Heterologous Inhibition of G Protein-coupled Receptor Endocytosis Mediated by Receptor-specific Trafficking of beta -Arrestins*

Uwe KleinDagger, Claudia Müller, Peter Chu, Mariel Birnbaumer§, and Mark von Zastrow

From the Department of Psychiatry and Department of Cellular and Molecular Pharmacology, Program in Cell Biology, University of California, San Francisco, California 94143-0984 and § Department of Anesthesiology, University of California, Los Angeles, California 90024

Received for publication, October 10, 2000, and in revised form, February 1, 2001


    ABSTRACT
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS AND DISCUSSION
REFERENCES

We have observed an unexpected type of nonreciprocal "cross-regulation" of the agonist-induced endocytosis of G protein-coupled receptors by clathrin-coated pits. Isoproterenol-dependent internalization of beta 2-adrenergic receptors in stably transfected HEK293 cells was specifically blocked (>65% inhibition) by vasopressin-induced activation of V2 vasopressin receptors co-expressed at similar levels. In contrast, activation of beta 2 receptors caused no detectable effect on V2 receptor internalization in the same cells. Several pieces of evidence suggest that this nonreciprocal inhibition of endocytosis is mediated by receptor-specific intracellular trafficking of beta -arrestins. First, previous studies showed that the activation of V2 but not beta 2 receptors caused pronounced recruitment of beta -arrestins to endocytic membranes (Oakley, R. H., Laporte, S. A., Holt, J. A., Barak, L. S., and Caron, M. G. (1999) J. Biol. Chem. 274, 32248-32257). Second, overexpression of arrestin 2 or 3 (beta -arrestin 1 or 2) abolished the V2 receptor-mediated inhibition of beta 2 receptor internalization. Third, mutations of the V2 receptor that block endomembrane recruitment of beta -arrestins eliminated the V2 receptor-dependent blockade of beta 2 receptor internalization. These results identify a novel type of heterologous regulation of G protein-coupled receptors, define a new functional role of receptor-specific intracellular trafficking of beta -arrestins, and suggest an experimental method to rapidly modulate the functional activity of beta -arrestins in intact cells.


    INTRODUCTION
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS AND DISCUSSION
REFERENCES

G protein-coupled receptors (GPCRs)1 are regulated by a set of highly conserved molecular mechanisms (1-3). Because most cells express multiple types of GPCR that serve distinct functions, the specificity of these regulatory mechanisms is of great physiological interest. Extensive studies have led to the classification of distinct "heterologous" and "homologous" mechanisms of GPCR regulation. An example of a heterologous regulatory mechanism is rapid desensitization of the beta 2-adrenergic receptor (beta 2AR) mediated by the cyclic AMP-dependent protein kinase. Activated cAMP-dependent protein kinase can phosphorylate both the beta 2AR as well as a number of other GPCRs irrespective of whether or not these receptors have bound agonist. A well characterized mechanism of homologous regulation is desensitization of the beta 2AR mediated by G protein-coupled receptor kinases. In general G protein-coupled receptor kinases preferentially phosphorylate ligand-activated receptors without affecting other GPCRs present in the same cells that are not activated by their respective agonist (4-6).

Many GPCRs, such as the prototypic beta 2AR, are regulated by agonist-induced endocytosis via clathrin-coated pits (7-10). This process is promoted by G protein-coupled receptor kinase-mediated phosphorylation of receptors followed by membrane recruitment of nonvisual arrestins (or beta -arrestins), which link receptors to the clathrin/AP-2 endocytic coat (9, 11, 12). Because high affinity interaction of GPCRs with arrestins is promoted by both G protein-coupled receptor kinase-mediated phosphorylation and an agonist-induced conformation of the receptor protein (13, 14), endocytosis by clathrin-coated pits is thought to represent a highly homologous mechanism of GPCR regulation (8, 11). Consistent with this, distinct GPCRs, even when co-expressed at high levels, are endocytosed by coated pits in a highly selective manner after activation by their respective agonist (15).

Arrestins play a highly conserved role in promoting endocytosis of various GPCRs by clathrin-coated pits (11). Recent studies indicate that particular GPCRs differ substantially in their effects on the intracellular trafficking of arrestins. Many G protein-coupled receptors (including the beta 2AR) recruit arrestins to the plasma membrane but do not remain associated with arrestins after endocytosis (16, 17). In contrast, the V2 vasopressin receptor (V2R) has been shown to mediate recruitment of arrestins both to the plasma membrane and to V2R-containing endocytic vesicles (17). This receptor-specific difference in the endomembrane recruitment of arrestins is mediated by a persistent phosphorylation of internalized V2Rs, which inhibits recycling of receptors to the plasma membrane and causes a prolonged state of receptor desensitization (17, 18). Endosome-associated arrestins have been proposed to play an important role in determining the specificity of downstream signal transduction by endocytosed GPCRs (19, 20). However, to our knowledge no previous studies have examined the possibility that arrestin trafficking may modulate the specificity of GPCR endocytosis itself. In the present study we have observed an unexpected type of heterologous and nonreciprocal inhibition of beta 2AR endocytosis mediated by agonist-induced activation of the V2R. This heterologous inhibition is dependent on endomembrane recruitment of beta -arrestins, suggesting a novel role of receptor-specific trafficking of arrestins in modulating endocytosis of certain GPCRs.

    EXPERIMENTAL PROCEDURES
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS AND DISCUSSION
REFERENCES

cDNA Constructs and Mutagenesis-- The coding sequence of the human V2R was amplified by PCR and inserted into a tagging vector containing the influenza hemagglutinin signal sequence and the FLAG epitope (21). The N-terminal epitope-tagged receptor sequence was subcloned into the mammalian expression vector pcDNA3.1 (Invitrogen). The human beta 2AR and the murine delta -opioid receptor were tagged in the N-terminal extracellular domains with the HA epitope (YPYDVPDYA) and subcloned into pcDNA3.1, as previously described (22). Two truncated forms of the V2R were obtained through oligonucleotide-directed mutagenesis using the polymerase chain reaction with 3' antisense primers incorporating a stop codon followed by a suitable restriction site (345T and 362T: stop codons substituted for residues at positions 345 and 362 of the human V2R amino acid sequence, respectively) (18). PCR products were subcloned into pcDNA3.1. GFP-tagged arrestin 3 (beta -arrestin 2) was a generous gift of Dr. Marc Caron. A C-terminally EE epitope-tagged version of arrestin 2 (beta -arrestin-1, a generous gift of Dr. Jeffrey Benovic) was constructed by ligating a synthetic adaptor oligonucleotide encoding the epitope tag sequence and cloning into pcDNA3. All constructs were verified by DNA sequencing (University of California San Francisco Biomolecular Resource Center).

Cell Lines and Transfection-- HEK293 cells (ATCC) were cultured in Dulbecco's modified Eagle's medium supplemented with 10% fetal calf serum, 100 units/ml penicillin, and 0.1 mg/ml streptomycin (University of California San Francisco Cell Culture Facility) in a humidified incubator with 7% CO2 at 37 °C. Cells were transiently transfected using calcium phosphate co-precipitation (23). For generation of stable transformants, cells after transfection were selected for resistance to 0.5 mg/ml G418 (Geneticin, Life Technologies, Inc.). Single clones were isolated after 2-3 weeks of culture and screened for expression by immunofluorescence microscopy and radioligand binding assays.

Immunofluorescence Microscopy-- Cells were grown on glass coverslips in 6-well tissue culture plates. For co-localization of FLAG-tagged V2R and HA-tagged beta 2AR or HA-tagged delta -opioid receptor, cells were incubated with the monoclonal anti-epitope antibodies anti-FLAG M1 (Sigma) and anti-HA HA.11 (Covance), respectively, both at a 1:500 dilution for 30 min at 37 °C. Cells were then treated under the respective conditions with agonists, fixed using 4% formaldehyde in phosphate-buffered saline, and permeabilized with blocking buffer (3% nonfat dry milk, 0.1% Triton X-100, 50 mM Tris/HCl, pH 7.5, 1 mM CaCl2). Bound M1 antibody was detected using incubations with a rabbit anti-mouse IgG2b subtype-specific linker antibody (1:800 dilution, 45 min at room temperature) followed by a Texas Red-conjugated donkey anti-rabbit antibody (1:1000 dilution, 20 min at room temperature). Bound HA.11 antibody was detected using fluorescein isothiocyanate-conjugated goat anti-mouse IgG1 subtype-specific antibody (1:800 dilution, 20 min at room temperature). Immuno-stained coverslips were mounted on microscopy slides and examined by epifluorescence or confocal microscopy (see below).

Co-localization of beta -Arrestin with Vasopressin V2 Receptor and beta 2-Adrenergic Receptor-- Cells stably co-expressing FLAG epitope-tagged V2R and HA epitope-tagged beta 2AR were transiently transfected with an expression plasmid encoding GFP-tagged beta -arrestins as described previously (24). Cells were incubated with the monoclonal anti-FLAG M1 antibody at a 1:500 dilution for 30 min at 37 °C. Cells were treated and fixed as mentioned above. FLAG epitope-tagged V2R was detected using a 6-((7-amino-4-methylcoumarin-3-acetyl)amino) hexanoic acid, succinimidyl ester (AMCA-S)-conjugated goat anti-mouse secondary antibody (Molecular Probes, OR, diluted 1:250, blue fluorescence). The beta 2AR was detected by staining with a rabbit polyclonal antibody raised against the C-terminal 15-amino acid residues of the human beta 2AR (22) diluted 1:500 and a Texas-Red conjugated anti-rabbit secondary antibody (Jackson Immunoresearch Laboratories, diluted 1:500, red fluorescence). beta -Arrestin 2-GFP was detected by GFP fluorescence (green fluorescence). Immuno-stained coverslips were examined by epifluorescence microscopy using a Nikon 60× NA1.4 objective and Chroma filter sets optimized for these fluorophores. Confocal microscopy was performed using a Bio-Rad MRC1000 and a Zeiss 100X NA1.3 objective. In all experiments, minimal bleed-through between channels was confirmed using single-labeled control specimens.

Membrane Preparation-- Individual cell clones stably expressing the beta 2AR and/or the V2R were grown on 10-cm dishes to confluency. Cells were lifted in phosphate-buffered saline containing 2 mM EDTA and centrifuged at 500 × g for 10 min. Pellets were resuspended in 1 ml of lysis buffer (5 mM Hepes/NaOH, pH 7.4, 5 mM EDTA, 5 mM EGTA, 0.5 mM Pefabloc SC) and homogenized using a glass Dounce homogenizer. The suspension was centrifuged for 15 min at 20,000 × g, and the pellets were resuspended and homogenized as above. The final pellet was resuspended in 200 µl of membrane buffer (20 mM Hepes/NaOH, pH 7.4, 1 mM EDTA, 5 mM MgCl2, 0.5 mM Pefabloc SC). Protein concentration was estimated by the method of Bradford (25). Membranes were stored frozen at -80 °C.

Radioligand Binding Assays-- To assess expression levels for the V2R, 10 µg of membrane protein were incubated in 125 µl of V2 binding buffer (50 mM Hepes/NaOH, pH 7.4, 5 mM MgCl2, 0.2% bovine serum albumin) with 10 nM [3H-Arg8]vasopressin ([3H]AVP) (PerkinElmer Life Sciences, 68.5 Ci/mmol) for 30 min at 30 °C. Nonspecific binding was determined in the presence of 4 µM nonlabeled AVP. The binding reaction was stopped by rapid filtration over GF/C glass-fiber filters (Whatman) using a Brandel cell harvester. Filters were washed 3 times with ice-cold V2 filtration buffer (20 mM Hepes/NaOH, pH 7.4, 2 mM MgCl2, 0.02% bovine serum albumin). For the beta 2AR binding assay, 10 µg of membrane protein were incubated in 500 µl of beta 2 binding buffer (75 mM Tris/HCl, pH 7.4, 12.5 mM MgCl2, 1 mM EDTA, 0.2% bovine serum albumin) with 10 nM [3H]dihydroalprenolol (Amersham Pharmacia Biotech, 88 Ci/mmol) for 90 min at 25 °C. Nonspecific binding was determined in the presence of 10 µM nonlabeled alprenolol. The binding reaction was stopped by rapid filtration over GF/C glass-fiber filters (Whatman). Filters were washed 3 times with ice-cold beta 2 filtration buffer (75 mM Tris/HCl, pH 7.4, 12.5 mM MgCl2, 1 mM EDTA). Filter-bound radioactivity was counted by liquid scintillation. All tests were performed in triplicate.

Scoring of Receptor Localization by Fluorescence Microscopy-- Coverslips were processed for indirect immunofluorescence staining, as above, and coded such that the identity and treatment conditions of the specimen were not specified. Examination of coded specimens by epifluorescence microscopy was performed by a second individual not familiar with the details of the experiment. The localization of fluorochromes representing individual receptors were classified in multiple cells examined at random, positive for expression of both receptors. Cells with detectable expression of only one of the two receptors were excluded from the scoring. Immunostaining was classified according to the following criteria: non-internalized localization (bright staining around the cell periphery with <10 immunoreactive puncta visualized within the cytoplasm); intermediate appearance (10-20 internal puncta); internalized (>20 immunoreactive puncta resolved within the cytoplasm). At least 25 cells per coverslip were scored by these criteria, and all specimens from an individual experiment were scored in a single session before the code was broken and results were tabulated. For studies employing GFP-tagged arrestins, receptor localization for GFP positive and negative cells were scored in the same sitting, and the specificity of observed effects was confirmed by transfection of GFP not fused to beta -arrestin. Numbers in figures represent the mean ± S.D. for results from a representative experiment, and all reported experiments were performed independently at least three times with similar results.

Estimation of Receptor Internalization by Fluorescence Flow Cytometry-- A previously established (26) flow cytometric assay was used to quantitate immunoreactive receptors present on the surface of intact cells after dissociation from the cell culture dish. All data points represent quantitation of 20,000 cells using a FACScan cytometer (Becton Dickenson) performed in triplicate (representing three independently treated dishes) for each experiment. The extent of receptor internalization was calculated according to the agonist-induced reduction in mean surface immunoreactivity (26). Figures indicate mean ± S.D. for results compiled from three separate experiments.

    RESULTS AND DISCUSSION
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS AND DISCUSSION
REFERENCES

We chose to examine membrane trafficking of the beta 2AR and V2R in HEK293 cells because both receptors stimulate adenylyl cyclase activity via the Gs-signaling pathway and undergo agonist-induced endocytosis by an arrestin-dependent mechanism mediated by clathrin-coated pits, yet these receptors are well established to differ substantially in their effects on the intracellular trafficking of beta -arrestins in this cell type (17). A functional HA-tagged beta 2AR and FLAG-tagged V2R were co-expressed in HEK293 cells by stable transfection. Because a single selection marker (neomycin) was used to isolate stably transfected cells after transfection of separate plasmids encoding the beta 2AR and V2R, many cell populations obtained by this method expressed either the beta 2AR or V2R but not both. Dual label fluorescence microscopy was used to identify stably transfected cell populations that expressed both the beta 2AR and V2R in the majority (~70%) of cells. A cell clone expressing both receptors at moderate levels (3.8 ± 0.3 pmol/mg for the V2R and 4.7 ± 0.2 pmol/mg for the beta 2AR, respectively) was selected from this group for further analysis because these levels of expression are comparable to those used previously for studies of GPCR endocytosis and arrestin trafficking in HEK293 cells (16, 17, 22).

Both the beta 2AR and V2R were visualized by fluorescence microscopy in the plasma membrane of cells incubated in the absence of agonist (Fig. 1, panels a and b). In the presence of saturating concentrations (10 µM) of the beta 2AR agonist isoproterenol (ISO), beta 2AR redistributed from the plasma membrane to endocytic vesicles within 15 min, whereas co-expressed V2Rs remained in the plasma membrane and did not exhibit any detectable internalization (Fig. 1, panels c and d). Conversely, in the presence of saturating concentrations (10 µM) of the V2R agonist AVP, V2Rs were selectively endocytosed without any detectable internalization of co-expressed beta 2ARs (Fig. 1, panels e and f). Agonist-induced endocytosis of the beta 2AR in HEK293 cells is mediated by clathrin-coated pits, as indicated by morphological studies and the effects of biochemical and genetic inhibitors of clathrin-coated pit function (7-9, 22). Recent studies indicate that agonist-induced endocytosis of the V2R is also mediated by clathrin-coated pits (17, 27). Consistent with this, we confirmed that AVP-induced internalization of the FLAG-tagged V2R was blocked by mild hypertonicity (0.45 M sucrose) and was also specifically inhibited by overexpression of K44E (dominant-negative) mutant dynamin (not shown).


View larger version (81K):
[in this window]
[in a new window]
 
Fig. 1.   Cellular trafficking of the V2R and beta 2AR upon agonist treatment. HEK293 cells stably expressing both FLAG epitope-tagged V2R and HA epitope-tagged beta 2AR at equal levels were treated for 20 min at 37 °C with 10 µM either no agonist (a and b), ISO alone (c and d), AVP alone (e and f), or both ISO and AVP (g and h). The distribution of both receptors after treatment was visualized by immunofluorescence microscopy. The upper panels show the localization of the V2R, and the lower panels show the localization of the beta 2AR in the same field. Black arrows in panels g and h indicate a cell co-expressing both receptors, and white arrows indicate a cell expressing only the beta 2AR without any detectable V2R.

In contrast to the ability of the beta 2AR or V2R to endocytose selectively when activated separately by ISO or AVP, respectively, surprising results were observed in cells exposed to a saturating concentration (10 µM) of both ISO and AVP. Under these conditions, pronounced internalization of the V2R was still observed, but co-expressed beta 2ARs remained in the plasma membrane and appeared to be completely resistant to agonist-induced internalization (Fig. 1, panels g and h, the black arrow indicates an example of a cell co-expressing both receptors). Indeed, the only cells in which beta 2AR internalization was observed under these conditions were the minority of cells in the transfected population (~10-20% of total, depending on the cell line) that expressed only beta 2AR without any detectable V2R (white arrow in Fig. 1, panels g and h, indicates an example of such a cell). Quantitation of multiple cells (selected at random in coded specimens, see "Experimental Procedures") confirmed these observations (Fig. 2A). Similar results were also observed in transiently transfected cells that vary more widely in beta 2AR and V2R expression levels (see below). Transient transfection of Chinese hamster ovary cells yielded comparable results, demonstrating that the inhibition can be observed in more than one cell type (not shown).


View larger version (20K):
[in this window]
[in a new window]
 
Fig. 2.   Quantitation of agonist-induced internalization of the V2R and beta 2AR. HEK293 cells stably expressing both FLAG epitope-tagged V2R and HA epitope-tagged beta 2AR at equal levels were treated for 20 min at 37 °C either with no agonist or 10 µM ISO alone, AVP alone, or both ISO and AVP. A, internalization for both receptors was estimated by scoring receptor localization after visualization by immunofluorescence microscopy (see "Experimental Procedures"). Only cells with detectable expression of both receptors were included in the scoring. B, quantitation of receptor internalization by fluorescence flow cytometry. In the presence of ISO alone, cells show 60 ± 1% loss of cell surface immunoreactivity (internalization) for the beta 2AR. In the presence of AVP alone, the V2R shows 29 ± 3% internalization. Co-stimulation with both AVP and ISO reduces the extent of beta 2AR internalization by ~65% to 21 ± 3%, whereas V2R internalization remains unaffected (33 ± 3%). The statistical significance of the AVP-induced inhibition of beta 2AR internalization was confirmed using Student's t test (p < 0.001).

The heterologous inhibition of beta 2AR internalization caused by activation of co-expressed V2Rs was further confirmed using an established flow cytometric assay (26) applied to stably transfected cells expressing both receptors at closely similar levels (~4 pmol/mg), as assessed by radioligand binding assays (see "Experimental Procedures"). Consistent with observations made by fluorescence microscopy, AVP and ISO specifically promoted internalization of the V2R and beta 2AR, respectively, in a strictly homologous manner when added separately to the culture medium (Fig. 2B, first, second, fourth, and fifth bars). In contrast, in cells exposed to both AVP and ISO, ISO-induced activation of the beta 2AR was markedly and specifically inhibited (Fig. 2B, third and sixth bars). The magnitude of AVP-induced inhibition of beta 2AR internalization measured by this assay was ~65% and was statistically highly significant (Fig. 2B legend), whereas ISO-induced activation of the beta 2AR caused no detectable effect on AVP-induced internalization of the V2R. Furthermore, the extent of V2R-mediated inhibition of beta 2AR internalization measured in these experiments was underestimated because of the presence of a subpopulation of stably transfected cells expressing beta 2AR without detectable V2R (~15% of total), in which no AVP-induced inhibition of beta 2AR internalization is observed (Fig. 1, panels g and h). A similar extent of inhibition of beta 2AR internalization was observed 60 min after incubation of co-transfected cells with ISO and AVP (not shown). When stimulated separately, the extent of internalization of both receptors reached steady state within 15 min after the addition of agonist (AVP or ISO, respectively). Taken together, these observations suggest that AVP-induced activation of the V2R in these cells causes a nearly complete blockade of the internalization of co-expressed beta 2ARs.

The nonreciprocal nature of the cross-inhibition of beta 2AR internalization induced by V2R activation was particularly remarkable because both receptors were expressed at closely similar levels (see above), and the flow cytometric analysis indicated that the beta 2AR (when activated individually in the absence of V2R activation) can internalize in these cells to a significantly greater extent at steady state than the V2R (Fig. 2B). To begin to examine the mechanism of the selective V2R-mediated endocytic inhibition, we examined its generality to several other receptors that endocytose via clathrin-coated pits. AVP-induced activation of the V2R caused no detectable inhibition of constitutive endocytosis of endogenously expressed transferrin receptors, as visualized by fluorescence microscopy using Texas Red-conjugated transferrin (not shown). However, V2R activation did cause a nearly complete blockade of etorphine-stimulated internalization of co-expressed HA-tagged delta -opioid receptor, which was similar to that observed for the beta 2AR and was also nonreciprocal (Fig. 3). Together, these observations indicate that the V2R-mediated inhibition is specific to the mechanism mediating agonist-induced endocytosis of certain GPCRs and does not reflect a more general inhibition or saturation of the clathrin-mediated endocytic pathway.


View larger version (64K):
[in this window]
[in a new window]
 
Fig. 3.   Block of agonist-induced internalization of the delta -opioid receptor by agonist stimulation of co-expressed V2R. HEK293 cells were transiently transfected with the HA epitope-tagged delta -opioid receptor (delta OR) and the FLAG epitope-tagged V2R (see "Experimental Procedures"). Cells were treated for 30 min at 37 °C with either 5 µM etorphine alone (a and b) or with both 5 µM etorphine and 10 µM AVP (c and d). The distribution of both receptors after treatment was visualized by immunofluorescence microscopy. The upper panels show the localization of the FLAG-V2R, and the lower panels show the localization of the delta -opioid receptor, respectively.

Opioid receptors, like the beta 2AR, recruit beta -arrestins to the plasma membrane of transfected HEK293 cells but fail to mediate detectable endomembrane recruitment of arrestins (28-31). In contrast, the V2R recruits beta -arrestins both to the plasma membrane and to V2R-containing endocytic vesicles (17). These observations suggest that the nonreciprocal inhibition of beta 2AR internalization by V2R activation might be mediated by receptor-specific trafficking of arrestins to endomembranes. To begin to test this hypothesis, we examined the effect of overexpressing beta -arrestins on the V2R-mediated blockade of beta 2AR internalization. Cells co-expressing FLAG-tagged V2R and HA-tagged beta 2AR were transiently transfected with a plasmid encoding a GFP-tagged version of arrestin 3 (beta -arrestin 2) (24). Then cells were incubated as above in the presence of both AVP and ISO, and triple-color fluorescence microscopy was used to visualize the subcellular distribution of V2R, beta 2AR, and GFP-tagged arrestin 3 in the same cells. In contrast to the complete lack of detectable beta 2AR internalization in cells expressing beta -arrestins at endogenous levels (Figs. 1 and 2), internalization of the beta 2AR was readily observed under the same conditions in cells overexpressing GFP-tagged arrestin 3 (Fig. 4A). Many of the punctate structures containing beta 2ARs observed in cells overexpressing beta -arrestins could be resolved from the plasma membrane by confocal optical sectioning and were inaccessible to antibody in nonpermeabilized cells (not shown), demonstrating that these structures represent bona fide beta 2AR-containing endocytic vesicles (rather than clusters of receptors present in the plasma membrane). These results were confirmed in multiple cells examined in coded specimens (Fig. 4B). Similar results were observed when transiently transfecting both receptors into cells stably overexpressing an EE epitope-tagged arrestin 2 (beta -arrestin 1, not shown). Significantly, overexpression of beta -arrestins has little effect on ISO-induced internalization of the beta 2AR in HEK293 cells not expressing the V2R (32). Together these results suggest that overexpression of beta -arrestins is sufficient to specifically "rescue" the V2R-mediated inhibition of beta 2AR internalization. Furthermore we observed that beta 2ARs and V2Rs visualized in arrestin-transfected cells were observed in an overlapping population of endocytic vesicles that colocalized extensively with GFP-tagged arrestin 3 (Fig. 4A, corresponding arrows in each panel indicate examples of such colocalized vesicles in a representative high-power field including several arrestin-transfected cells). These results demonstrate that once the V2R-mediated "sequestration" of arrestin activity is overcome, the co-expressed beta 2AR and V2R can enter a similar endocytic pathway.


View larger version (71K):
[in this window]
[in a new window]
 
Fig. 4.   Reversal of the V2R-mediated blockade of beta 2AR internalization by overexpression of beta -arrestin. HEK293 cells stably expressing both FLAG epitope V2R and HA epitope-tagged beta 2AR were transiently transfected with an expression plasmid encoding GFP-tagged arrestin 3. Cells were treated for 20 min at 37 °C with both 10 µM ISO and AVP. A, the distribution of the beta 2AR (a), GFP-arrestin 3 (b), and the V2R (c) after treatment was visualized in the same cells by triple-label fluorescence microscopy (see "Experimental Procedures"). The white arrows indicate examples of intracellular vesicles in which all three proteins are co-localized. B, receptor internalization for both receptors was estimated by scoring of receptor localization after visualization by immunofluorescence microscopy (see "Experimental Procedures"). Only cells with detectable expression of both receptors were included in the scoring; cells with and without detectable expression of GFP-arrestin 3 were scored separately in the same specimen.

To examine whether endomembrane trafficking of beta -arrestins is actually necessary for the V2R-mediated inhibition of beta 2AR internalization, we used a mutational strategy to disrupt V2R-mediated recruitment of beta -arrestins to endocytic vesicles. The wild type V2R is highly resistant to dephosphorylation in endocytic vesicles, leading to endomembrane recruitment of arrestins and inhibited recycling of receptors (17, 18). Truncation mutations (362T and 345T) of the cytoplasmic tail create functional receptors that undergo agonist-induced endocytosis with similarly rapid kinetics as the wild type receptor (18) but exhibit distinct defects in agonist-dependent phosphorylation/dephosphorylation. The 345T mutant receptor does not exhibit detectable phosphorylation after exposure to agonist; the 362T mutant receptor undergoes agonist-induced phosphorylation but is rapidly dephosphorylated after endocytosis (17, 18). Significantly, neither mutant receptor mediates detectable endomembrane recruitment of beta -arrestins (17). We confirmed this result (not shown) and then compared the ability of FLAG-tagged versions of full-length, 362T and 345T mutant V2Rs to mediate AVP-induced inhibition of the beta 2AR (co-expressed in HEK293 cells without overexpression of beta -arrestins). Consistent with observations in stably transfected cells (Figs. 1 and 2), activation of the full-length V2R strongly inhibited agonist-induced internalization of co-expressed beta 2ARs in transiently transfected HEK293 cells (Fig. 5A, panels a and b). In contrast, neither the 345T (Fig. 5A, panels c and d) nor the 362T (Fig. 5A, panels e and f) mutant receptor mediated detectable inhibition of beta 2AR internalization when examined under the same conditions. The ability of these mutations to abrogate the AVP-induced inhibition of beta 2AR internalization was not a result of differences in expression levels of the mutant receptors, as immunofluorescence staining intensity present in individual transfected cells (estimated using an electronic camera connected to the fluorescence microscope, not shown) confirmed that 345T, 362T, and full-length V2Rs were expressed over a similar range of expression levels in individual transfected cells. The differential effects of mutant V2Rs on internalization of the co-expressed beta 2AR were confirmed by examining multiple cells at random in coded specimens (Fig. 5B). We also observed extensive colocalization of the beta 2AR with both the 362T and 345T mutant receptors in endocytic membranes visualized in the same cells by dual label fluorescence microscopy (arrows in Fig. 5A indicate examples of such colocalized endocytic vesicles). These results further confirm that, once the beta 2AR endocytic inhibition is rescued either by overexpression of beta -arrestins (Fig. 4) or by mutations of the V2R that block endomembrane recruitment of beta -arrestins (Fig. 5), the beta 2AR can enter a similar endocytic pathway as the V2R.


View larger version (69K):
[in this window]
[in a new window]
 
Fig. 5.   Trafficking of beta 2AR with co-expressed full-length or truncated forms of the V2R. HEK293 cells were transiently transfected with the beta 2AR and either full-length (wt) V2R or one of the two truncated mutant V2 receptors, 345T or 362T (see "Experimental Procedures"). Cells were treated for 20 min at 37 °C with both 10 µM ISO and AVP. A, the distribution of both receptors after treatment was visualized by immunofluorescence microscopy. The upper panels show the localization of the different forms of V2R, and the lower panels show the localization of the beta 2AR. The white arrows indicate examples of intracellular vesicles in which both receptors are co-localized. B, receptor internalization for both receptors was estimated by scoring of receptor localization after visualization by immunofluorescence microscopy (see "Experimental Procedures"). Only cells with detectable expression of both receptors were included in the scoring.

We conclude that AVP-induced activation of the V2R mediates a pronounced heterologous inhibition of agonist-induced endocytosis of the beta 2AR as well as certain other GPCRs that endocytose via clathrin-coated pits. This inhibition is clearly not reciprocal, because activation of the beta 2AR with saturating concentrations of agonist does not detectably inhibit agonist-induced endocytosis of the V2R, even in cells in which both receptors are expressed at closely similar levels and in which steady state internalization of the beta 2AR (induced by ISO in the absence of AVP) is significantly greater than that of the V2R. The nonreciprocal nature of this inhibition is correlated with a previously described difference in the effects of the beta 2AR and V2R on intracellular trafficking of beta -arrestins (17). We established that overexpression of beta -arrestins is sufficient to rescue the heterologous inhibition of beta 2AR internalization mediated by the wild type V2R. Moreover, mutations of the V2R that prevent endomembrane recruitment of beta -arrestins abrogate the AVP-induced inhibition of beta 2AR internalization in cells expressing beta -arrestins at endogenous levels. Thus we propose that receptor-specific intracellular trafficking of beta -arrestins depletes the functional pool of cytoplasmic beta -arrestins below that required to promote ligand-induced endocytosis of certain GPCRs.

Whereas receptor-specific differences in the intracellular trafficking of various GPCRs have been examined in considerable detail, relatively little is known about the functional consequences of receptor-specific differences in the intracellular trafficking of beta -arrestins. Recent studies suggest that beta -arrestins associated with endocytic membranes may play an important role in organizing or modulating downstream signal transduction via endocytosed GPCRs (19, 20, 33). However, to our knowledge the present results provide the first direct evidence for a specific functional consequence of beta -arrestin trafficking in regulating GPCR endocytosis itself. Furthermore, we believe these results provide the first data implicating beta -arrestins in a heterologous form of GPCR regulation, which may complement previous studies suggesting heterologous regulation of certain G protein-coupled receptor kinases (34, 35).

In this study we have focused primarily on V2R-mediated effects on regulated internalization of a subset of GPCRs, which have been established most definitively to endocytose in HEK293 cells by clathrin-coated pits. Therefore further studies will be necessary to examine the generality of our observations to other cell types and other GPCRs, particularly receptors that are endocytosed by "alternative" mechanisms (8, 36) or further differ in their effects on intracellular trafficking of beta -arrestins (16, 37). It will also be important to determine whether the mechanism characterized in the present study is capable of mediating cross-inhibition of other important arrestin-dependent functions such as functional uncoupling of receptors from heterotrimeric G proteins or receptor-mediated signaling via mitogenic kinase cascades. The levels of receptor expression examined in the present studies, although moderate compared with other studies of transfected cells, may still be considerably higher than observed in native tissues. Thus the relevance of the present observations to endogenously expressed GPCRs remains to be determined. Nevertheless, as the process of endomembrane recruitment of beta -arrestins can be observed in natively expressing neurons (37), we anticipate that the heterologous regulatory mechanism described in the present study may be of considerable physiological importance. For example it is tempting to speculate that this mechanism may be relevant to the recently reported ability of the V2R to bypass desensitization of myocardial adrenergic receptors in an experimental model of congestive heart failure (38). The potential physiological relevance of this regulatory mechanism notwithstanding, the present results also establish a novel experimental method by which an important functional activity of cytoplasmic beta -arrestins can be modulated acutely in living cells. We anticipate that this approach may be useful as an adjunct to previously described methods (e.g. dominant loss-of-function mutations (39, 40), antisense knockdown (41, 42), and gene knockout approaches (43, 44)) that typically allow cellular beta -arrestins to be manipulated only over a much longer time scale.

    ACKNOWLEDGEMENTS

We thank Dr. J. L. Benovic for providing cDNA encoding arrestin 2 and Dr. M. G. Caron for providing cDNA encoding GFP-arrestin 3. We thank Dr. B. Kobilka for use of his cell harvester and for valuable discussion.

    FOOTNOTES

* These studies were supported by research grants from the National Institutes of Health.The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Dagger Supported during part of these studies by a postdoctoral fellowship from the American Heart Association. To whom correspondence should be addressed: Advanced Medicine, Inc., Dept. of Biochemistry, 901 Gateway Blvd., South San Francisco, CA 94080. Tel.: 650-808-6088; Fax: 313-557-2727; E-mail: uklein@advmedicine.com.

Published, JBC Papers in Press, February 8, 2001, DOI 10.1074/jbc.M009214200

    ABBREVIATIONS

The abbreviations used are: GPCR, G protein-coupled receptor; HEK, human embryonic kidney; beta 2AR, beta 2-adrenergic receptor; V2R, V2 vasopressin receptor; PCR, polymerase chain reaction; HA, hemagglutinin; GFP, green fluorescent protein; AVP, [Arg8]vasopressin; ISO, isoproterenol.

    REFERENCES
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS AND DISCUSSION
REFERENCES

1. Ferguson, S. S., Zhang, J., Barak, L. S., and Caron, M. G. (1998) Life Sci. 62, 1561-1565[CrossRef][Medline] [Order article via Infotrieve]
2. Carman, C. V., and Benovic, J. L. (1998) Curr. Opin. Neurobiol. 8, 335-344[CrossRef][Medline] [Order article via Infotrieve]
3. Lefkowitz, R. J., Pitcher, J., Krueger, K., and Daaka, Y. (1998) Adv. Pharmacol. 42, 416-420[Medline] [Order article via Infotrieve]
4. Hausdorff, W. P., Bouvier, M., O'Dowd, F., Irons, G. P., Caron, M. G., and Lefkowitz, R. J. (1989) J. Biol. Chem. 264, 12657-12665[Abstract/Free Full Text]
5. Bouvier, M. (1990) Ann. N. Y. Acad. Sci. 594, 120-129[Abstract]
6. Hausdorff, W. P., Lohse, M. J., Bouvier, M., Liggett, S. B., Caron, M. G., and Lefkowitz, R. J. (1990) Symp. Soc. Exp. Biol. 44, 225-240[Medline] [Order article via Infotrieve]
7. von Zastrow, M., and Kobilka, B. K. (1994) J. Biol. Chem. 269, 18448-18452[Abstract/Free Full Text]
8. Zhang, J., Ferguson, S. S. G., Barak, L. S., Menard, L., and Caron, M. G. (1996) J. Biol. Chem. 271, 18302-18305[Abstract/Free Full Text]
9. Goodman, O. B., Jr., Krupnick, J. G., Santini, F., Gurevich, V. V., Penn, R. B., Gagnon, A. W., Keen, J. H., and Benovic, J. L. (1996) Nature 383, 447-450[CrossRef][Medline] [Order article via Infotrieve]
10. Koenig, J. A., and Edwardson, J. M. (1997) Trends Pharmacol. Sci. 18, 276-287[CrossRef][Medline] [Order article via Infotrieve]
11. Goodman, O. B., Jr., Krupnick, J. G., Santini, F., Gurevich, V. V., Penn, R. B., Gagnon, A. W., Keen, J. H., and Benovic, J. L. (1998) Adv. Pharmacol. 42, 429-433[Medline] [Order article via Infotrieve]
12. Laporte, S. A., Oakley, R. H., Holt, J. A., Barak, L. S., and Caron, M. G. (2000) J. Biol. Chem. 275, 23120-23126[Abstract/Free Full Text]
13. Gurevich, V. V., and Benovic, J. L. (1997) Mol. Pharmacol. 51, 161-169[Abstract/Free Full Text]
14. Gurevich, V. V., and Benovic, J. L. (2000) Methods Enzymol. 315, 422-437[Medline] [Order article via Infotrieve]
15. Tsao, P. I., and von Zastrow, M. (2000) J. Biol. Chem. 275, 11130-11140[Abstract/Free Full Text]
16. Zhang, J., Barak, L. S., Anborgh, P. H., Laporte, S. A., Caron, M. G., and Ferguson, S. S. (1999) J. Biol. Chem. 274, 10999-11006[Abstract/Free Full Text]
17. Oakley, R. H., Laporte, S. A., Holt, J. A., Barak, L. S., and Caron, M. G. (1999) J. Biol. Chem. 274, 32248-32257[Abstract/Free Full Text]
18. Innamorati, G., Sadeghi, H. M., Tran, N. T., and Birnbaumer, M. (1998) Proc. Natl. Acad. Sci. U. S. A. 95, 2222-2226[Abstract/Free Full Text]
19. Luttrell, L. M., Ferguson, S. S., Daaka, Y., Miller, W. E., Maudsley, S., Della Rocca, G. J., Lin, F., Kawakatsu, H., Owada, K., Luttrell, D. K., Caron, M. G., and Lefkowitz, R. J. (1999) Science 283, 655-661[Abstract/Free Full Text]
20. DeFea, K. A., Zalevsky, J., Thoma, M. S., Dery, O., Mullins, R. D., and Bunnett, N. W. (2000) J. Cell Biol. 148, 1267-1281[Abstract/Free Full Text]
21. Guan, X. M., Kobilka, T. S., and Kobilka, B. K. (1992) J. Biol. Chem. 267, 21995-21998[Abstract/Free Full Text]
22. von Zastrow, M., and Kobilka, B. K. (1992) J. Biol. Chem. 267, 3530-3538[Abstract/Free Full Text]
23. Chen, C., and Okayama, H. (1987) Mol. Cell. Biol. 7, 2745-2752[Medline] [Order article via Infotrieve]
24. Barak, L. S., Ferguson, S. S., Zhang, J., and Caron, M. G. (1997) J. Biol. Chem. 272, 27497-27500[Abstract/Free Full Text]
25. Bradford, M. M. (1976) Anal. Biochem. 72, 248-254[CrossRef][Medline] [Order article via Infotrieve]
26. Keith, D. E., Murray, S. R., Zaki, P. A., Chu, P. C., Lissin, D. V., Kang, L., Evans, C. J., and von Zastrow, M. (1996) J. Biol. Chem. 271, 19021-19024[Abstract/Free Full Text]
27. Pfeiffer, R., Kirsch, J., and Fahrenholz, F. (1998) Exp. Cell Res. 244, 327-339[CrossRef][Medline] [Order article via Infotrieve]
28. Zhang, J., Ferguson, S. S., Barak, L. S., Bodduluri, S. R., Laporte, S. A., Law, P. Y., and Caron, M. G. (1998) Proc. Natl. Acad. Sci. U. S. A. 95, 7157-7162[Abstract/Free Full Text]
29. Whistler, J. L., and von Zastrow, M. (1998) Proc. Natl. Acad. Sci. U. S. A. 95, 9914-9919[Abstract/Free Full Text]
30. Whistler, J. L., Chuang, H. H., Chu, P., Jan, L. Y., and von Zastrow, M. (1999) Neuron 23, 737-746[CrossRef][Medline] [Order article via Infotrieve]
31. Zhang, X., Bao, L., Arvidsson, U., Elde, R., and Hokfelt, T. (1998) Neuroscience 82, 1225-1242[CrossRef][Medline] [Order article via Infotrieve]
32. Menard, L., Ferguson, S. S., Zhang, J., Lin, F. T., Lefkowitz, R. J., Caron, M. G., and Barak, L. S. (1997) Mol. Pharmacol. 51, 800-808[Abstract/Free Full Text]
33. DeFea, K. A., Vaughn, Z. D., O'Bryan, E. M., Nishijima, D., Dery, O., and Bunnett, N. W. (2000) Proc. Natl. Acad. Sci. U. S. A. 97, 11086-11091[Abstract/Free Full Text]
34. Aragay, A. M., Ruiz-Gomez, A., Penela, P., Sarnago, S., Elorza, A., Jimenez-Sainz, M. C., and Mayor, F., Jr. (1998) FEBS Lett. 430, 37-40[CrossRef][Medline] [Order article via Infotrieve]
35. Sarnago, S., Elorza, A., and Mayor, F., Jr. (1999) J. Biol. Chem. 274, 34411-34416[Abstract/Free Full Text]
36. Pals-Rylaarsdam, R., Gurevich, V. V., Lee, K. B., Ptasienski, J. A., Benovic, J. L., and Hosey, M. M. (1997) J. Biol. Chem. 272, 23682-23689[Abstract/Free Full Text]
37. McConalogue, K., Corvera, C. U., Gamp, P. D., Grady, E. F., and Bunnett, N. W. (1998) Mol. Biol. Cell 9, 2305-2324[Abstract/Free Full Text]
38. Laugwitz, K. L., Ungerer, M., Schoneberg, T., Weig, H. J., Kronsbein, K., Moretti, A., Hoffmann, K., Seyfarth, M., Schultz, G., and Schomig, A. (1999) Circulation 99, 925-933[Abstract/Free Full Text]
39. Zhang, J., Barak, L. S., Winkler, K. E., Caron, M. G., and Ferguson, S. S. (1997) J. Biol. Chem. 272, 27005-27014[Abstract/Free Full Text]
40. Krupnick, J. G., Santini, F., Gagnon, A. W., Keen, J. H., and Benovic, J. L. (1997) J. Biol. Chem. 272, 32507-32512[Abstract/Free Full Text]
41. Mundell, S. J., Loudon, R. P., and Benovic, J. L. (1999) Biochemistry 38, 8723-8732[CrossRef][Medline] [Order article via Infotrieve]
42. Mundell, S. J., and Benovic, J. L. (2000) J. Biol. Chem. 275, 12900-12908[Abstract/Free Full Text]
43. Blanchard, E. M., Iizuka, K., Christe, M., Conner, D. A., Geisterfer-Lowrance, A., Schoen, F. J., Maughan, D. W., Seidman, C. E., and Seidman, J. G. (1997) Circ. Res. 81, 1005-1010[Abstract/Free Full Text]
44. Bohn, L. M., Lefkowitz, R. J., Gainetdinov, R. R., Peppel, K., Caron, M. G., and Lin, F. T. (1999) Science 286, 2495-2498[Abstract/Free Full Text]


Copyright © 2001 by The American Society for Biochemistry and Molecular Biology, Inc.