Clathrin Adaptor AP2 Regulates Thrombin Receptor Constitutive Internalization and Endothelial Cell...

12
MOLECULAR AND CELLULAR BIOLOGY, Apr. 2006, p. 3231–3242 Vol. 26, No. 8 0270-7306/06/$08.000 doi:10.1128/MCB.26.8.3231–3242.2006 Copyright © 2006, American Society for Microbiology. All Rights Reserved. Clathrin Adaptor AP2 Regulates Thrombin Receptor Constitutive Internalization and Endothelial Cell Resensitization† May M. Paing, 1 Christopher A. Johnston, 1 David P. Siderovski, 1 and JoAnn Trejo 1,2 * Department of Pharmacology 1 and Department of Cell and Developmental Biology, 2 School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599-7365 Received 18 August 2005/Returned for modification 30 September 2005/Accepted 17 January 2006 Protease-activated receptor 1 (PAR1), a G protein-coupled receptor for the coagulant protease thrombin, is irreversibly activated by proteolysis. Unactivated PAR1 cycles constitutively between the plasma membrane and intracellular stores, thereby providing a protected receptor pool that replenishes the cell surface after thrombin exposure and leads to rapid resensitization to thrombin signaling independent of de novo receptor synthesis. Here, we show that AP2, a clathrin adaptor, binds directly to a tyrosine-based motif in the cytoplasmic tail of PAR1 and is essential for constitutive receptor internalization and cellular recovery of thrombin signaling. Expression of a PAR1 tyrosine mutant or depletion of AP2 by RNA interference leads to significant inhibition of PAR1 constitutive internalization, loss of intracellular uncleaved PAR1, and failure of endothelial cells and other cell types to regain thrombin responsiveness. Our findings establish a novel role for AP2 in direct regulation of PAR1 trafficking, a process critically important to the temporal and spatial aspects of thrombin signaling. G protein-coupled receptors (GPCRs) comprise the largest family of receptors in the mammalian genome and elicit cel- lular responses to diverse extracellular stimuli (30). Intracellu- lar trafficking of GPCRs controls temporal and spatial aspects of receptor signaling, including signal termination via removal of activated receptors from G-proteins and signaling effectors at the plasma membrane. Recent studies indicate that activated GPCRs can also signal internally at endocytic vesicles (1, 34). Once agonist dissociates from internalized receptor, GPCRs are then recycled back to the cell surface in a resensitized state com- petent to signal again. Trafficking of internalized GPCRs from endosomes to lysosomes with the consequent receptor degrada- tion is also an important process that terminates receptor signal- ing (36, 37). The regulation of GPCR internalization, recycling, and lysosomal sorting involves specific interactions between re- ceptor “sorting” motifs and endocytic adaptor proteins. However, the mechanisms that mediate trafficking of GPCRs through the endocytic pathway remain poorly defined. Protease-activated receptor 1 (PAR1), prototype of a family of proteolytically activated GPCRs, is a receptor for the coagu- lant protease thrombin. PAR1 is the predominant mediator of thrombin signaling in human platelets, endothelial cells, fibro- blasts, and smooth muscle cells and elicits a variety of cellular responses critical for normal vascular responses as well as cardiovascular disease processes (6, 21). PAR1 is activated by an unusual, irreversible proteolytic mechanism. Thrombin cleaves the extracellular amino terminus of the receptor, un- masking a new amino terminus that acts as a tethered ligand by binding intramolecularly to the receptor to trigger signaling (5, 38, 39). Synthetic peptides that mimic this newly formed amino- terminus can activate PAR1 independent of thrombin and receptor cleavage. The irreversible nature of proteolytic PAR1 activation, by generating a tethered ligand that cannot diffuse away, is distinct from the reversible activation of most GPCRs, raising the question, How do cells regulate thrombin signaling? PAR1 trafficking is essential for the fidelity of thrombin signaling. In unstimulated fibroblasts and endothelial cells, PAR1 cycles constitutively between the cell surface and an intracellular compartment, forming a cytosolic receptor pool protected from thrombin cleavage and activation (13, 15, 18). Upon thrombin exposure, cell surface PAR1 is cleaved, acti- vated and then internalized, sorted predominantly to lyso- somes, and degraded (16, 36). Internalization and lysosomal sorting of irreversibly activated PAR1 are both critical for signal termination (36, 37). After thrombin is removed, un- cleaved PAR1 moves from the intracellular protected pool to the cell surface. This replenishment of the cell surface with uncleaved PAR1 allows for rapid recovery of thrombin signal- ing independent of de novo receptor synthesis (13). However, the sorting motifs and endocytic adaptor proteins that specify the distinct trafficking behaviors of PAR1 are not known. Arrestins are multifunctional adaptor proteins known to in- teract with the clathrin endocytic machinery to mediate GPCR internalization. We previously found that PAR1 internaliza- tion, although dependent on clathrin and dynamin, occurs in- dependent of arrestins (4, 25, 35). Given this observation, and the presence of tyrosine-based motifs in the cytoplasmic tail of PAR1, we examined the function of the adaptor protein com- plex 2 (AP2). AP2 is a plasma membrane-localized clathrin adaptor composed of , 2, 2, and 2 adaptin subunits (3). The 2 subunit binds directly to tyrosine-based sorting signals within the cytoplasmic regions of transmembrane proteins to fa- cilitate internalization through clathrin-coated pits. Our studies here reveal that AP2 directly regulates PAR1 constitutive inter- nalization and is essential for resensitization of endothelial cells and other cell types to thrombin signaling. * Corresponding author. Mailing address: Department of Pharma- cology, University of North Carolina at Chapel Hill, 1106 Mary Ellen Jones Bldg., Chapel Hill, NC 27599-7365. Phone: (919) 843-7691. Fax: (919) 966-5640. E-mail: [email protected]. † Supplemental material for this article may be found at http://mcb .asm.org/. 3231 on May 6, 2016 by guest http://mcb.asm.org/ Downloaded from

Transcript of Clathrin Adaptor AP2 Regulates Thrombin Receptor Constitutive Internalization and Endothelial Cell...

MOLECULAR AND CELLULAR BIOLOGY, Apr. 2006, p. 3231–3242 Vol. 26, No. 80270-7306/06/$08.00�0 doi:10.1128/MCB.26.8.3231–3242.2006Copyright © 2006, American Society for Microbiology. All Rights Reserved.

Clathrin Adaptor AP2 Regulates Thrombin Receptor ConstitutiveInternalization and Endothelial Cell Resensitization†

May M. Paing,1 Christopher A. Johnston,1 David P. Siderovski,1 and JoAnn Trejo1,2*Department of Pharmacology1 and Department of Cell and Developmental Biology,2 School of Medicine,

University of North Carolina, Chapel Hill, North Carolina 27599-7365

Received 18 August 2005/Returned for modification 30 September 2005/Accepted 17 January 2006

Protease-activated receptor 1 (PAR1), a G protein-coupled receptor for the coagulant protease thrombin, isirreversibly activated by proteolysis. Unactivated PAR1 cycles constitutively between the plasma membraneand intracellular stores, thereby providing a protected receptor pool that replenishes the cell surface afterthrombin exposure and leads to rapid resensitization to thrombin signaling independent of de novo receptorsynthesis. Here, we show that AP2, a clathrin adaptor, binds directly to a tyrosine-based motif in thecytoplasmic tail of PAR1 and is essential for constitutive receptor internalization and cellular recovery ofthrombin signaling. Expression of a PAR1 tyrosine mutant or depletion of AP2 by RNA interference leads tosignificant inhibition of PAR1 constitutive internalization, loss of intracellular uncleaved PAR1, and failure ofendothelial cells and other cell types to regain thrombin responsiveness. Our findings establish a novel role forAP2 in direct regulation of PAR1 trafficking, a process critically important to the temporal and spatial aspectsof thrombin signaling.

G protein-coupled receptors (GPCRs) comprise the largestfamily of receptors in the mammalian genome and elicit cel-lular responses to diverse extracellular stimuli (30). Intracellu-lar trafficking of GPCRs controls temporal and spatial aspectsof receptor signaling, including signal termination via removalof activated receptors from G-proteins and signaling effectorsat the plasma membrane. Recent studies indicate that activatedGPCRs can also signal internally at endocytic vesicles (1, 34).Once agonist dissociates from internalized receptor, GPCRs arethen recycled back to the cell surface in a resensitized state com-petent to signal again. Trafficking of internalized GPCRs fromendosomes to lysosomes with the consequent receptor degrada-tion is also an important process that terminates receptor signal-ing (36, 37). The regulation of GPCR internalization, recycling,and lysosomal sorting involves specific interactions between re-ceptor “sorting” motifs and endocytic adaptor proteins. However,the mechanisms that mediate trafficking of GPCRs through theendocytic pathway remain poorly defined.

Protease-activated receptor 1 (PAR1), prototype of a familyof proteolytically activated GPCRs, is a receptor for the coagu-lant protease thrombin. PAR1 is the predominant mediator ofthrombin signaling in human platelets, endothelial cells, fibro-blasts, and smooth muscle cells and elicits a variety of cellularresponses critical for normal vascular responses as well ascardiovascular disease processes (6, 21). PAR1 is activatedby an unusual, irreversible proteolytic mechanism. Thrombincleaves the extracellular amino terminus of the receptor, un-masking a new amino terminus that acts as a tethered ligand bybinding intramolecularly to the receptor to trigger signaling (5, 38,39). Synthetic peptides that mimic this newly formed amino-

terminus can activate PAR1 independent of thrombin andreceptor cleavage. The irreversible nature of proteolytic PAR1activation, by generating a tethered ligand that cannot diffuseaway, is distinct from the reversible activation of most GPCRs,raising the question, How do cells regulate thrombin signaling?

PAR1 trafficking is essential for the fidelity of thrombinsignaling. In unstimulated fibroblasts and endothelial cells,PAR1 cycles constitutively between the cell surface and anintracellular compartment, forming a cytosolic receptor poolprotected from thrombin cleavage and activation (13, 15, 18).Upon thrombin exposure, cell surface PAR1 is cleaved, acti-vated and then internalized, sorted predominantly to lyso-somes, and degraded (16, 36). Internalization and lysosomalsorting of irreversibly activated PAR1 are both critical forsignal termination (36, 37). After thrombin is removed, un-cleaved PAR1 moves from the intracellular protected pool tothe cell surface. This replenishment of the cell surface withuncleaved PAR1 allows for rapid recovery of thrombin signal-ing independent of de novo receptor synthesis (13). However,the sorting motifs and endocytic adaptor proteins that specifythe distinct trafficking behaviors of PAR1 are not known.

Arrestins are multifunctional adaptor proteins known to in-teract with the clathrin endocytic machinery to mediate GPCRinternalization. We previously found that PAR1 internaliza-tion, although dependent on clathrin and dynamin, occurs in-dependent of arrestins (4, 25, 35). Given this observation, andthe presence of tyrosine-based motifs in the cytoplasmic tail ofPAR1, we examined the function of the adaptor protein com-plex 2 (AP2). AP2 is a plasma membrane-localized clathrinadaptor composed of �, �2, �2, and �2 adaptin subunits (3).The �2 subunit binds directly to tyrosine-based sorting signalswithin the cytoplasmic regions of transmembrane proteins to fa-cilitate internalization through clathrin-coated pits. Our studieshere reveal that AP2 directly regulates PAR1 constitutive inter-nalization and is essential for resensitization of endothelial cellsand other cell types to thrombin signaling.

* Corresponding author. Mailing address: Department of Pharma-cology, University of North Carolina at Chapel Hill, 1106 Mary EllenJones Bldg., Chapel Hill, NC 27599-7365. Phone: (919) 843-7691. Fax:(919) 966-5640. E-mail: [email protected].

† Supplemental material for this article may be found at http://mcb.asm.org/.

3231

on May 6, 2016 by guest

http://mcb.asm

.org/D

ownloaded from

MATERIALS AND METHODS

Reagents and antibodies. Human �-thrombin was obtained from EnzymeResearch Laboratories. The PAR1 agonist peptides SFLLRN and TFLLRNPNDK were synthesized as the carboxyl amide and purified by high-pressureliquid chromatography by the University of North Carolina Peptide Facility.N-terminal biotinylated peptides corresponding to the carboxy terminus of hu-man PAR1 (amino acids 396 to 425) were synthesized and purified by high-pressure liquid chromatography by the Tufts University Core Facility (Boston,MA). Hirudin, cycloheximide, carbachol, isoproterenol, uridine triphosphate(UTP), and sucrose were purchased from Sigma. Calcium indicator dye Fura2-acetoxymethyl-ester (Fura2-AM), pluronic acid, 4-bromo A-23187 ionophore,transferrin-Alexa-488, and Alexa-488- and Alexa-594-conjugated goat anti-mouse antibodies were obtained from Molecular Probes. Monoclonal M1 anti-FLAG, �-adaptin, �-actin, and glutathione S-transferase (GST) antibodies wereobtained from Sigma. Anti-AP50 (�2), anti-�-adaptin, and anti-early endosomeantigen (EEA1) monoclonal antibodies were from BD Biosciences. Polyclonal anti-His6 antibody was obtained from Abcam Inc. Anti-�-arrestin polyclonal antibodyA1CT was generously provided by R. J. Lefkowitz (Duke University). A rabbitpolyclonal anti-PAR1 antibody was generated against the amino-terminal peptidesequence YEPFWEDEEKNESGLTEYC, as previously described (17). Horse-radish peroxidase-conjugated goat anti-mouse and anti-rabbit secondary anti-bodies were from Bio-Rad.

cDNAs and cell lines. A previously published cDNA encoding wild-type PAR1containing an amino-terminal FLAG epitope was used to generate receptormutants (18). Mutations were introduced using QuickChange site-directed mu-tagenesis (Stratagene) and confirmed by dideoxy sequencing. FLAG-tagged�2-adrenergic receptor cDNA was a gift from M. von Zastrow (University ofCalifornia, San Francisco). A plasmid encoding green fluorescent protein (GFP)-tagged, dominant-negative (K44A) dynamin 2 was generously provided by M.McNiven (Mayo Clinic and Foundation). HeLa cells stably expressing FLAG-tagged PAR1 wild-type and mutants were generated and maintained as previ-ously described (35). Human umbilical vein endothelial cells (HUVECs) wereobtained from Clonetics and maintained according the manufacturer’s instruc-tions. HUVECs at early passages were used for all experiments.

siRNAs. HeLa cells were transiently transfected with 50 nM of nonspecific(NS) or �2-specific small interfering RNAs (siRNAs) using Lipofectamine 2000according to the manufacturer’s instructions. Experiments were performed �60 hafter transfections. HUVECs (1 � 106) were electroporated with a 600 nMconcentration of either NS or �2 siRNA using technology developed by Amaxa,Inc., and experiments were carried out �48 h later. The �2 siRNA targeting themRNA sequence 5�-GTG GAT GCC TTT CGG GTC A-3� was previouslydescribed (20) and synthesized by Dharmacon, Inc. The NS siRNA 5�-CTA CGTCCA GGA GCG CAC C -3� was used as a negative control.

Internalization assays. Constitutive and agonist-induced PAR1 internaliza-tion were assessed using our previously published receptor-antibody uptakeassay (26, 35).

Immunofluorescence confocal microscopy. HeLa cells or HUVECs were pre-incubated with rabbit polyclonal anti-PAR1 antibody for 1 h at 4°C to label thesurface cohort, washed, and then left untreated or treated in the absence orpresence of agonist for various times at 37°C. Cells were fixed and processed forconfocal microscopy as we previously described (25, 40). Colocalization of �2-adaptin was assessed by incubating permeabilized cells with anti-�-adaptin antibodyfor 1 h at 25°C, followed by species-specific fluorophore-conjugated secondaryantibodies, and then imaged by confocal microscopy. Colocalization of PAR1with �-adaptin was examined in fixed cells permeabilized with 0.2% Triton X-100diluted in phosphate-buffered saline (PBS), after incubation with species-specificfluorophore-conjugated secondary antibodies using confocal microscopy. Theextent of colocalization of PAR1 with either �2-adaptin or �-adaptin was quan-titated by counting the number of PAR1-positive puncta that costained with�2-adaptin or �-adaptin, indicated by the yellow area in the merged image aspreviously described (40). The data are expressed as the percentage of PAR1-positive puncta that costained with either �2-adaptin or �-adaptin and representthe averages of many cell samples examined in independent experiments. Toassess PAR1 colocalization with EEA1, permeabilized cells were incubated withanti-EEA1 antibody for 1 h at 25°C, washed, and then incubated with species-specific secondary fluorophore-conjugated antibodies and imaged by confocalmicroscopy. Transferrin uptake was assessed in serum-deprived HeLa cells byincubation with Alexa-488-conjugated transferrin for 1 h at 4°C; cells werewashed and then warmed to 37°C for 10 min and internalized transferrin–Alexa-488 was imaged by confocal microscopy. Images were collected using a Fluoview300 laser scanning confocal imaging system (Olympus) configured with an IX70fluorescent microscope fitted with a PlanApo 60� oil objective. Fluorescent

images, X-Y sections at 0.28 �m, were collected sequentially at 800 � 600resolution with 2� optical zoom. The final composite image was created usingAdobe Photoshop CS.

Immunoblotting. To detect �2 expression, total cell lysates were resolved bysodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis, transferred tomembranes, and immunoblotted with a monoclonal anti-AP50 (�2) antibody.Blots were then stripped and reprobed with a monoclonal antiactin antibody.Immunoblots were developed with ECL-PLUS (Amersham) and imaged byautoradiography.

GST pull-down and surface plasmon resonance (SPR) binding assays. Togenerate GST fused to the PAR1 cytoplasmic (C)-tail domain, an EcoR I andBamH I insert containing a glycine/serine spacer (GSSG) and PAR1 C-tailresidues 374 to 425 was ligated into compatible sites of pGEX2TK (Amersham).GST constructs were transformed into BL21(DE3) Escherichia coli, and fusionproteins were induced and purified using standard techniques previously de-scribed (41). A His6-tagged �2 expression plasmid (24), encompassing the carboxylterminal amino acid residues 160 to 435 (provided by D. J. Owen, University ofCambridge, United Kingdom), was transformed into BL21(DE3)pLys-S E. coli. Sixliters of culture was grown at 37°C to an optical density at 600 nm (OD600) of 0.8with progressive temperature reduction to 18°C prior to induction of His6-�2expression with 50 �M IPTG (isopropyl-�-D-thiogalactopyranoside) for 20 h at18°C. The soluble cell lysate fraction was subjected to Ni2�-nitrilotriacetic acid(Ni-NTA) resin affinity chromatography, followed by size exclusion chromatog-raphy using established methods (41). Monodispersed fractions of His6-�2 pro-tein were pooled and concentrated using a YM-10 centrifugal filter (Millipore)in a final buffer containing 20 mM HEPES, pH 7.5, 200 mM NaCl, 2 mMdithiothreitol, and 10% glycerol.

For GST pull-down binding assays, 20 �g of GST-PAR1 C-tail fusion proteinor GST alone was immobilized on glutathione-Sepharose 4B beads and thenincubated with 20 �g of His-tagged �2 protein for 3 h at 4°C in binding buffer (10mM Tris-HCl, pH 7.4, 5 mM EDTA, 0.2% Triton X-100). Binding reactions werethen washed three times with binding buffer (without Triton X-100). Proteinsthat remained bound were eluted in 2� SDS gel loading buffer (100 mM Tris-HCl, pH 6.8, 10% SDS, 0.2% bromophenol blue, 1 mM dithiothreitol, and 20%glycerol), resolved by SDS-polyacrylamide gel electrophoresis, transferred tomembranes, and immunoblotted with anti-His antibody or anti-GST antibody.Immunoblots were developed with ECL or ECL-PLUS (Amersham) and imagedby autoradiography.

SPR binding assays were performed at 25°C using a BIAcore 3000 in theUniversity of North Carolina Pharmacology Protein Core. N-terminally biotin-ylated PAR1 C-tail peptides (diluted to 1 �g/ml in BIA running buffer [10 mMHEPES, pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.005% NP-40]) were bound toseparate flow cells of a streptavidin-coated biosensor chip (SA5; Biacore) to asurface density of �1,000 resonance units. Prior to injection, His6-�2 protein wasdiluted to desired concentrations in BIA running buffer. A total of 50 �l ofHis6-�2 protein was then simultaneously injected over flow cells at 10 �l/min,followed by a 200-s dissociation in BIA running buffer. All sensorgram curveswere corrected for bulk buffer refractive index shifts and nonspecific binding bysubtracting corresponding traces from a negative control blank surface. Followingeach injection, surfaces were regenerated with a 10-�l injection of 500 mM NaClplus 20 mM NaOH at 20 �l/min. Binding curves and affinity calculations wereconducted using BIAevaluation (version 3.0) and GraphPad Prism (version 4.0b).

Intracellular calcium measurements. Cells were grown on glass coverslips toa cell density of approximately 30 to 40% of confluence. Intracellular calcium wasmeasured and quantified essentially as previously described (27). Cells loadedwith 5 �M Fura2 were placed in a flowthrough chamber and superfused contin-uously with Hanks balanced salt solution in the presence or absence of agonists.Cells were exposed to alternating excitation wavelengths of 340 and 380 nm, andfluorescence emission at 510 nm was monitored using an integrating charge-coupled-device camera. The 340/380-nm fluorescence emission ratio was deter-mined, and intracellular Ca2� concentration was determined using the equationof Gyrnkiewicz et al. (12). The data were recorded and processed using InCytIM2 digital imaging system (Intracellular Imaging Inc.).

Data analysis. Data were analyzed using Prism 4.0 software, and statisticalsignificance was determined using InStat 3.0 (GraphPAD). Group comparisonswere made using an unpaired Student’s t test.

RESULTS

PAR1 interacts directly with �2 in vitro and colocalizes withendogenous AP2 in vivo. Using arrestin-deficient mouse em-bryonic fibroblasts, we previously found that PAR1 internalizes

3232 PAING ET AL. MOL. CELL. BIOL.

on May 6, 2016 by guest

http://mcb.asm

.org/D

ownloaded from

through a clathrin- and dynamin-dependent pathway indepen-dent of arrestin function (25). Arrestin-independent PAR1internalization through clathrin-coated pits is also observed inHeLa cells and other cell types (4, 35) (see Fig. S1 in thesupplemental material). To delineate the mechanism respon-sible for PAR1 internalization through clathrin-coated pits, weinitially examined whether PAR1 could interact with the �2subunit of AP2. Purified, His6-tagged �2 protein was found tobind directly to a GST-PAR1 C-tail fusion protein absorbed toglutathione-Sepharose beads, but not to GST protein alone(Fig. 1A). We next determined whether PAR1 colocalizes withthe clathrin adaptor AP2 in intact cells using confocal micros-copy. HeLa cells stably expressing FLAG-tagged PAR1 wereincubated with a polyclonal anti-PAR1 antibody at 4°C toensure that only cell surface receptors would be labeled. Cells

were then warmed to 37°C for 2.5 min in the absence ofagonist; under these conditions unactivated PAR1 is recruitedto clathrin-coated pits (35). Cells were then fixed and immu-nostained for PAR1 and the AP2 subunit �2-adaptin. Approx-imately 30% of PAR1-positive puncta costained for �2-adaptin(Fig. 1B), suggesting that the majority of constitutively inter-nalized receptors recruited to clathrin-coated pits colocalizewith AP2 in intact cells (Fig. 1B).

AP2 is essential for PAR1 constitutive internalization. Toassess AP2 function in PAR1 trafficking, we used siRNA tar-geting the �2 subunit to deplete HeLa cells of endogenousAP2 complex (20). Expression of �2 was virtually abolished incells transiently transfected with �2 siRNA compared to cellstransfected with a nonspecific control (Fig. 2A, inset). Cellstransfected with �2 siRNA also showed significant decreases in�2-adaptin expression (Fig. 2C), consistent with a loss of en-dogenous AP2 complex (20). Inhibition of transferrin uptakein �2 siRNA-transfected cells (see Fig. S2 in the supplementalmaterial) further indicates that �2 knockdown is sufficient todeplete HeLa cells of AP2 function, as the transferrin receptoris known to internalize constitutively and recycle through anAP2-, clathrin- and dynamin-dependent pathway (3).

To determine whether AP2 is necessary for PAR1 internal-ization, a stable HeLa cell line expressing FLAG-tagged PAR1was labeled with anti-FLAG antibody and then incubated withor without agonist at 37°C for various times to allow internal-ization of receptor-bound antibody. In control siRNA-trans-fected cells not exposed to agonist, �20% of antibody initiallybound to the cell surface was internalized at steady state (Fig.2A and B), consistent with the extent of PAR1 constitutiveinternalization observed in other cell types (13, 32). In con-trast, PAR1 constitutive internalization was inhibited signifi-cantly in �2 siRNA-transfected cells (Fig. 2A and B), in spiteof an increase in steady-state amounts of surface receptor inthese cells (data not shown). These findings suggest that AP2is necessary for PAR1 constitutive internalization. Exposure tothe agonist peptide SFLLRN for 10 min caused substantialPAR1 internalization irrespective of siRNA treatment (Fig.2B). Interestingly, in cells depleted of �2, activated PAR1internalization occurred through a clathrin- and dynamin-de-pendent pathway (see Fig. S3 in the supplemental material).These findings suggest that agonist-activated PAR1 internal-ization is independent of AP2 function.

Immunofluorescence studies of PAR1-expressing HeLa cellswere consistent with an AP2-dependent regulation of receptorconstitutive internalization. In control siRNA-transfected cells,a 10-min incubation at 37°C caused PAR1 to redistribute fromthe cell surface into endocytic vesicles (Fig. 2D), consistentwith tonic cycling between the plasma membrane and anintracellular compartment. In contrast, cell surface PAR1failed to internalize in unstimulated �2 siRNA-transfectedcells after 10 min at 37°C (Fig. 2D). Similar results were ob-served in cells transfected with a different �2 siRNA targetinga distinct mRNA sequence (9) (data not shown). The additionof SFLLRN, however, induced significant accumulation ofinternalized PAR1 in both control- and �2 siRNA-transfectedcells (Fig. 2D), confirming that AP2 is not required for agonist-activated PAR1 internalization.

To determine whether AP2 regulates trafficking of endoge-nous receptor in native cells, we examined PAR1 internaliza-

FIG. 1. The �2 subunit of AP2 interacts with the PAR1 C tail invitro and PAR1 colocalizes with endogenous AP2 in intact cells.(A) GST-PAR1 C-tail fusion protein or GST protein alone absorbedto glutathione-Sepharose beads was incubated with purified His-tagged �2. Bound proteins were eluted and immunoblotted with ananti-His antibody (upper panel). An aliquot of purified His-tagged �2representing 10% input is shown in the last lane. Membranes werestripped and reprobed with an anti-GST antibody to determine thetotal amount of GST-PAR1 C tail and GST protein loaded in variouslanes (lower panel). (B) HeLa cells stably expressing FLAG-taggedPAR1 were preincubated with anti-PAR1 antibody for 1 h at 4°C suchthat only cell surface receptor-bound antibody. Cells were then incu-bated for 2.5 min at 37°C with no agonist, fixed, permeabilized, andimmunostained for PAR1 (red) using a polyclonal anti-PAR1 antibodyand for endogenous �2-adaptin (green) using a monoclonal anti-�2-adaptin antibody and imaged by confocal microscopy. Colocalizationof PAR1 with AP2 is shown in yellow in the merged image. The imageshown is representative of many cells examined in three independentexperiments. Scale bar, 2 �m.

VOL. 26, 2006 AP2 DIRECTLY REGULATES PAR1 SIGNALING AND TRAFFICKING 3233

on May 6, 2016 by guest

http://mcb.asm

.org/D

ownloaded from

tion in HUVECs. HUVECs electroporated with �2 siRNAshowed significant loss of �2 protein and �2-adaptin expres-sion compared to control siRNA-treated cells, suggesting thatin these cells endogenous AP2 complex has been depleted(Fig. 3). A significant amount of internalized PAR1 was foundin endocytic vesicles in control cells after 15 min at 37°C (Fig.3B). By contrast, PAR1 failed to redistribute to endocyticvesicles in �2 siRNA-transfected cells (Fig. 3B), suggesting anAP2-dependent regulation of PAR1 constitutive internaliza-tion in HUVECs. Incubation with the PAR1-specific agonistpeptide TFLLRNPNDK caused a marked increase in recep-tor-containing endocytic vesicles in both control- and �2siRNA-transfected cells (Fig. 3C), indicating that activatedPAR1 internalizes independent of AP2. Together these find-ings reveal a novel function for AP2 in regulating PAR1 con-stitutive internalization.

A distal tyrosine-based motif within PAR1 is required for �2binding and constitutive internalization. The �2 subunit ofAP2 binds directly to tyrosine-based sequences in cytoplasmic

domains of transmembrane proteins and thereby facilitatesrecruitment to clathrin-coated pits at the cell surface (3). Asearch of the PAR1 sequence for such sorting signals revealedtwo highly conserved sequences in the C-tail region conform-ing to the YXXØ motif (where Y denotes tyrosine, X is anyamino acid, and Ø is a bulky hydrophobic amino acid) (Fig.4A). The interaction of �2 with tyrosine-based sequences isstrictly dependent on the tyrosine and bulky hydrophobic res-idue positions; however, residues flanking the motif also con-tribute to �2 binding (3). To test the role of each tyrosine-based motif in PAR1 trafficking, we generated receptormutants in which critical positions (Y383 and L386 or Y420 andL423) were converted to alanines (Fig. 4A). The L424 residueflanking the distal tyrosine-based motif was also mutated toalanine to avoid the possibility of compensatory effects on �2binding. In wild-type PAR1-expressing HeLa cells, �10 to 20%of antibody-labeled cell surface receptor was constitutivelyinternalized at steady state (Fig. 4B and C). The PAR1A383SIA386 mutant exhibited levels of constitutive and agonist-

FIG. 2. PAR1 constitutive internalization, but not agonist-induced internalization, is inhibited in AP2-depleted HeLa cells. (A) HeLa cellsstably expressing FLAG-tagged, wild-type PAR1 were transiently transfected with 50 nM siRNA targeted to either �2 or nonspecific (ns) mRNAsequences. Surface FLAG-tagged PAR1 receptors were then labeled with the Ca2�-dependent M1 anti-FLAG antibody for 1 h at 4°C. Cells werethen incubated (without agonist) for various times at 37°C to allow constitutive internalization of receptor-bound antibody. After incubations, cellswere stripped of antibody remaining bound to the cell surface with PBS–0.04% EDTA and lysed, and internalized antibody was then quantifiedby enzyme-linked immunosorbent assay. Data (mean standard error of the mean; n 3) are expressed as a percentage of initial cell surfacereceptor-bound antibody, defined as total antibody bound to cells at 0 min and not washed with PBS-EDTA. Similar results were obtained in atleast three independent experiments. The immunoblot of equivalent amounts of cell lysates shown in the inset confirms the loss of �2 protein in�2 siRNA-transfected cells, whereas actin expression was unaffected. (B) PAR1-expressing HeLa cells transiently transfected with �2 or ns siRNAswere labeled with antibody as described above and then incubated in the presence or absence of 50 �M SFLLRN agonist peptide for 10 min at37°C. Internalized receptor-bound antibody was quantified as described above. The data (mean standard error of the mean; n 3) shown arerepresentative of at least three separate experiments. (C) PAR1-expressing HeLa cells transiently transfected with �2 or ns siRNAs were fixed,permeabilized, and immunostained for �2-adaptin expression and imaged by confocal microscopy. The differential interference contrast (DIC)image is the same field of �2-transfected cells shown in the adjacent anti-�2-adaptin fluorescence image. (D) PAR1-expressing HeLa cellstransiently transfected with �2 or ns siRNAs were either left untreated (0 min) or treated in the absence or presence of 50 �M SFLLRN for 10min at 37°C. Cells were fixed, immunostained for PAR1, and imaged by confocal microscopy. The imaged cells are representative of many cellsexamined in three different experiments. Insets are magnifications of boxed areas. Scale bar, 10 �m.

3234 PAING ET AL. MOL. CELL. BIOL.

on May 6, 2016 by guest

http://mcb.asm

.org/D

ownloaded from

induced internalization similar to wild-type (Fig. 4B and C),indicating that the proximal tyrosine-based motif is not essen-tial for either process. In striking contrast, however, PAR1A420KKA423A424 mutants failed to constitutively internalize,

suggesting that the distal tyrosine-based motif functions inPAR1 tonic cycling (Fig. 4B and C). A similar defect in con-stitutive internalization was observed upon mutation of onlythe distal tyrosine (Y420) to alanine, whereas mutation of theL423L424 residues had no significant effect (Fig. 4D), indicatingthat the critical Y420 is essential for function. However, whenactivated, the PAR1 A420KKA423A424 mutant internalized sim-ilar to wild-type receptor (Fig. 4C), indicating that the distaltyrosine-based motif is not essential for agonist-activated re-ceptor internalization.

Immunofluorescence confocal microscopy was performed toconfirm the function of tyrosine-based motifs in receptor traf-ficking. In untreated control cells, wild-type PAR1 and YXX Ømutants were each found predominantly at the cell surface anddid not colocalize with EEA1, a specific marker of early en-dosomes (Fig. 5a to d). After 10 min at 37°C, wild-type PAR1and A383SIA386 mutant moved from the cell surface to endo-cytic vesicles and markedly colocalized with EEA1, suggestingthat constitutively internalized receptor transits through anearly endosomal compartment (Fig. 5a� and b�). Addition ofSFLLRN caused an even greater increase in wild-type PAR1and A383SIA386 mutant internalization (Fig. 5a� and b�). Incontrast, A420KKA423A424 mutant PAR1 failed to redistribute toEEA1-positive endosomes after 10 min at 37°C (Fig. 5c� and d�),consistent with a lack of receptor constitutive internalization.However, agonist treatment induced a significant amount ofA420KKA423A424 mutant internalization comparable to that ofwild-type receptor (Fig. 5c� and d�).

We next determined whether the distal tyrosine-based motif(Y420KKL423) was important for PAR1 colocalization with en-dogenous AP2 in cells. HeLa cells expressing wild-type PAR1or A420KKA423A424 mutant were labeled with a polyclonalanti-PAR1 antibody at 4°C so that only the surface receptorcohort binds antibody. PAR1 recruitment to clathrin-coatedpits was then initiated by warming cells to 37°C for 2.5 min(35). Cells were fixed and immunostained for PAR1 and�-adaptin, and colocalization was assessed by confocal mi-croscopy. Approximately 29% of wild-type PAR1 localized todistinct puncta that costained for the AP2 subunit �-adaptin(Fig. 6a). In contrast, A420KKA423A424 mutant remained pre-dominantly in puncta that showed minimal colocalization with�-adaptin (Fig. 6b); only �8% of A420KKA423A424 mutant-positive puncta costained for the �-adaptin subunit. Thus, mu-tation of the distal tyrosine-based motif disrupts PAR1 colo-calization with endogenous AP2 in intact cells, consistent withthe failure of mutant receptor to undergo constitutive inter-nalization.

These findings from cellular expression of PAR1 YXXØ mu-tants suggest that the distal tyrosine-based motif Y420KKL423 iscritical to PAR1 constitutive internalization and colocalizationwith AP2 in intact cells. We therefore tested whether this distalmotif was necessary for the observed direct binding of �2protein to the PAR1 C-tail (Fig. 1A). Recombinant His6-tagged �2 protein, purified from E. coli expression by nickel-NTA and size exclusion chromatography (Fig. 7A), was in-jected over streptavidin-coated SPR biosensors containingimmobilized biotinylated PAR1 C-tail peptides (Fig. 7B andC). The dissociation constant (KD) for the binding of �2 to thewild-type PAR1 C-tail was 7.5 1.3 �M. In contrast, no

FIG. 3. AP2 is essential for endogenous PAR1 constitutive inter-nalization, but not agonist-induced internalization, in HUVECs.(A) HUVECs were electroporated with �2 or nonspecific (ns) siRNAs.Cell lysates were prepared and immunoblotted (IB) for �2 expressionor actin expression. (B and C) HUVECs electroporated with �2 or nssiRNAs were incubated with anti-PAR1 antibody for 1 h at 4°C; underthese conditions only receptors on the cell surface bound antibody.Cells were washed and then incubated in the absence or presence of100 �M TFLLRNPNDK agonist peptide for 15 min at 37°C. Cellswere fixed, immunostained for PAR1 and �2-adaptin expression, andimaged by confocal microscopy. These images are representative ofmany cells examined in at least three independent experiments. Theinsets are magnifications of boxed areas. Scale bar, 10 �m.

VOL. 26, 2006 AP2 DIRECTLY REGULATES PAR1 SIGNALING AND TRAFFICKING 3235

on May 6, 2016 by guest

http://mcb.asm

.org/D

ownloaded from

detectable binding was observed to the A420KKA423A424 mu-tant PAR1 C-tail peptide at any concentration of �2 proteintested (Fig. 7C), indicating that the distal YXXØ motif isindeed required for direct �2 association.

Recovery of cellular thrombin signaling is dependent onAP2-regulated PAR1 constitutive internalization. Constitutivecycling of PAR1 between the plasma membrane and an intra-cellular compartment results in a protected pool of uncleavedreceptors that repopulate the cell surface following thrombinexposure without de novo receptor synthesis (13). This processis critical for rapid cellular resensitization to thrombin signal-ing. We therefore assessed the role of AP2 and tyrosine-basedmotifs in the recovery of cellular thrombin responsiveness bymeasuring thrombin-stimulated increases in intracellular Ca2�

after an initial thrombin exposure in the presence of cyclohex-imide, a protein synthesis inhibitor. Adding a saturating con-centration of thrombin resulted in cleavage and activation ofall surface-bound wild-type or mutant PAR1 expressed inHeLa cells (data not shown). In wild-type PAR1-expressingcells, an initial exposure to thrombin induced a transient in-crease in cytosolic Ca2� (Fig. 8A), comparable to that ob-served in other cell types (13). Cells were immediately refrac-tory to thrombin. After thrombin removal, cells were allowedto recover for 20 min in the presence of hirudin (a thrombininhibitor) and cycloheximide and then exposed to thrombinagain, which elicited a second rise in intracellular Ca2� (Fig.8A). This time course of recovery is similar to that observedwith PAR1-expressing fibroblasts (13). Cells expressing thePAR1 A383SIA386 mutant also exhibited recovery of thrombinsignaling after an initial thrombin exposure (Fig. 8B). Thesefindings are consistent with the idea that wild-type PAR1 andA383SIA386 mutant undergo constitutive internalization, gen-erating an internal pool of protected receptors that recyclesback to the cell surface (13, 26). Cells expressing the PAR1A420KKA423A424 mutant displayed an initial thrombin-stimu-lated increase in cytosolic Ca2� comparable to wild-type receptor;however, these cells failed to elicit a second response to thrombin(Fig. 8C and D). The ability of PAR1 A420KKA423A424 mu-tant-expressing cells to respond to carbachol, an agonist forendogenous muscarinic acetylcholine receptors, indicatesthat the cells were not globally defective in GPCR-stimu-lated Ca2� mobilization after initial thrombin exposure(Fig. 8E and F). Thus, the failure of PAR1 A420KKA423A424

mutant-expressing cells to regain thrombin signaling ap-pears to be linked to the lack of an internal pool of un-cleaved receptors that replenish the cell surface and permitrecovery of thrombin responsiveness.

We next examined whether AP2 regulation of PAR1 consti-tutive internalization affected cellular resensitization to throm-bin signaling. In PAR1-expressing HeLa cells transfected with

FIG. 4. A PAR1 cytoplasmic tail distal tyrosine-based motif regu-lates constitutive but not agonist-induced receptor internalization.(A) PAR1 cytoplasmic tail amino acid sequence is shown, and the posi-tions of the tyrosine-based motifs are indicated. Superscripted numbersindicate the critical tyrosines and leucines that were mutated to alanines.The asterisk indicates the end of the protein sequence. (B) HeLa cellsstably expressing similar amounts of FLAG-tagged, wild-type PAR1 orreceptor mutants were labeled with antibody and incubated (withoutagonist) for various times at 37°C, and internalized receptor was quan-tified as described above. The data (mean standard error of themean; n 3) shown are expressed as a percentage of total cell surfacereceptor-bound antibody at 0 min and are representative of at leastthree separate experiments. (C) HeLa cells stably expressing wild-typePAR1 or receptor mutants were labeled with antibody and incubatedin the presence or absence of 50 �M SFLLRN agonist peptide for 10min at 37°C, and the amount of receptor internalization was quanti-tated as described above. Data are representative of at least threeindependent experiments. (D) HeLa cells expressing comparableamounts of FLAG-tagged PAR1 wild-type, LL423/424AA, orY420A mutants were labeled with antibody, washed, incubated inthe presence or absence of 50 �M SFLLRN agonist peptide for 30min at 37°C, and then quantitated. The data (mean standard errorof the mean; n 3) shown are representative of at least three

independent experiments. A significant difference in constitutive en-docytosis of the Y420A mutant compared to wild-type receptor wasdetected (�, P � 0.05), whereas no significant difference in constitutiveendocytosis of the LL423/424AA mutant compared to wild-typereceptor was detected. Statistical analysis was determined using anunpaired Student’s t test. WT, wild-type; ASIA, A383SIA386; AKKAA,A420KKA423A424.

3236 PAING ET AL. MOL. CELL. BIOL.

on May 6, 2016 by guest

http://mcb.asm

.org/D

ownloaded from

control siRNA, thrombin elicited an initial increase in cytosolicCa2�, and after recovery virtually all cells regained thrombinresponsiveness (Fig. 9A and C). An initial Ca2� response tothrombin was also observed in �90% of �2 siRNA-transfectedcells; however, after recovery only �20% of cells were capableof eliciting a second response to thrombin (Fig. 9B and C). Incontrast, carbachol-induced calcium mobilization was main-tained in the majority of �2 siRNA-transfected cells followingthrombin exposure (Fig. 9D), indicating that cells were capableof responding to other GPCR agonists.

Resensitization of HUVECs to thrombin signaling was alsoassessed. Nearly 80% of HUVECs electroporated with controlsiRNA were initially responsive to thrombin and immedi-ately refractory to a second thrombin exposure, but after a20-min recovery period, almost all cells (�75%) again eliciteda thrombin response (Fig. 9E and G). These findings are con-sistent with the recovery of thrombin responsiveness previouslyobserved in these cells (13). In contrast, however, only �10%of HUVECs with siRNA-mediated �2 knockdown regainedthrombin responsiveness following initial thrombin exposureand a recovery period (Fig. 9F and G). Signaling by UTP, anagonist for endogenous P2Y2 and/or P2Y4 GPCRs, was unper-turbed, indicating that �2 siRNA-electroporated cells are re-sponsive in general to GPCR activation after an initial throm-bin exposure (Fig. 9H). Taken together, these findings stronglysuggest that AP2-dependent regulation of PAR1 constitutiveinternalization is essential for recovery of cellular responses tothrombin.

FIG. 5. Immunofluorescence reveals a critical role for the PAR1 cytoplasmic tail distal tyrosine-based motif in constitutive but not agonist-induced internalization. HeLa cells expressing comparable amounts of wild-type (WT) PAR1 or receptor mutants were either left untreated (a tod; 0 min) or incubated in the absence (a� to d�; 10 min) or presence (a� to d�; �SFLLRN 10 min) of 50 �M SFLLRN agonist peptide for 10 minat 37°C. Cells were fixed and coimmunostained for PAR1 (green), using a polyclonal anti-PAR1 antibody, and for the early endosome markerEEA1 (red), using a monoclonal anti-EEA1 antibody, and imaged by confocal microscopy. The insets are magnifications of boxed areas. Theimaged cells are representative of many cells examined in at least three independent experiments. Scale bar, 10 �m. ASIA, A383SIA386; AKKAA,A420KKA423A424.

FIG. 6. The distal tyrosine motif is important for PAR1 colocaliza-tion with AP2 in intact cells. HeLa cells expressing PAR1 wild-type(WT) or the distal tyrosine motif mutant A420KKA423A424 (AKKAA)were prelabeled with anti-PAR1 antibody, washed, and then warmedto 37°C for 2.5 min to initiate PAR1 recruitment to clathrin-coatedpits. Cells were fixed and coimmunostained for PAR1, (green) using apolyclonal anti-PAR1 antibody, or for the AP2 subunit �-adaptin(red), using a monoclonal anti-�-adaptin antibody, and imaged byconfocal microscopy. Colocalization of PAR1 with AP2 is revealed bythe yellow areas in the merged image. The insets are magnifications ofboxed areas. The image shown is representative of many cells exam-ined in three different experiments. Scale bar, 10 �m.

VOL. 26, 2006 AP2 DIRECTLY REGULATES PAR1 SIGNALING AND TRAFFICKING 3237

on May 6, 2016 by guest

http://mcb.asm

.org/D

ownloaded from

DISCUSSION

In the present study, we have defined a hitherto unknownrole for the clathrin adaptor AP2 in regulating PAR1 consti-tutive internalization and restoring thrombin responsiveness toendothelial cells and other cell types. The �2 subunit of AP2directly interacts with a tyrosine-based motif present at the

extreme carboxy terminus of PAR1. This tyrosine-based motifand AP2 function are each critical for maintaining an intracel-lular store of protected receptors that replenish the cell surfacewith uncleaved PAR1 after thrombin exposure, leading to re-covery of thrombin signaling independent of de novo receptorsynthesis. Interestingly, internalization of agonist-activated PAR1through clathrin-coated pits is independent of AP2, suggestingthat constitutive and agonist-induced receptor internalizationare specified by distinct molecular machinery. These findingsdemonstrate a direct role for AP2 in regulation of PAR1 sig-naling and trafficking.

Most GPCRs, including the well-characterized archetype ofthe �2-adrenergic receptor, are rapidly phosphorylated followingactivation, bind arrestins, and are then recruited to a clathrin-and dynamin-dependent pathway for internalization from theplasma membrane (33). Arrestins facilitate GPCR internaliza-tion by binding directly to the clathrin heavy chain and the�2-adaptin subunit of AP2 (8, 10). We have shown in multiplecell types that arrestins are not essential for PAR1 consti-tutive or agonist-induced internalization (4, 25). In contrast,we report here that the �2 subunit of AP2 directly binds to atyrosine-based motif in the cytoplasmic tail of PAR1 to medi-ate constitutive internalization. Mutation of PAR1 distal ty-rosine-based motif or depleting cells of AP2 by RNA interfer-ence resulted in significant inhibition of receptor constitutiveinternalization. Moreover, the PAR1 distal tyrosine-based mo-tif was also important for receptor colocalization with endog-enous AP2 in intact cells and critical for direct �2 binding invitro. The low-affinity binding of PAR1 with the �2 subunit ofAP2 as observed in our studies (KD, 7.5 �M) is typical of themoderate to weak interaction of �2 with proteins bearing ty-rosine-based motifs previously reported (22, 31) and may haveprecluded our ability to detect PAR1 interaction with AP2using cell lysate-based approaches of coimmunoprecipitationor pull-downs. Such low-affinity protein-protein interactionsare biologically relevant and, in fact, are required to allow forthe dynamic assembly and disassembly of the protein networkthat drives clathrin-coated vesicle budding at the plasma mem-brane (23). A high-affinity interaction between PAR1 and �2would be detrimental, resulting in significant internalization ofthe receptor at steady state and rendering it unavailable to itsextracellular stimuli.

In addition to PAR1, we found that several other humanGPCRs contain sequences conforming to the canonical YXXØmotif within their cytoplasmic tails (see Fig. S4 in the supple-mental material), suggesting that AP2 might function in traf-ficking of other GPCRs. Indeed, the �2 subunit of AP2 waspreviously shown to directly interact with the C tail of the�1b-adrenergic receptor (7). However, in this case, �2 binds toan unusual stretch of eight arginine residues (rather than acanonical YXXØ motif) within the C-tail of the �1b-adrenergicreceptor. This study also reported that an �lb-adrenergic re-ceptor mutant, in which the arginine stretch was deleted, failedto bind �2 in vitro, and displayed defects in agonist-inducedendocytosis, suggesting that this region (and perhaps AP2binding) is important for activated �lb-adrenergic receptor in-ternalization. The constitutively active human viral chemo-kine US28 GPCR has also been reported to internalizethrough a clathrin-dependent pathway involving AP2 andnot arrestins (9). However, whether AP2 directly binds to a

FIG. 7. The distal tyrosine-based motif is necessary for direct as-sociation between the PAR1 cytoplasmic tail and purified recombinant�2 protein. (A) His6-tagged �2 protein was expressed in E. coli byIPTG induction, and soluble, monodispersed protein was purified bysequential application of Ni-NTA resin and size exclusion chromatog-raphy. (B) Representative binding curves were generated by injecting15 �M purified His6-tagged �2 protein over immobilized PAR1 C-tailpeptide surfaces, followed by subtracting corresponding traces from ablank negative control surface. (C) Increasing concentrations of His6-tagged �2 protein (0.01 to 50 �M) were injected separately overpeptide biosensor surfaces, and binding curves were generated as de-scribed in panel B. The resulting maximal resonance units obtainedduring the analyte association phase were then plotted versus analyteconcentration to generate a one-site binding curve, indicating a disso-ciation constant (KD) of 7.5 1.3 �M for wild-type PAR1 C tail. Nodetectable binding was observed for the PAR1 A420KKA423A424

(AKKAA) mutant peptide surface, even at the saturating concen-tration of 50 �M.

3238 PAING ET AL. MOL. CELL. BIOL.

on May 6, 2016 by guest

http://mcb.asm

.org/D

ownloaded from

YXXØ motif present in the C-tail region of US28 receptorremains to be determined. Together, these findings suggestthat the clathrin adaptor AP2 functions in distinct modes ofGPCR trafficking, including constitutive internalization ofunactivated PAR1, activated �1b-adrenergic receptor inter-nalization, and uptake of the constitutively active viral che-mokine US28 receptor.

Our study also indicates that the trafficking behaviors ofunactivated versus activated PAR1 are regulated by distinctendocytic machinery. PAR1 constitutive internalization was vir-tually abolished in cells depleted of AP2, whereas agonist-acti-vated receptor internalization remained intact (Fig. 2 and 3).Activated PAR1 internalized via a clathrin- and dynamin-dependent pathway in AP2 depleted cells, excluding the pos-sibility that the receptor uses an alternate non-clathrin endo-cytic route (see Fig. S3 in the supplemental material). Ourprevious studies suggest that internalization of agonist-acti-vated PAR1 is critically dependent on phosphorylation of the

C-tail region (25, 32). Activated PAR1 phosphorylation mightinduce a conformational change within the C-tail region toexpose an endocytic-sorting motif and/or to promote bindingof clathrin adaptor proteins. We previously showed that mu-tation of the proximal tyrosine-based motif partially inhibitsthe endocytosis of activated PAR1 that is only evident afterprolonged agonist exposure (26). However, AP2 does not ap-pear to be involved in this process since, in the current studies,we find that AP2 is not essential for activated PAR1 internal-ization (Fig. 2 and 3). Thus, whether other clathrin adaptorproteins such as epsin or CALM bind directly to this motif orelsewhere to PAR1 intracytosolic domains to regulate AP2-independent internalization of activated PAR1 through clath-rin-coated pits is not known. Internalization of activated PAR1may be additionally dependent on ubiquitination or deubiq-uitination processes. The transient modification of GPCRsand/or associated proteins by ubiquitination is known to facil-itate trafficking through the endocytic pathway (14).

FIG. 8. PAR1 mutants defective in constitutive internalization fail to regain thrombin responsiveness. HeLa cells stably expressing similaramounts of wild-type (WT) PAR1 or receptor mutants grown on coverslips were pretreated with 10 �M cycloheximide for 90 min to block de novoprotein synthesis, loaded with Fura2-AM, and then stimulated with thrombin (�-Th) as indicated. The Ca2� responses (i.e., [Ca2�]i, intracellularCa2� concentration) were quantified as described in Materials and Methods. Cells were first exposed to 30 nM thrombin for 10 min at 37°C, andthe traces shown represent Ca2� responses of at least 10 cells from a single field, all of which responded to thrombin. The presence of agonistsand time of exposure are indicated by the bars below and above the traces. Between stimulations, cells were washed with medium containing thethrombin inhibitor hirudin (0.5 U/ml) and allowed to recover for 20 min at 37°C. (A and B) Wild-type (WT) PAR1 and PAR1 A383SIA386

mutant-expressing cells responded to a second 10 nM thrombin stimulus as well as to 2 �M A23187, a calcium ionophore. Similar results wereobserved in three independent experiments. (C to F) HeLa cells expressing the PAR1 A420KKA423A424 mutant failed to elicit a second responseto 10 nM thrombin but retained the capacity to respond to 500 �M carbachol (CARB), an agonist for endogenous muscarinic acetylcholineGPCRs, or to the calcium ionophore A23187 (2 �M). Similar findings were observed in three separate experiments. ASIA, A383SIA386; AKKAA,A420KKA423A424.

VOL. 26, 2006 AP2 DIRECTLY REGULATES PAR1 SIGNALING AND TRAFFICKING 3239

on May 6, 2016 by guest

http://mcb.asm

.org/D

ownloaded from

The proteolytic activation of PAR1 is unique amongGPCRs, and distinct intracellular pathways have evolved todispose of irreversibly activated receptors and to replenish thecell surface with uncleaved receptors after thrombin exposure.

Our findings strongly suggest that AP2-dependent constitutiveinternalization of PAR1 is critical for resensitizing cells tothrombin signaling. In human platelets, which presumably re-spond to thrombin only once, the majority of PAR1 is retained

FIG. 9. AP2 is essential for restoring thrombin signaling in HUVECs and other cell types. PAR1-expressing HeLa cells were transiently transfectedwith 50 nM �2 or NS siRNA, and HUVECs were electroporated with 600 nM �2 or nonspecific (ns) siRNAs as described in Materials and Methods.Cells were grown on coverslips, pretreated with 10 �M cycloheximide, loaded with Fura2-AM, and then exposed to thrombin (�-Th) as indicated. Theintracellular Ca2� response traces from individual cells are shown in various panels, whereas the responses from an average of 20 to 30 cells imaged fromthree separate experiments are shown as bar graphs in panels C, D, G, and H. Cells were first exposed to 30 nM thrombin for 10 min at 37°C, and �80%of these cells responded to thrombin. Cells were washed with medium containing hirudin (0.5 U/ml) and then exposed to 10 nM thrombin again. Incontrol ns siRNA-treated cells, thrombin elicited a second Ca2� response in virtually the same percentage of cells that were initially responsive tothrombin. In �2 siRNA-treated cells, only 1 to 2 cells out of �20 cells were responsive to thrombin after an initial thrombin exposure (panels C and G).The difference between the number of cells eliciting �-thrombin-induced calcium response in �2 siRNA-treated cells compared to ns siRNA control-treated cells was significant (��, P � 0.01; �, P � 0.05) as determined using an unpaired Student’s t test. HeLa cells and HUVECs retained the capacityto respond to 500 �M carbachol (CARB) or 100 �M UTP, agonists for endogenously expressed muscarinic and purinergic GPCRs, respectively (panelsD and H), and to the calcium ionophore A23187 (2 �M). Similar results were observed in at least three independent experiments.

3240 PAING ET AL. MOL. CELL. BIOL.

on May 6, 2016 by guest

http://mcb.asm

.org/D

ownloaded from

on the cell surface and fails to internalize (19). Thus, plateletslack an internal pool of protected receptors as recovery ofthrombin responsiveness is not a physiological requirement forthis cell type. In contrast, endothelial cells and fibroblasts areexposed to thrombin repeatedly and need to recover thrombinsignaling in a timely manner. This appears to be accomplishedby the movement of uncleaved PAR1 from an intracellularcompartment to the cell surface to permit rapid recovery ofthrombin signaling independent of de novo receptor synthesis(13). Evidence presented here supports this model. In cellsexpressing PAR1 tyrosine mutants or in cells depleted of AP2by RNA interference, uncleaved receptor fails to constitutivelyinternalize and accumulate in an intracellular store. Conse-quently, PAR1 is not protected from thrombin cleavage andcells are unable to regain thrombin responsiveness. However,cells retained the capacity to elicit calcium responses to otherendogenous GPCR agonists, indicating that receptor activa-tion is not globally disrupted. Several other GPCRs includingPAR2 and the thromboxane A2 receptor � isoform have alsobeen reported to constitutively internalize, resulting in an in-tracellular store of unactivated receptors (2, 28). A noncanoni-cal YX3Ø motif is present in the cytoplasmic tail of thrombox-ane A2 receptor (29), whereas PAR2 lacks such tyrosine-basedsequences. The mechanism mediating constitutive internaliza-tion of these receptors as well as its physiological relevanceremains poorly defined.

Our studies provide the first insight into the molecularmechanisms responsible for PAR1 constitutive internalizationand cellular resensitization to thrombin signaling. The proteo-lytic nature of PAR1 activation is distinct from most reversiblyactivated GPCRs. Consequently, endothelial cells and othercell types, which need to respond to thrombin repeatedly overtime, have evolved specialized mechanisms to permit rapidrecovery or maintenance of thrombin responsiveness. Throm-bin and PAR1 are important mediators of endothelial cellactivation and migration during vascular development and inresponse to vascular injury (11, 42). Cell migration requiresrepeated sampling of the local environment in order to detectchanges in agonist concentrations and gradients. It will beimportant to determine whether disruption of PAR1 toniccycling and failure of endothelial cells to rapidly recoverthrombin signaling are critical for endothelial cell activationand/or migration in vivo.

ACKNOWLEDGMENTS

We thank T. K. Harden and R. A. Nicholas for comments andhelpful discussions and David J. Owen and Robert J. Lefkowitz forgenerously providing reagents.

This work was supported by National Institutes of Health grantsHL067697 and HL073328 (to J.T.) and GM065533 (to D.P.S.). M.M.P.and C.A.J. were each supported by American Heart Association Pre-doctoral Fellowships.

REFERENCES

1. Ahn, S., S. K. Shenoy, H. Wei, and R. J. Lefkowitz. 2004. Differential kineticand spatial patterns of �-arrestin and G protein-mediated ERK activation bythe angiotensin II receptor. J. Biol. Chem. 279:35518–35525.

2. Bohm, S. K., L. M. Khitin, E. F. Grady, G. Aponte, D. G. Payan, and N. W.Bunnett. 1996. Mechanisms of desensitization and resensitization of protein-ase-activated receptor-2. J. Biol. Chem. 271:22003–22016.

3. Bonifacino, J. S., and L. M. Traub. 2003. Signals for sorting of transmem-brane proteins to endosomes and lysosomes. Annu. Rev. Biochem. 72:395–447.

4. Chen, C. H., M. M. Paing, and J. Trejo. 2004. Termination of protease-activated receptor-1 signaling by �-arrestins is independent of receptor phos-phorylation. J. Biol. Chem. 279:10020–10031.

5. Chen, J., M. Ishii, L. Wang, K. Ishii, and S. R. Coughlin. 1994. Thrombinreceptor activation: confirmation of the intramolecular tethered ligandinghypothesis and discovery of an alternative intermolecular liganding mode.J. Biol. Chem. 269:16041–16045.

6. Coughlin, S. R. 2000. Thrombin signalling and protease-activated receptors.Nature 407:258–264.

7. Diviani, D., A.-L. Lattion, L. Abuin, O. Staub, and S. Cotecchia. 2003. Theadaptor complex 2 directly interacts with the �1b-adrenergic receptor andplays a role in receptor endocytosis. J. Biol. Chem. 278:19331–19340.

8. Ferguson, S. S., W. r. Downey, A. M. Colapietro, L. S. Barak, L. Menard, andM. G. Caron. 1996. Role of �-arrestin in mediating agonist-promoted Gprotein-coupled receptor internalization. Science 271:363–366.

9. Fraile-Ramos, A., T. A. Kohout, M. Waldhoer, and M. Marsh. 2003. Endo-cytosis of the viral chemokine receptor US28 does not require �-arrestins butis dependent on the clathrin-mediated pathway. Traffic 4:243–253.

10. Goodman, O. B., Jr., J. G. Krupnick, F. Santini, V. V. Gurevich, R. B. Penn,A. W. Gagnon, J. H. Keen, and J. L. Benovic. 1996. �-Arrestin acts as aclathrin adaptor in endocytosis of the beta2-adrenergic receptor. Nature383:447–450.

11. Griffin, C. T., Y. Srinivasan, Y.-W. Zheng, W. Huang, and S. R. Coughlin.2001. A role for thrombin receptor signaling in endothelial cells duringembryonic development. Science 293:1666–1670.

12. Grynkiewicz, G., M. Poenie, and R. Y. Tsien. 1985. A new generation of Ca2�

indicators with greatly improved fluorescence properties. J. Biol. Chem.260:3440–3450.

13. Hein, L., K. Ishii, S. R. Coughlin, and B. K. Kobilka. 1994. Intracellulartargeting and trafficking of thrombin receptors: a novel mechanism for re-sensitization of a G protein-coupled receptor. J. Biol. Chem. 269:27719–27726.

14. Hicke, L., and R. Dunn. 2003. Regulation of membrane protein transport byubiquitin and ubiquitin-binding proteins. Annu. Rev. Cell Dev. Biol. 19:141–172.

15. Horvat, R., and G. E. Palade. 1995. The functional thrombin receptor isassociated with the plasmalemma and a large endosomal network in culturedhuman umbilical vein endothelial cells. J. Cell Sci. 108:1155–1164.

16. Hoxie, J. A., M. Ahuja, E. Belmonte, S. Pizarro, R. Parton, and L. F. Brass.1993. Internalization and recycling of activated thrombin receptors. J. Biol.Chem. 268:13756–13763.

17. Hung, D. T., T. K. Vu, V. I. Wheaton, K. Ishii, and S. R. Coughlin. 1992.Cloned platelet thrombin receptor is necessary for thrombin-induced plate-let activation. J. Clin. Investig. 89:1350–1353.

18. Ishii, K., L. Hein, B. Kobilka, and S. R. Coughlin. 1993. Kinetics of thrombinreceptor cleavage on intact cells: relation to signaling. J. Biol. Chem. 268:9780–9786.

19. Molino, M., D. F. Bainton, J. A. Hoxie, S. R. Coughlin, and L. F. Brass. 1997.Thrombin receptors on human platelets. Initial localization and subsequentredistribution during platelet activation. J. Biol. Chem. 272:6011–6017.

20. Motley, A., N. A. Bright, M. N. J. Seaman, and M. Robinson. 2003. Clathrin-mediated endocytosis in AP-2 depleted cells. J. Cell Biol. 162:909–918.

21. O’Brien, P. J., M. Molino, M. Kahn, and L. F. Brass. 2001. Protease acti-vated receptors: theme and variations. Oncogene 20:1570–1581.

22. Ohno, H., J. Stewart, M. Fournier, H. Bosshart, I. Rhee, S. Miyatake, T.Saito, A. Gallusser, Y. Kirchhausen, and J. Bonifacino. 1995. Interactions oftyrosine-based sorting signals with clathrin-associated proteins. Science 269:1872–1875.

23. Owen, D. J., B. M. Collins, and P. R. Evans. 2004. Adaptors for clathrincoats: structure and function. Annu. Rev. Cell Dev. Biol. 20:153–191.

24. Owen, D. J., and P. R. Evans. 1998. A structural explanation for the recog-nition of tyrosine-based endocytic signals. Science 282:1327–1332.

25. Paing, M. M., A. B. Stutts, T. A. Kohout, R. J. Lefkowitz, and J. Trejo. 2002.�-Arrestins regulate protease-activated receptor-1 desensitization but notinternalization or down-regulation. J. Biol. Chem. 277:1292–1300.

26. Paing, M. M., B. R. S. Temple, and J. Trejo. 2004. A tyrosine-based sortingsignal regulates intracellular trafficking of protease-activated receptor-1:multiple regulatory mechanisms for agonist-induced G protein-coupled re-ceptor internalization. J. Biol. Chem. 279:21938–21947.

27. Palmer, R. K., J. L. Boyer, J. B. Schachter, R. A. Nicholas, and T. K. Harden.1998. Agonist action of adenosine triphosphates at the human P2Y1 recep-tor. Mol. Pharm. 54:1118–1123.

28. Parent, J. L., P. Labrecque, M. J. Orsini, and J. L. Benovic. 1999. Internalizationof the TXA2 receptor � and � isoforms. J. Biol. Chem. 274:8941–8948.

29. Parent, J. L., P. Lebrecque, M. D. Rochdi, and J. L. Benovic. 2001. Role ofthe differentially spliced carboxyl terminus in thromboxane A2 receptortrafficking. J. Biol. Chem. 276:7079–7085.

30. Pierce, K. L., R. T. Premont, and R. J. Lefkowitz. 2002. Seven-transmem-brane receptors. Nat. Rev. Mol. Cell. Biol. 3:639–650.

31. Rapoport, I., M. Miyazaki, W. Boll, B. Duckworth, L. C. Cantley, S. Shoelson,and T. Kirchhausen. 1997. Regulatory interactions in the recognition ofendocytic sorting signals by AP-2 complexes. EMBO J. 16:2240–2250.

VOL. 26, 2006 AP2 DIRECTLY REGULATES PAR1 SIGNALING AND TRAFFICKING 3241

on May 6, 2016 by guest

http://mcb.asm

.org/D

ownloaded from

32. Shapiro, M. J., J. Trejo, D. Zeng, and S. R. Coughlin. 1996. Role of thethrombin receptor’s cytoplasmic tail in intracellular trafficking. Distinct de-terminants for agonist-triggered versus tonic internalization and intracellularlocalization. J. Biol. Chem. 271:32874–32880.

33. Shenoy, S. K., and R. J. Lefkowitz. 2003. Multifaceted role of �-arrestins inthe regulation of seven-membrane-spanning receptor trafficking and signal-ing. Biochem. J. 375:503–515.

34. Tohgo, A., E. W. Choy, D. Getsy-Palmer, K. L. Pierce, S. A. Laporte, R. H.Oakley, M. G. Caron, R. J. Lefkowitz, and L. M. Luttrell. 2003. The stability ofthe G protein-coupled receptor-�-arrestin interaction determines the mecha-nism and functional consequence of ERK activation. J. Biol. Chem. 278:6258–6267.

35. Trejo, J., Y. Altschuler, H.-W. Fu, K. E. Mostov, and S. R. Coughlin. 2000.Protease-activated receptor downregulation: a mutant HeLa cell line sug-gests novel requirements for PAR1 phosphorylation and recruitment toclathrin-coated pits. J. Biol. Chem. 275:31255–31265.

36. Trejo, J., and S. R. Coughlin. 1999. The cytoplasmic tails of protease-activated receptor-1 and substance P receptor specify sorting to lysosomesversus recycling. J. Biol. Chem. 274:2216–2224.

37. Trejo, J., S. R. Hammes, and S. R. Coughlin. 1998. Termination of signalingby protease-activated receptor-1 is linked to lysosomal sorting. Proc. Natl.Acad. Sci. USA 95:13698–13702.

38. Vu, T.-K. H., D. T. Hung, V. I. Wheaton, and S. R. Coughlin. 1991. Molecularcloning of a functional thrombin receptor reveals a novel proteolytic mech-anism of receptor activation. Cell 64:1057–1068.

39. Vu, T.-K. H., V. I. Wheaton, D. T. Hung, and S. R. Coughlin. 1991. Domainsspecifying thrombin-receptor interaction. Nature 353:674–677.

40. Wang, Y., Y. Zhou, K. Szabo, C. R. Haft, and J. Trejo. 2002. Down-regulationof protease-activated receptor-1 is regulated by sorting nexin 1. Mol. Biol.Cell 13:1965–1976.

41. Willard, F. S., A. J. Kimple, C. A. Johnston, and D. P. Siderovski. 2005. Adirect fluorescence-based assay for RGS domain GTPase accelerating activ-ity. Anal. Biochem. 340:341–351.

42. Woolkalis, M. J., T. J. DeMelfi, N. Blanchard, J. A. Hoxie, and L. F. Brass.1995. Regulation of thrombin receptors on human umbilical vein endothelialcells. J. Biol. Chem. 270:9868–9875.

3242 PAING ET AL. MOL. CELL. BIOL.

on May 6, 2016 by guest

http://mcb.asm

.org/D

ownloaded from