capacity to couple to the cy-clic AMP-regulated Epac exchangefactors.For screening of GPCR-mediated

R-Ras activation, we used HEK-293cells stably expressing the M3mAChR, which can couple to allmajor subtypes of heterotrimeric Gproteins, most prominently to Gqproteins (26–28). Surprisingly, wefound that GTP loading of R-Ras bythis rather promiscuous GPCR isspecifically dependent on cyclic

AMP formation by adenylyl cyclase, which is not considered aprimary function of the M3 mAChR. Subsequent studies withforskolin, a direct adenylyl cyclase activator, cyclic 8-Br-AMP, amembrane-permeable cyclic AMP analog, and particularly the�2-adrenergic receptor and the PGE1 receptor, two typical Gs-coupled receptors, confirmed that R-Ras activation by GPCRsis indeed dependent on cyclic AMP formation. However, the

FIGURE 7. Involvement of R-Ras in M3 mAChR-mediated PLD stimulation. HEK-293 cells were transfectedwith empty vector (Control), S43N R-Ras, G38V R-Ras, or GAP1 IP4BP DNA at the indicated amounts. At 48 h aftertransfection, PLC- and PLD-catalyzed formation of IP3 (A) and PtdEtOH (B and C ), respectively, was measured inthe absence (Basal, open bars) and presence of 100 �M carbachol (filled bars) as described under “ExperimentalProcedures.” Data shown are means � S.E. (n � 3– 4). Insets, immunoblot detection of R-Ras and GAP1 IP4BP inlysates of transfected cells.

R-Ras Activation by Epac

21844 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 31 • AUGUST 4, 2006

by on January 14, 2007 w

ww

.jbc.orgD

ownloaded from

activation of R-Ras was apparently not mediated by PKA, themajor cellular cyclic AMP effector. Instead, Epac1 proteins,endogenously expressed in HEK-293 and N1E-115 neuroblas-toma cells (34), apparently served as mediators of cyclic AMP-dependent R-Ras activation. First, specific activation of Epacproteins with the cyclic AMP analog, 8-pCPT-2Me-cAMP (43),induced a PKA-independent R-Ras activation. Second, expres-sion of R279K Epac1, an Epac1 mutant deficient in cyclic AMPbinding (34), suppressed activation of R-Ras by both 8-pCPT-2Me-cAMP and the GPCR agonists in HEK-293 and N1E-115neuroblastoma cells. Third, silencing of cellular Epac1 by siR-NAs either by transient transfection of Epac1-specific siRNAorstable expression of retroviral encoded siRNA Epac1 in HEK-293 cells (data not shown) impaired activation of R-Ras by theM3mAChR and by the Epac-specific cyclic AMP analog. Epac1directly binds to R-Ras and, when activated by cyclic AMP,catalyzes the GDP/GTP exchange at this GTPase, probablyleading to activation of R-Ras under cellular settings.We have recently reported that the same GPCRs shown here

to activate R-Ras inHEK-293 andN1E-115 neuroblastoma cellsinduce activation of the Rap GTPase Rap2B and that this proc-ess is mediated by cyclic AMP-activated Epac proteins (31),similar as shown here for activation of R-Ras. As Epac proteinshave been characterized as GEFs specific for Rap GTPases (35,42), we studied whether Rap GTPases may mediate activationof R-Ras. However, co-expression of dominant negative Rap2B,which on the other hand prevented activation of H-Ras andextracellular signal-regulated kinases byGs-coupled receptor inHEK-293 cells (34), or Rap1A did not prevent R-Ras activationby 8-pCPT-2Me-cAMP and theM3mAChR. Thus, in line withthe direct interaction of Epac1 with R-Ras observed in vitrowith purified proteins, these data suggest that cyclic AMP-ac-tivated Epac proteins can induce activation of R-Ras in intactcells largely independent of Rap GTPases. The ineffectivenessof the dominant negative Rap mutants to prevent Epac-medi-ated R-Ras activation in intact cellsmay be due to discrete func-tional and physical Epac signaling units. However, althoughEpac1 bound to R-Ras and Rap2B with similar efficiency, theGEF activity of�DEP Epac1 toward R-Ras was less than towardRap2B but similar to the GEF activity of the R-Ras GEF C3G.Thus, despite previous studies (46–48) we have obtained con-vincing evidence that the cyclic AMP-dependent activation ofR-Ras is directly mediated by Epac proteins both in vivo andin vitro. Several factors may contribute to explain our observa-tion of Epac-mediated R-Ras activation, including the need forspecific cofactors that direct the catalytic activity and/or sub-strate selectivity of Epac, a divergence in the pattern of proteinsexpressed by different cell lines, and the subcellular partition-ing of signaling factors. Similarly, activation of R-Ras by C3Gwas also disputed, and inconsistent observations to some extentwere explained by the notification that R-Ras may act as a pref-erable substrate for C3G in specific subcellular domains (22, 24,

FIGURE 8. Involvement of cyclic AMP and Epac proteins in M3 mAChR-mediated PLD stimulation. A, HEK-293 cells were first treated for 15 minwithout (Control) and with 10 �M H-89 or 10 �M dd-Ado. Thereafter, PLD

activity was determined in the absence (Basal) and presence of 100 �M car-bachol. B and C, HEK-293 cells were transfected without (Control) and withR279K Epac1. After 48 h, PLD activities were measured in the absence (Basal)and presence of 100 �M carbachol (B) or at the indicated concentrations of8-pCPT-2Me-cAMP (C ). Data shown are means � S.E. (n � 3–5).

R-Ras Activation by Epac

AUGUST 4, 2006 • VOLUME 281 • NUMBER 31 JOURNAL OF BIOLOGICAL CHEMISTRY 21845

by on January 14, 2007 w

ww

.jbc.orgD

ownloaded from

49). It may be considered that Epac proteins not only act asGEFs for these GTPases but additionally function as effector ofthe activated GTPase or may translocate the active GTPase topotential effector target sites in the cells. Indeed, Quilliam andco-workers (50) recently reported that Epac2 is able to coupleH-Ras and cyclic AMP signals at the plasma membrane.In search for a biological function of R-Ras in the GPCR

actions, a possible role of R-Ras in the M3 mAChR-mediatedstimulation of PLC andPLDwas analyzed, using dominant neg-ative and constitutively active R-Ras mutants. In line with find-

ings on the cholecystokinin A receptor expressed in Chinesehamster ovary cells (12), no effects of these R-Ras mutants onPLC stimulation by theM3mAChR expressed inHEK-293 cellswere observed. In contrast, dominant negative R-Ras largelyinhibited and constitutively active R-Ras enhanced PLD stimu-lation by theM3mAChR. TheGTPase-activating proteinGAPIIP4BP, known to stimulate GTP hydrolysis on R-Ras in vitro(45), mimicked the inhibitory effect of dominant negativeR-Ras on PLD stimulation by the M3 mAChR. Furthermore, inline with the findings observed on R-Ras activation, PLD stim-ulation by theM3mAChRwas reduced by inhibition of adenylylcyclase, but not PKA, and by expression of R279K Epac1, anddirect activation of Epac proteins increased PLD activity. Mostimportant, silencing of cellular Epac1 decreased M3 mAChRsignaling to PLD as well. As dominant negative Rap GTPasesdid not alter the PLD response (data not shown), and aswe havereported previously that M3 mAChR-mediated PLD stimula-tion is Ras- and Ral-independent (51), such mechanisms prob-ably do not contribute to PLD regulation by R-Ras. However, asshown here Epac-activated R-Ras apparently plays a prominentand specific role in PLD stimulation by GPCRs. Studies are inprogress to define the mechanisms by which R-Ras, in combi-nation with Rho and ARF GTPases (28, 36, 41), controls PLDactivity. As Epac-activated Rap2B has been shown to controlstimulation of PLC-� byGPCRs (29, 31, 32), it appears that Epacproteins by activating the GTPases, Rap2B and R-Ras, act asmultifunctional proteins in GPCR signaling systems. Interest-ingly, R-Ras and PLC-� have been shown recently to acttogether in sustaining protrusivemotility inMCF10A cells (52).In conclusion, our findings obtained with three different

GPCRs in two cell types indicate that activation of R-Ras is arapid and prominent response to GPCR activation and thatthese receptors activate R-Ras by enhancing cyclic AMP forma-tion with subsequent activation of Epac proteins, apparentlydirectly controlling the GDP/GTP exchange reaction at R-Rasindependent of Rap GTPases. As studied with the M3 mAChR,Epac-activated R-Ras is apparently not involved in PLC stimu-lation but plays a major role in stimulation of PLD by thisGPCR.

Acknowledgments—We thank K. Baden, H. Geldermann, D.Petermeyer, and M. Hagedorn for expert technical assistance. Wealso thank Drs. J. de Gunzburg, R. Jockers, H. Rehmann, M.Spaargaren, D. Bouyoucef, P. Cullen, and J. Bos for providingcDNA constructs and antibodies.

REFERENCES1. Lowe, D. G., and Goeddel, D. V. (1987)Mol. Cell. Biol. 7, 2845–28562. Spaargaren, M., and Bischoff, J. R. (1994) Proc. Natl. Acad. Sci. U. S. A. 91,

12609–126133. Spaargaren, M., Martin, G. A., McCormick, F., Fernandez-Sarabia, M. J.,

and Bischoff, J. R. (1994) Biochem. J. 300, 303–3074. Marte, B. M., Rodriguez-Viciana, P., Wennstrom, S., Warne, P. H., and

Downward, J. (1997) Curr. Biol. 7, 63–705. Cullen, P. J., and Lockyer, P. J. (2002) Nat. Rev. Mol. Cell Biol. 3, 339–3486. Quilliam, L. A., Rebhun, J. F., and Castro, A. F. (2002) Prog. Nucleic Acids

Res. Mol. Biol. 71, 391–4447. Kinbara, K., Goldfinger, L. E., Hansen, M., Chou, F.-L., and Ginsberg,

M. H. (2003) Nat. Rev. Mol. Cell Biol. 4, 767–776

FIGURE 9. Effect of Epac1 depletion on M3 mAChR-mediated PLD stimu-lation. HEK-293 cells either transfected with unspecific siRNA (Alexa Fluor488) or Epac1-specific siRNA (200 pmol each) (A), and HEK-293 cells stablyexpressing the vector control (Vector) or Epac1-specific siRNA (siRNA Epac1-expressing cells) (B), were processed to determine PLD activity in the absence(Basal) and presence of 5 �M carbachol. Data shown are means � S.E. (n � 3).B, upper panel, mRNA expression of Epac1 in vector controls and siRNA Epac1-expressing cells (n � 5). Higher �CT values represent lower mRNA levels.Immunoblot detection of Epac1 and R-Ras in lysates of control cells (VectorControl) and siRNA Epac1-expressing cells.

R-Ras Activation by Epac

21846 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 31 • AUGUST 4, 2006

by on January 14, 2007 w

ww

.jbc.orgD

ownloaded from

8. Bernards, A., and Settleman, J. (2004) Trends Cell Biol. 14, 377–3859. Zhang, Z., Vuori, K.,Wang,H., Reed, J. C., and Ruoslahti, E. (1996)Cell 85,

61–6910. Sethi, T., Ginsberg, M. H., Downward, J., and Hughes, P. E. (1999) Mol.

Biol. Cell 10, 1799–180911. Self, A. J., Caron, E., Paterson, H. F., and Hall, A. (2001) J. Cell Sci. 114,

1357–136612. Koopman,W. J. H., Bosch, R. R., van Emst-de Vries, S. E., Spaargaren, M.,

De Pont, J. J. H. H. M., andWillems, P. H. G. M. (2003) J. Biol. Chem. 278,13672–13679

13. Zou, J. X.,Wang, B., Kalo,M. S., Zisch, A.H., Pasquale, E. B., andRuoslahti,E. (1999) Proc. Natl. Acad. Sci. U. S. A. 96, 13813–13818

14. Dodelet, V. C., Pazzagli, C., Zisch, A. H., Hauser, C. A., and Pasquale, E. B.(1999) J. Biol. Chem. 274, 31941–31946

15. Zou, J. X., Liu, Y., Pasquale, E. B., and Ruoslahti, E. (2002) J. Biol. Chem.277, 1824–1827

16. Bos, J. L. (2005) Curr. Opin. Cell Biol. 17, 123–12817. Oinuma, I., Ishikawa, Y., Katoh, H., and Negishi, M. (2004) Science 305,

862–86518. Wozniak,M.A., Kwong, L., Chodniewicz, D., Klemke, R. L., andKeely, P. J.

(2005)Mol. Biol. Cell 16, 84–9619. Jeong, H.-W., Nam, J.-O., and Kim, I.-S. (2005) Cancer Res. 65, 507–51520. Holly, S. P., Larson, M. K., and Parise, L. V. (2005) Mol. Biol. Cell 16,

2458–246921. Komatsu, M., and Ruoslahti, E. (2005) Nat. Med. 11, 1346–135022. Ohba, Y., Mochizuki, N., Yamashita, S., Chan, A. M., Schrader, J. W.,

Hattori, S., Nagashima, K., and Matsuda, M. (2000) J. Biol. Chem. 275,20020–20026

23. Yamashita, S.,Mochizuki, N., Ohba, Y., Tobiume,M., Okada, Y., Sawa, H.,Nagashima, K., and Matsuda, M. (2000) J. Biol. Chem. 275, 25488–25493

24. Mochizuki, N., Ohba, Y., Kobayashi, S., Otsuka, N., Graybiel, A. M.,Tanaka, S., and Matsuda, M. (2000) J. Biol. Chem. 275, 12667–12671

25. Gotoh, T., Cai, D., Tian, X., Feig, L. A., and Lerner, A. (2000) J. Biol. Chem.275, 30118–30123

26. Peralta, E. G., Ashkenazi, A., Winslow, J. W., Ramachandran, J., andCapon, D. J. (1988) Nature 334, 434–437

27. Offermanns, S., Wieland, T., Homann, D., Sandmann, J., Bombien, E.,Spicher, K., Schultz, G., and Jakobs, K. H. (1994) Mol. Pharmacol. 45,890–898

28. Rumenapp, U., Asmus, M., Schablowski, H., Woznicki, M., Han, L.,Jakobs, K. H., Fahami-Vahid, M., Michalek, C., Wieland, T., andSchmidt, M. (2001) J. Biol. Chem. 276, 10168–10174

29. Evellin, S., Nolte, J., Tysack, K., vom Dorp, F., Thiel, M., Oude Weernink,P. A., Jakobs, K. H., Webb, E. J., Lomasney, J. W., and Schmidt, M. (2002)J. Biol. Chem. 277, 16805–16813

30. Kooistra, M. R. H., Corada, M., Decana, E., and Bos, J. L. (2005) FEBS Lett.579, 4966–4972

31. Schmidt, M., Evellin, S., Oude Weernink, P. A., vom Dorp, F., Rehmann,H., Lomasney, J. W., and Jakobs, K. H. (2001)Nat. Cell Biol. 3, 1020–1024

32. von Dorp, F., Sari, A. Y., Sanders, H., Keiper, M., Oude Weernink, P. A.,

Jakobs, K. H., and Schmidt, M. (2004) Cell. Signal. 16, 921–92833. Stope,M. B., vomDorp, F., Szatkowski, D., Bohm, A., Keiper, M., Nolte, J.,

OudeWeernink, P. A., Rosskopf, D., Evellin, S., Jakobs, K.H., and Schmidt,M. (2004)Mol. Cell. Biol. 24, 4664–4676

34. Keiper, M., Stope, M. B., Szatkowski, D., Bohm, A., Tysack, K., vom Dorp,F., Saur, O., OudeWeernink, P. A., Evellin, S., Jakobs, K. H., and Schmidt,M. (2004) J. Biol. Chem. 279, 46497–46508

35. de Rooij, J., Rehmann,H., vanTriest,M., Cool, R. H.,Wittinghofer, A., andBos, J. L. (2000) J. Biol. Chem. 275, 20829–20836

36. Schurmann, A., Schmidt, M., Asmus, M., Bayer, S., Fliegert, F., Koling, S.,Ma�mann, S., Schilf, C., Subauste,M. C., Vo�,M., Jakobs, K.H., and Joost,H.-G. (1999) J. Biol. Chem. 274, 9744–9751

37. Ozaki, N., Shibasaki, T., Kashima, Y., Miki, T., Takahashi, K., Ueno, H.,Sunaga, Y., Yano, H., Matsuura, Y., Iwanaga, T., Takai, Y., and Seino, S.(2000) Nat. Cell Biol. 2, 805–811

38. Lutz, S., Freichel-Blomquist, A., Rumenapp,U., Schmidt,M., Jakobs, K.H.,and Wieland, T. (2004) Naunyn-Schmiedeberg’s Arch. Pharmacol. 369,540–546

39. Barton, G. M., and Medzhitov, R. (2002) Proc. Natl. Acad. Sci. U. S. A. 99,14943–14945

40. Livak, K. J., and Schmittgen, T. D. (2001)Methods (Orlando) 25, 402–40841. Keller, J., Schmidt,M., Hussein, B., Rumenapp,U., and Jakobs, K.H. (1997)

FEBS Lett. 403, 299–30242. Bos, J. L. (2003) Nat. Rev. Mol. Cell Biol. 4, 733–73843. Enserink, J. M., Christensen, A. E., de Rooij, J., van Triest, M., Schwede, F.,

Genieser, H. G., Døskeland, S. O., Blank, J. L., and Bos, J. L. (2002)Nat. CellBiol. 4, 901–906

44. Riese, M. J., Wittinghofer, A., and Barbieri, J. T. (2001) Biochemistry 40,3289–3294

45. Cullen, P. J., Hsuan, J., Truong, O., Letcher, A. J., Jackson, T. R., Dawson,A. P., and Irvine, R. F. (1995) Nature 376, 527–530

46. de Rooij, J., Zwartkruis, F. J. T., Verheijen, M. H. G., Cool, R. H., Nijman,S. M. B., Wittinghofer, A., and Bos, J. L. (1998) Nature 396, 474–477

47. Kawasaki, H., Springett, G. M., Mochizuki, N., Toki, S., Nakaya, M.,Matsuda, M., Housman, D. E., and Graybiel, A. M. (1998) Science 282,2275–2279

48. Kawasaki, H., Springett, G. M., Toki, S., Canales, J. J., Harlan, P., Blumen-stiel, J. P., Chen, E. J., Bany, I. A., Mochizuki, N., Ashbacher, A., Matsuda,M., Housman, D. E., and Graybiel, A. M. (1998) Proc. Natl. Acad. Sci.U. S. A. 95, 13278–13283

49. Ohba, Y., Ikuta, K., Ogura, A., Matsuda, J., Mochizuki, N., Nagashima, K.,Kurokawa, K., Mayer, B. J., Maki, K., Miyazaki, J.-I., and Matsuda, M.(2001) EMBO J. 20, 3333–3341

50. Li, Y., Asuri, S., Rebhuhn, J. F., Castro, A. F., Paranavitana, N. C., andQuilliam, L. A. (2006) J. Biol. Chem. 281, 2506–2514

51. Voss, M., Oude Weernink, P. A., Haupenthal, S., Moller, U., Cool, R. H.,Bauer, B., Camonis, J. H., Jakobs, K. H., and Schmidt, M. (1999) J. Biol.Chem. 274, 34691–34698

52. Ada-Nguema, A. S., Xenias, H., Sheetz, M. P., and Keely, P. J. (2006) J. CellSci. 119, 1307–1319

R-Ras Activation by Epac

AUGUST 4, 2006 • VOLUME 281 • NUMBER 31 JOURNAL OF BIOLOGICAL CHEMISTRY 21847

by on January 14, 2007 w

ww

.jbc.orgD

ownloaded from