GIPC Interacts with the {beta}1-Adrenergic Receptor and Regulates {beta}1-Adrenergic Receptor-mediated ERK Activation*

Liaoyuan A. Hu {ddagger}, Wei Chen, Negin P. Martin, Erin J. Whalen, Richard T. Premont and Robert J. Lefkowitz, An investigator of the Howard Hughes Medical Institute. §

From the Howard Hughes Medical Institute, Departments of Medicine and Biochemistry, Duke University Medical Center, Durham, North Carolina 27710

Received for publication, December 4, 2002 , and in revised form, April 23, 2003.


    ABSTRACT
 TOP
 ABSTRACT
 INTRODUCTION
 EXPERIMENTAL PROCEDURES
 RESULTS
 DISCUSSION
 REFERENCES
 
{beta}1-adrenergic receptors, expressed at high levels in the human heart, have a carboxyl-terminal ESKV motif that can directly interact with PDZ domain-containing proteins. Using the {beta}1-adrenergic receptor carboxyl terminus as bait, we identified the novel {beta}1-adrenergic receptor-binding partner GIPC in a yeast two-hybrid screen of a human heart cDNA library. Here we demonstrate that the PDZ domain-containing protein, GIPC, co-immunoprecipitates with the {beta}1-adrenergic receptor in COS-7 cells. Essential for this interaction is the Ser residue of the {beta}1-adrenergic receptor carboxyl-terminal ESKV motif. Our data also demonstrate that {beta}1-adrenergic receptor stimulation activates the mitogen-activated protein kinase, ERK1/2. {beta}1-adrenergic receptor-mediated ERK1/2 activation was inhibited by pertussis toxin, implicating Gi, and was substantially decreased by the expression of GIPC. Expression of GIPC had no observable effect on {beta}1-adrenergic receptor sequestration or receptor-mediated cAMP accumulation. This GIPC effect was specific for the {beta}1-adrenergic receptor and was dependent on an intact PDZ binding motif. These data suggest that GIPC can regulate {beta}1-adrenergic receptor-stimulated, Gi-mediated, ERK activation while having no effect on receptor internalization or Gs-mediated cAMP signaling.


    INTRODUCTION
 TOP
 ABSTRACT
 INTRODUCTION
 EXPERIMENTAL PROCEDURES
 RESULTS
 DISCUSSION
 REFERENCES
 
{beta}-Adrenergic receptors ({beta}ARs)1 play a critical role in the regulation of cardiovascular function (1). There are three subtypes of {beta}AR, {beta}1AR, {beta}2AR, and {beta}3AR. Both the {beta}1AR and {beta}2AR subtypes are expressed in the heart and appear to regulate cardiac function through similar intracellular signaling pathways (2). These receptors are members of the seven transmembrane spanning receptor superfamily and are thought to signal largely through heterotrimeric G proteins; primarily Gs. Activation of Gs stimulates adenylate cyclase increasing intracellular cAMP. Increased cAMP activates cAMP-dependent protein kinase (PKA), which is believed to mediate many of the cellular responses associated with {beta}AR activation (3, 4).

Despite similarities in signaling, there are several striking physiological differences between the {beta}1AR and {beta}2AR subtypes (5, 6). For example, {beta}1AR and {beta}2AR subtypes play opposing roles in regulating cardiac myocyte apoptosis: stimulation of the {beta}1AR increases apoptosis, whereas stimulation of the {beta}2AR inhibits apoptosis (7, 8). {beta}1AR-induced apoptosis is implicated in the transition from cardiac hypertrophy to heart failure (9, 10). The differential regulation of cardiac cell survival by these {beta}AR subtypes has been partially explained by the ability of the {beta}2AR to couple to the pertussis toxin (PTX)-sensitive G protein, Gi (11). {beta}2ARs in human and mouse cardiomyocytes have been shown to activate both Gs and Gi, whereas {beta}1AR stimulation has, thus far, only been shown to activate Gs (12). Activation of the {beta}2AR, stimulates a Gi-mediated PI3K-Akt-dependent cell-survival signaling pathway and prevents cardiomyocytes from undergoing Gs-mediated apoptosis (11). Recent studies suggest that Gs to Gi switching by the {beta}2AR is the consequence of PKA-mediated receptor phosphorylation (13, 14).

{beta}2AR/Gi coupling has also been implicated in receptor-meditated ERK activation (13, 15). {beta}2AR-stimulated ERK1/2 activation has been reported in cultured HEK-293 and COS-7 cells, and in isolated cardiac myocytes (16, 17). This {beta}2AR-stimulated ERK1/2 activation is mediated by {beta}{gamma} subunits of PTX-sensitive G proteins (Gi) through a pathway involving the non-receptor tyrosine kinase c-Src, small G protein Ras and Raf-1 kinase (15, 18). The mechanism(s) of {beta}1AR-stimulated ERK activation is somewhat more controversial. A number of groups have reported that the {beta}1AR is unable to stimulate ERK activation, a conclusion based on the perceived inability of the {beta}1AR to couple to Gi (19, 20). However, recent data from cardiac myocytes suggest that the {beta}1AR can activate ERK and p38 in a Gi-dependent manner; even though the {beta}1AR is less potent in stimulating ERK activation than the {beta}2AR (11, 21). The mechanisms behind the observed differential activation of ERK are still unknown.

Recent data suggest that {beta}ARs may transduce signals that are both dependent and independent of heterotrimeric G proteins (22, 23). Well known receptor-interacting proteins, such as {beta}-arrestins and G protein-coupled receptor kinases (GRKs), which were thought previously only to regulate receptor desensitization, have been implicated in signaling pathways that may be G protein-independent (2426). Another G protein-independent mechanism underlying {beta}AR-mediated cell signaling has recently been demonstrated. Both {beta}1ARs and {beta}2ARs have carboxyl-terminal PDZ binding motifs reported to bind to PDZ domain-containing proteins (27, 28). The {beta}2AR has been shown to interact with Na+/H+ exchanger regulatory factors (NHERF) in an agonist-dependent manner via this PDZ binding domain (27); while the {beta}1AR interacts with the postsynaptic density-enriched proteins, PSD-95 and MAGI-2 (28, 29). Association with PSD-95 regulates agonist stimulated {beta}1AR internalization and may also provide a molecular mechanism by which {beta}1ARs are localized to the synapse, regulating synaptic plasticity (28). Interactions between the {beta}1AR and PDZ domain-containing proteins can be regulated by GRK5 (30). Furthermore, studies in cardiac myocytes have recently demonstrated that the PDZ binding motif of the {beta}1AR modulates receptor trafficking and signaling (31).

In order to identify proteins that interact with the PDZ binding domain of the {beta}1AR, we performed a yeast two-hybrid screen on a human heart cDNA library, using the {beta}1AR carboxyl-terminal tail as bait. From this screen we identified GIPC (GAIP-interacting protein, carboxyl (C) terminus), a PDZ-containing protein, as a novel binding partner of the {beta}1AR. Here we characterize this interaction and its functional consequences.


    EXPERIMENTAL PROCEDURES
 TOP
 ABSTRACT
 INTRODUCTION
 EXPERIMENTAL PROCEDURES
 RESULTS
 DISCUSSION
 REFERENCES
 
Plasmids and Antibodies—Mammalian expression plasmids pcDNA3/FLAG-{beta}1AR, pcDNA3/FLAG-{beta}1AR (S475A), and pcDNA3/FLAG-{beta}2AR have been described previously (28, 32). The construction of yeast plasmids pAS2–1/{beta}1AR-CT and pAS2–1/{beta}2AR-CT has also been described previously (28). GFP-tagged ERK2 in pEGFP-N1 expression vector was a gift from Dr. Bunnett at UCSF (33). Rat pACT2/GIPC was a gift from Dr. Farquhar (UCSD) (34). Myc-tagged rat GIPC in pCMV-Tag2B expression plasmid was constructed by cloning a GIPC fragment from pACT2/GIPC into BamH I/XhoI sites of the pCMV Tag-2B vector (Stratagene). The expression plasmid for the PTX-resistant G protein {alpha}i1 (C351S) was purchased from Guthrie cDNA Resource Cener.

Polyclonal (A-14) and monoclonal (9E10) anti-Myc antibodies, and polyclonal anti-ERK2 antibody were from Santa Cruz Biotechnologies. Anti-FLAG M2 and Anti-FLAG M2 affinity gel were from Sigma. Polyclonal anti-phospho-ERK1/2 antibody was from Cell Signaling Technology. Horseradish peroxidase (HRP)-conjugated anti-mouse IgG and anti-rabbit IgG secondary antibodies were from Amersham Biosciences.

Yeast Two-hybrid Screening—Plasmid pAS2–1/{beta}1AR-CT was used as bait to screen a human heart cDNA library (in pACT2) (Clontech). Bait plasmid and library cDNAs were co-transformed into the yeast strain PJ69–4A using a standard yeast transformation protocol. Yeast were plated on selective medium (S.D.-Leu/Trp/His, +10 mM 3AT) and allowed to grow for 4–6 days at 30 °C. Positive colonies were then restreaked on selective medium (S.D.-Leu/Trp/Ade or S.D.-Leu/Trp/His) plates. Plasmid DNA was rescued from positive colonies that grew on both -His and -Ade plates and subject to further sequence analysis. To further confirm positive interactions, isolated library cDNAs were co-transformed back into yeast together with a bait plasmid, either pAS2–1/{beta}1AR-CT, empty vector pAS2–1, or other test plasmids as indicated in the text. Yeast was then subjected to growth tests on selective plates.

Cell Culture and Transfection—All tissue culture media and related reagents were purchased from Invitrogen. COS-7 cells were maintained in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin in a 37 °C incubator under 5% CO2. Cells in 100-mm dishes were transfected with LipofectAMINE (at 4:1 ratio with DNA) according to the manufacturer's protocol. HEK-293 cells were maintained under the same conditions as COS-7 cells except minimal essential medium (MEM) was used. HEK-293 cells were transfected with FuGENE 6 (at 2.5:1 ratio with DNA) according to the manufacturer's protocol (Roche Applied Science). After transfection, cells were grown 36–48 h before agonist stimulation.

Cellular Immunoprecipitations and Immunoblotting—Transfected COS-7 cells in 100-mm plates were incubated in serum-free medium for 60 min before agonist stimulation. The cells were then incubated in the presence or absence of 10 µM isoproterenol (ISO) for 10 min at 37 °C. The cells were rinsed with cold Dulbecco's phosphate-buffered saline (DPBS) and replaced with 1 ml of cross-linking buffer (DPBS containing 10 mM HEPES, pH 7.4, and 2.5 mM of the cell permeable cross-linking reagent dithiobis(succinimidyl propionate) (DSP, Pierce)). For the stimulated cells, 10 µM ISO was present in the cross-linking buffer. The cells were incubated for 30 min at room temperature with continuous slow rocking. The cross-linking reaction was terminated by quickly removing the cross-linking buffer and replacing it with 1 ml of ice-cold lysis buffer (50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 2 mM EDTA, 10% glycerol, 0.5% Nonidet P-40, and protease inhibitors). The cells were lysed by incubation on ice for 30 min and then clarified by centrifugation at 21,000 x g for 12 min at 4 °C. The clarified supernatants were then used in cellular coimmunoprecipitation experiments. 50 µl of each supernatant was diluted into an equal amount of 3x SDS-PAGE sample buffer to serve as whole cell extract controls.

1 ml of clarified cell extract was then incubated with 25 µl of anti-FLAG M2 affinity gel slurry at 4 °C with gentle rotation for 4 h to overnight. Beads were washed four to five times with ice-cold lysis buffer, and the bound proteins were eluted with 50 µl of 3x SDS-PAGE sample buffer. Equivalent amounts of sample in 3x sample buffer were resolved on precast 4–20% polyacrylamide gels (Invitrogen) and then transferred to nitrocellulose membranes by semi-dry blotting. Nitrocellulose membranes were blocked with 5% fat-free milk in TBST (20 mM Tris, pH 7.4, 500 mM NaCl, and 0.1% Tween 20) and incubated with the appropriate primary antibody at room temperature for 1 h. After washing with TBST, membranes were incubated for 1 h with horseradish peroxidase-conjugated anti-mouse or rabbit IgG secondary antibody (1:2000). Protein bands were visualized via SuperSignal Chemiluminescence Substrate (Pierce). Quantitation of band density was performed with Bio-Rad Fluor-S MultiImager.

ERK Phosphorylation—Twenty-four hours after transfection, cells were split into 6-well dishes and then incubated in serum-free media (DMEM for COS-7 and MEM for HEK-293, 0.1% bovine serum albumin, 10 mM HEPES, pH 7.4) overnight before agonist stimulation. Agonist stimulation was performed at 37 °C in serum-free media for the times indicated in the figure legends. The medium was removed, and cells were then solubilized in 3x SDS-PAGE sample buffer. The whole cell lysates were sonicated and resolved by SDS-PAGE. ERK phosphorylation was detected by Western blot using anti-phospho-ERK1/2 antibody as described above. The total ERK2 in the cell lysates was detected using anti-ERK2 polyclonal antibody. Protein bands were visualized via SuperSignal Chemiluminescence substrate and quantitated with Bio-Rad Fluor-S MultiImager.

Receptor Internalization and Cyclic AMP Accumulation Assays—For receptor internalization assays, HEK-293 cells in 100-mm dishes were transiently transfected with pcDNA3/FLAG-{beta}1AR in the presence and absence of Myc-GIPC. One day after transfection, cells were split into poly-D-lysine-coated 6-well plates (Biocoat) and grown overnight at 37 °C. Cells were serum-starved for 1 h before stimulation with 10 µM ISO for 30 min at 37 °C. Cells were placed on ice and cell surface FLAG-tagged receptors were detected with anti-FLAG M2 antibody followed by FITC-conjugated anti-mouse IgG as described previously (35). Receptor internalization was defined as the percentage of cell surface receptors lost after agonist stimulation, measured by cell flow cytometry.

For cyclic AMP accumulation assays, HEK-293 cells in 100-mm dishes were transiently transfected with pcDNA3/FLAG-{beta}1AR in the presence and absence of transfected Myc-GIPC. Twenty-four hours after transfection, cells were split into 12-well collagen-coated plates and then labeled with modified essential medium supplemented with 5% fetal bovine serum and 2 µCi/ml [3H]adenine for 4 h to overnight. Cells were serum-starved for 30 min and then stimulated with {beta}1AR selective agonist dobutamine or 10 µM forskolin for 10 min. Cyclic AMP accumulation was quantitated by chromatography and expressed as a percentage of 3H incorporated into cyclic AMP as described previously (35).


    RESULTS
 TOP
 ABSTRACT
 INTRODUCTION
 EXPERIMENTAL PROCEDURES
 RESULTS
 DISCUSSION
 REFERENCES
 
GIPC Binds Specifically to the {beta}1AR Carboxyl-terminal Tail—In our search for potential {beta}1AR binding partners, we used the {beta}1AR carboxyl-terminal tail ({beta}1AR-CT) as bait in a yeast two-hybrid screen of a human heart cDNA library. From a total of 4 million independent colonies screened, 24 positive clones were obtained. Four of the positive clones yielded identical sequence encoding a portion of the gene for human GIPC (hGIPC), with an N-terminal truncation at residue 74. We also isolated rat GIPC from a yeast two-hybrid screen of rat brain. The isolated rat sequence (rGIPC) encoded full-length GIPC plus a portion of the 5'-untranslated region (Fig. 1).



View larger version (30K):
[in this window]
[in a new window]
 
FIG. 1.
Identification of GIPC as a novel binding partner of the {beta}1AR carboxyl terminus in yeast two-hybrid screens. A, domain structure of GIPC and two isolated GIPC clones. The {beta}1AR carboxyl terminus was used as bait to screen a human heart cDNA library in a yeast two-hybrid system. The GIPC clones isolated from a human library (hGIPC) encoded a partial sequence of GIPC; while the clone isolated from the rat library (rGIPC) encoded a full-length GIPC plus some 5'-untranslated region. The PDZ domain of GIPC has been high-lighted for comparison. B, GIPC interacts specifically with the {beta}1AR-CT in a yeast two-hybrid system. Yeast strain PJ69–4A was co-transformed with bait DNAs (pAS2–1, pAS2–1/{beta}1ARCT, or pAS2–1/{beta}2ARCT) together with isolated preys. Yeast colonies were subjected to growth tests on selective medium -His (S.D.-Leu/Trp/His) or -Ade (S.D. -Leu/Trp/Ade). Yeast growth rates were rated from fast (+++) to slow (+) to no growth (-).

 

The specificity of the interaction between GIPC and the {beta}1AR-CT was confirmed by further yeast two-hybrid analysis. GIPC clones were transformed back into yeast strain PJ69-4A together with baits (either {beta}1AR-CT or {beta}2AR-CT). Transformed yeast containing both bait and GIPC were then subjected to growth tests on selective medium. As shown in Fig. 1, only yeast hosting both GIPC and the {beta}1AR-CT were able to grow on both -His and -Ade plates. The interaction between the {beta}1AR and GIPC is specific, since the {beta}2AR-CT did not support yeast growth under the same conditions when co-transformed with GIPC.

The {beta}1-Adrenergic Receptor Interacts with GIPC Through Its Carboxyl-terminal PDZ Binding Motif—In order to determine whether the full-length {beta}1AR could associate with GIPC in the cell, COS-7 cells were co-transfected with FLAG-{beta}1AR and Myc-GIPC. Immunoprecipitation of the {beta}1AR followed by Western blotting for Myc-GIPC revealed robust co-immunoprecipitation of a GIPC/{beta}1AR complex (Fig. 2). Treatment of the cells with the {beta}-adrenergic receptor agonist ISO had no significant effect on the GIPC/{beta}1AR association (data not shown). Consistent with the yeast two-hybrid results, GIPC did not co-immunoprecipitate with the full-length FLAG-{beta}2AR when the two proteins were co-expressed in COS-7 cells.



View larger version (45K):
[in this window]
[in a new window]
 
FIG. 2.
GIPC associates specifically with the {beta}1AR through a PDZ domain. HEK-293 or COS-7 cells were transiently transfected with FLAG-tagged wild-type receptors ({beta}1AR or {beta}2AR) or various {beta}1AR mutants (V477A, S475A, S475D, and S473A) and/or rat Myc-GIPC. Forty-eight hours post-transfection, cells were washed with Dulbecco's phosphate-buffered saline and exposed to the cross-linking reagent DSP for 30 min at room temperature. The reaction was quickly quenched by ice-cold lysis buffer. The cell lysates were clarified, and receptors were immunoprecipitated with anti-FLAG M2 beads as described under "Experimental Procedures." GIPC bound to the beads was resolved by SDS-PAGE and Western blotted with an anti-Myc antibody (upper panel). Expression of GIPC in the cell lysates is shown in the bottom panel. These data are representative of five similar experiments.

 

To examine the structural determinants of the GIPC/{beta}1AR interaction, we used several {beta}1AR mutants, in which three out of the last five residues were replaced with Ala individually (28). Similar to that observed with the {beta}1AR/PSD-95 interaction, when the Ser residue at the -2 position of the {beta}1AR was mutated to either Ala (S475A) or Asp (S475D), the association of GIPC with the {beta}1AR could no longer be detected. When GIPC was co-expressed with the {beta}1AR mutant S473A, co-immunoprecipitation of GIPC was reduced only slightly relative to the wild-type {beta}1AR. However, the {beta}1AR mutant V477A, which is unable to bind with PSD-95, showed a very strong interaction with GIPC. These studies demonstrate by both yeast two-hybrid screening and cellular co-immunoprecipitation experiments that the GIPC specifically interacts with the {beta}1AR through the PDZ domain. Unlike the association of the {beta}1AR with PSD-95, the Val in the last position (Val-477) of the receptor is not required for the interaction with GIPC.

GIPC Has No Effect on Receptor-stimulated Cyclic AMP Accumulation—Next we examined the effect of GIPC on {beta}1AR-mediated whole cell cAMP accumulation. HEK-293 cells were transiently transfected with wild-type {beta}1AR, and either Myc-tagged GIPC or vector alone. Dose-response curves of cAMP accumulation induced by the selective {beta}1AR agonist dobutamine are shown in Fig. 3. Similar to previous results with PSD-95, GIPC expression had no effect on {beta}1AR-mediated whole cell cAMP accumulation, changing neither the maximal level of cAMP accumulation nor the EC50. Because PDZ domain-containing proteins have previously been shown to regulate receptor internalization (28, 29), we also examined the effect of GIPC on {beta}1AR internalization in HEK-293 cells. GIPC expression had no effect on agonist stimulated {beta}1AR internalization (data not shown).



View larger version (15K):
[in this window]
[in a new window]
 
FIG. 3.
GIPC has no effect on {beta}1AR-mediated cyclic AMP accumulation. HEK-293 cells were transiently transfected with pcDNA3/FLAG-{beta}1AR alone or with Myc-GIPC. Forty-eight hours after transfection, cells were treated with serum-free medium for 30 min and then stimulated with the {beta}1AR-selective agonist dobutamine (at the concentrations indicated) or 50 µM forskolin for 10 min at 37 °C. It was necessary to use dobutamine instead of ISO for these experiments to avoid activation of endogenous {beta}2-adrenergic receptors in the HEK-293 cells. Whole cell cAMP accumulation was determined by chromatography and presented as percentage conversion of [3H]adenine into [3H]cAMP. Dobutamine-induced cAMP accumulation was normalized to that induced by 10 µM forskolin. These data are representative of three similar experiments.

 

{beta}1AR-stimulated ERK Activation—In order to examine {beta}1AR-mediated ERK activation, we used a plasmid encoding an ERK2-GFP fusion protein. This allowed for GFP-ERK2 to be easily separated from endogenous ERK1/2 by SDS-PAGE. In COS-7 cells transiently transfected with pcDNA3/FLAG-{beta}1AR and GFP-ERK2, ISO stimulation induced robust ERK phosphorylation/activation compared with non-stimulated cells (Fig. 4A). In control cells transfected with GFP-ERK2 alone, ISO-stimulated ERK phosphorylation was not detected using phospho-ERK antibody.



View larger version (24K):
[in this window]
[in a new window]
 
FIG. 4.
{beta}1AR-mediated ERK activation. COS-7 cells were transiently transfected with the {beta}1AR and GFP-ERK2. Forty-eight hours after transfection, cells were treated with serum-free medium overnight. ERK activation was performed as described under "Experimental Procedures." A, {beta}1AR stimulation enhanced ERK phosphorylation. Serum-starved COS-7 cells were stimulated for 5 min with 10 µM ISO at 37 °C. The cells were solubilized in 3x SDS-PAGE sample buffer. Phosphorylation of the ERK in the whole cell lysates was detected by Western blot analysis using an anti-phospho-ERK1/2 antibody. The data presented in A–D are representative of a minimum of three independent experiments. B, dose-dependent ISO-stimulated {beta}1AR-stimulated ERK activation. Serum-starved COS-7 cells were incubated with different concentration of ISO for 5 min and then phospho-ERK was determined by anti-phospho-ERK1/2 antibody. C, time course of the {beta}1AR-stimulated ERK activation. Serum-starved COS-7 cells were incubated with 10 µM ISO for the indicated times, and then ERK phosphorylation in the whole cell lysates was detected using an anti-phospho-ERK1/2 antibody. D, effect of different agonist stimulation on the {beta}1AR-mediated ERK activation. Serum-starved COS-7 cells were incubated with 10 µM ISO, norepinephrine (NE), dobutamine (Dobu), 1 µM PMA or 10 µM forskolin (Forsk) for 5 min. The ERK phosphorylation in the whole cell lysates was detected using anti-phospho-ERK1/2 antibody. The amount of phosphorylated-ERK from each sample was normalized to that from unstimulated cells. E, {beta}1AR-stimulated ERK activation was inhibited by {beta} antagonist or PTX pretreatment, but not by the PKA inhibitor H89. Serum-starved COS-7 cells were treated with 100 ng/ml PTX overnight, or 10 µM H89, 10 µM propranolol (Pro) or 10 µM alprenolol (ALP) for 30 mins prior to ISO stimulation. The amount of ERK phosphorylation from ISO-stimulated cells was set as control. The amount of phosphorylated-ERK from each sample was expressed as a percentage of control. The data are presented as means ± S.E. from four independent experiments. F, {beta}1AR-stimulated ERK activation was inhibited by PTX. The effects of PTX treatment were reversed by overexpressing a mutant Gi (C351), which could not be ADP-ribosylated. Serum-starved COS-7 cells were treated with 100 ng/ml of PTX overnight, stimulated with 10 µM ISO, and run on Western blots as described previously. The amount of phosphorylated-ERK from each sample was expressed as a percentage of control in cells expressing {beta}1AR. *, p < 0.05.

 

Similar to previous studies of the {beta}2AR, {beta}1AR-stimulated ERK activation was a relatively fast process, peaking at around 5 min. At maximum, ISO-stimulated ERK activation was 6–8 fold greater than that of non-stimulated control cells (Fig. 4B). After peaking at 5 min, {beta}1AR-stimulated ERK activation steadily decreased to ~50% of maximum at 15 min. {beta}1AR-stimulated ERK activation was observed over a range of agonist concentrations. The dose response curve for ISO-stimulated ERK activation is shown in Fig. 4C.

Next we examined the effect of different agonists on {beta}1AR-mediated ERK activation in COS-7 cells. The {beta}1AR has a high affinity for both norepinephrine and epinephrine (2). As predicted, norepinephrine (NE) stimulation dramatically increased ERK activation (Fig. 4D). The {beta}1AR specific agonist dobutamine (Dobu) also stimulated ERK activation, but was much less active than either NE or ISO. As a control, PMA, the direct activator of PKC, demonstrated robust ERK activation, whereas the direct activator of adenylate cyclase, forskolin (Forsk), showed no demonstrable ERK activation. As expected, ISO-stimulated ERK activation was completely blocked by the {beta}-adrenergic receptor antagonists propranolol (PRO) and alprenolol (ALP) (Fig. 4E).

Treatment of cells with 10 µM PTX overnight decreased {beta}1AR-mediated ERK activation considerably. To further demonstrate that PTX inhibited the activation of ERK by inhibiting the {alpha}i subunit of the G protein, we overexpressed a G protein {alpha}i1 subunit (C351S), which cannot be ADP-ribosylated and is therefore unaffected by PTX treatment. As shown in Fig. 4F, overexpression of Gi (C351S), while having little effect on the overall ISO-stimulated ERK activity, leads to a marked increase in ISO-stimulated ERK activation observed in the presence of PTX. The residual effect of PTX is presumably due to inhibition of endogenous Gi. These data further support the idea that the {beta}1AR activates ERK through a Gi-mediated pathway, similar to that reported for the {beta}2AR (13). In contrast to findings with the {beta}2AR (13), the PKA inhibitor H89 had no significant effect on {beta}1AR-mediated ERK activation.

GIPC Regulates {beta}1AR-mediated ERK Activation—GIPC is a PDZ domain-containing protein that interacts with a Gi-specific RGS protein called GAIP (G{alpha}-interacting protein) (34); therefore, GIPC could be involved in the regulation of Gi-mediated signaling. Here we tested the effect of GIPC on {beta}1AR-stimulated ERK activation, which may be mediated by the PTX-sensitive G protein, Gi. As shown in Fig. 5A, expression of GIPC substantially decreased ISO-stimulated {beta}1AR-mediated ERK activation. ISO-stimulated ERK activation in the presence of GIPC was decreased by 45% compared with control cells lacking GIPC (Fig. 5, A and D). Under similar conditions, expression of GIPC had little effect on {beta}2AR-mediated ERK activation (Fig. 5B). The inhibitory effect of GIPC appeared to be specific for the {beta}1AR. GIPC had no significant effect on the ability of the {beta}1AR mutant (S475A), which is unable to interact with GIPC, to activate ERK (Fig. 5C). These data suggest that the ability of GIPC to regulate {beta}1AR-mediated ERK activation is dependent on its ability to interact directly with the {beta}1AR via its PDZ domain.



View larger version (21K):
[in this window]
[in a new window]
 
FIG. 5.
GIPC specifically inhibits {beta}1AR-stimulated ERK activation. COS-7 cells were transiently transfected with {beta}1AR, {beta}2AR, or {beta}1AR S475A mutant and GIPC plus GFP-ERK2. Forty-eight hours after transfection, cells were treated with serum-free medium overnight. Serum-starved cells were stimulated with 10 µM ISO for 5 min at 37 °C. ERK activation was assayed as described under "Experimental Procedures." Expression of GIPC dramatically inhibits {beta}1AR-mediated ERK activation (A), but has little effect on {beta}2AR- (B) or {beta}1AR S475A ({beta}1AR(M)) mutant (C)-mediated ERK activation. D, summary of the effect of GIPC on {beta}1AR-, {beta}2AR-, and {beta}1AR S457A mutant ({beta}1AR(M))-mediated ERK activation. ISO-stimulated ERK activation in the absence of GIPC was set as control. The amount of phospho-ERK in the presence of GIPC was expressed as a percentage of the control. The data are presented as means ± S.E. from four independent experiments.

 


    DISCUSSION
 TOP
 ABSTRACT
 INTRODUCTION
 EXPERIMENTAL PROCEDURES
 RESULTS
 DISCUSSION
 REFERENCES
 
{beta}1ARs are of primary importance in regulating heart rate and contractility. Through a yeast two-hybrid screen of a human heart cDNA library, we identified GIPC as a novel binding partner for the {beta}1AR. GIPC is a PDZ domain-containing protein that was originally identified and named as a protein that interacts with GAIP, an RGS (regulator of G protein signaling) protein (34). In addition to GAIP, GIPC has also been reported to interact with several membrane proteins through its PDZ domain; these include the Glut-1 transporter, transmembrane Semaphorin-F (M-SemF) (36), neurophilin-1 (37), insulin-like growth factor 1 receptor (38), gp75 (tyrosinase-related protein-1), transforming growth factor {beta} (TGF-{beta}) receptor (39), 5T4 transmembrane glycoprotein (40), integrins {alpha}5, {alpha}6A {alpha}6B (41, 42), and the viral protein TAX (43).

Here we report a specific interaction between the {beta}1AR and GIPC. Despite the fact that both the {beta}1AR and {beta}2AR contain similar PDZ binding motifs, GIPC interacts only with the {beta}1AR, as demonstrated by both yeast two-hybrid screening and co-immunoprecipitation experiments. The GIPC PDZ domain facilitates the interaction with the {beta}1AR. Mutation of Ser-475 to either Ala (S475A) or Asp (S475D) completely eliminated the interaction. However, unlike that reported for PSD-95, changing the last residue Val-477 of the {beta}1AR to Ala had no effect on the {beta}1AR/GIPC association. TAX, a protein containing a similar carboxyl-terminal ETEA-COOH motif has also been shown to interact with GIPC (43). GIPC expression has no significant effect on {beta}1AR-induced cAMP accumulation. Therefore, similar to PSD-95, the association of GIPC with the {beta}1AR had no effect on {beta}1AR coupling to Gs. In contrast to PSD-95, which inhibits receptor internalization in HEK-293 cells (31), GIPC had little to no effect on agonist-stimulated receptor internalization.

Recently, many GPCRs have been reported to activate MAP kinase cascades regulating cell growth and/or proliferation (18). In COS-7 cells, it has been clearly demonstrated that stimulation of the {beta}2AR activates the MAP kinase ERK through a Gi-mediated pathway (13, 44). In cardiac myocytes, {beta}2AR stimulation has been shown to activate the MAP kinases ERK, JNK, and p38 (21, 45). However, {beta}1AR-mediated ERK activation or coupling to Gi is still quite controversial. Here, we demonstrate {beta}1AR stimulated, PTX-sensitive, ERK activation; strongly suggesting that the {beta}1AR, like the {beta}2AR, can signal through both Gs and Gi.

GIPC, similar to many PDZ domain-containing proteins, has been shown to play an important role in organizing signaling cascades (46), anchoring proteins in specific subcellular compartments (40), and regulating cell signaling (39). GIPC regulates the expression of the TGF-{beta} receptor at the cell surface and enhances the cellular response to TGF-{beta} (39). GIPC has also been linked to G protein signaling by its ability to interact with the RGS protein GAIP (46). Overexpression of GIPC in PC12 cells decreases NGF induced MAP kinase (ERK1/2) activation (38). Interestingly, a GIPC-like protein Kermit, which interacts with the Frizzed receptor, has been reported to regulate frizzed 3 signaling in neural crest development (47). Here we provided further evidence that GIPC regulates G protein signaling by directly interacting with the {beta}1AR in a PDZ-dependent manner. Expression of GIPC decreased the ability of the {beta}1AR to stimulate ERK activation, but had no effect on {beta}1AR-mediated cAMP accumulation. This regulatory effect of GIPC on ERK activation is specific for the {beta}1AR and depends on a PDZ-mediated interaction, since expression of GIPC has no effect on the ERK activation stimulated by either the {beta}2AR or the {beta}1AR mutant (S475A). These data suggest that the GIPC/{beta}1AR association affects receptor Gi- but not Gs-mediated signaling.

In mouse cardiac myocytes, stimulation of the {beta}1AR leads to a PKA-dependent increase in the rate of contraction (48). {beta}2AR stimulation shows a biphasic effect on the rate of cardiac myocyte contraction, with an initial PKA-dependent increase followed by a PTX-sensitive decrease, mediated by Gi (48). Disruption of the PDZ binding motif in the {beta}1AR changed the signaling profile of the {beta}1AR in cardiac myocytes to more resemble that of the {beta}2AR (31). These data suggest that the {beta}1AR is capable of coupling to Gi, but that association with PDZ-containing proteins such as PSD-95 could prevent this interaction from occurring. GIPC interacts with the {beta}1AR through its PDZ domain and thereby specifically inhibits {beta}1AR-stimulated Gi-mediated signaling. Thus, the GIPC effect on {beta}1AR-mediated ERK activation might explain the differential effects of {beta}1AR and {beta}2AR stimulation on the rate of cardiac myocyte contraction.

The effect of GIPC on {beta}1AR/Gi-mediated signaling could also explain the differential effects of {beta}1AR and {beta}2AR stimulation on cardiac myocyte apoptosis. {beta}1AR stimulation leads to cardiac myocyte apoptosis, whereas stimulation of the {beta}2AR concurrently activates pro-apoptotic and anti-apoptotic signals, the net effect being increased cell survival (8). Differential coupling to Gs and Gi has been used to explain this differential effect of {beta}1AR versus {beta}2AR stimulation (10). Here we demonstrate that the {beta}1AR can couple to both Gs and Gi, similar to a number of other GPCRs including the histamine, serotonin, and glucagon receptors (20). The continued study of receptor-interacting proteins, such as PDZ-containing proteins, will further our understanding of differential signaling by receptors.


    FOOTNOTES
 
* This work was supported in part by National Institutes of Health Grant HL16037 (to R. J. L). The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. Back

{ddagger} Recipient of a fellowship from the American Heart Association. Back

§ To whom correspondence should be addressed: Howard Hughes Medical Institute, Box 3821, Duke University Medical Center, Durham, NC, 27710. Tel.: 919-684-2974; Fax: 919-684-8875; E-mail: lefko001{at}receptor-biol.duke.edu.

1 The abbreviations used are: {beta}AR, {beta}-adrenergic receptor; {beta}1AR, {beta}1-adrenergic receptor; {beta}2AR, {beta}2-adrenergic receptor; CT, carboxyl terminus; ERK, extracellular signal-regulated kinase; GAIP, G{alpha}-interacting protein, GIPC, GAIP-interacting protein, carboxyl-terminal; GPCR, G protein-coupled receptor; HEK, human embryonic kidney; GRK, G protein-coupled receptor kinase; MAP kinase, mitogen-activated protein kinase; NHERF, Na+/H+-exchanger regulatory factor; PDZ, PSD-95/Dlg/ZO-1 homology domain; PTX, pertussis toxin; ISO, isoproterenol. Back


    ACKNOWLEDGMENTS
 
We thank Dr. Farquhar for providing GIPC clones, Millie McAdams and Judy Phelps for DNA sequencing, and Julia Turnbough and Donna Addison for assistance with the preparation of this manuscript.



    REFERENCES
 TOP
 ABSTRACT
 INTRODUCTION
 EXPERIMENTAL PROCEDURES
 RESULTS
 DISCUSSION
 REFERENCES
 

  1. Brodde, O. E. (1993) Pharmacol. Ther. 60, 405-430[CrossRef][Medline] [Order article via Infotrieve]
  2. Brodde, O. E. (1991) Pharmacol. Rev. 43, 203-242[Medline] [Order article via Infotrieve]
  3. Pierce, K. L., Premont, R. T., and Lefkowitz, R. J. (2002) Nat. Rev. Mol. Cell. Biol. 3, 639-650[CrossRef][Medline] [Order article via Infotrieve]
  4. Lefkowitz, R. J. (2000) Nat. Cell Biol. 2, E133-E136[CrossRef][Medline] [Order article via Infotrieve]
  5. Steinberg, S. F. (1999) Circ. Res. 85, 1101-1111[Free Full Text]
  6. Xiao, R. P., Cheng, H., Zhou, Y. Y., Kuschel, M., and Lakatta, E. G. (1999) Circ. Res. 85, 1092-1100[Abstract/Free Full Text]
  7. Communal, C., Singh, K., Sawyer, D. B., and Colucci, W. S. (1999) Circulation 100, 2210-2212[Abstract/Free Full Text]
  8. Zhu, W. Z., Zheng, M., Koch, W. J., Lefkowitz, R. J., Kobilka, B. K., and Xiao, R. P. (2001) Proc. Natl. Acad. Sci. U. S. A. 98, 1607-1612[Abstract/Free Full Text]
  9. Kang, P. M., and Izumo, S. (2000) Circ. Res. 86, 1107-1113[Free Full Text]
  10. Xiao, R. P. (2001) Sci. STKE http://stke.sciencemag.org/cgi/content/full/OC_sigtrans;2001/104/re15
  11. Chesley, A., Lundberg, M. S., Asai, T., Xiao, R. P., Ohtani, S., Lakatta, E. G., and Crow, M. T. (2000) Circ. Res. 87, 1172-1179[Abstract/Free Full Text]
  12. Xiao, R. P., Avdonin, P., Zhou, Y. Y., Cheng, H., Akhter, S. A., Eschenhagen, T., Lefkowitz, R. J., Koch, W. J., and Lakatta, E. G. (1999) Circ. Res. 84, 43-52[Abstract/Free Full Text]
  13. Daaka, Y., Luttrell, L. M., and Lefkowitz, R. J. (1997) Nature 390, 88-91[CrossRef][Medline] [Order article via Infotrieve]
  14. Zamah, A. M., Delahunty, M., Luttrell, L. M., and Lefkowitz, R. J. (2002) J. Biol. Chem. 277, 31249-31256[Abstract/Free Full Text]
  15. Luttrell, L. M., van Biesen, T., Hawes, B. E., Koch, W. J., Krueger, K. M., Touhara, K., and Lefkowitz, R. J. (1997) Adv. Second Messenger Phosphoprotein Res. 31, 263-277[Medline] [Order article via Infotrieve]
  16. Crespo, P., Cachero, T. G., Xu, N., and Gutkind, J. S. (1995) J. Biol. Chem. 270, 25259-25265[Abstract/Free Full Text]
  17. Daaka, Y., Luttrell, L. M., Ahn, S., Della Rocca, G. J., Ferguson, S. S., Caron, M. G., and Lefkowitz, R. J. (1998) J. Biol. Chem. 273, 685-688[Abstract/Free Full Text]
  18. Luttrell, L. M. (2002) Can. J. Physiol. Pharmacol. 80, 375-382[CrossRef][Medline] [Order article via Infotrieve]
  19. Lavoie, C., Mercier, J. F., Salahpour, A., Umapathy, D., Breit, A., Villeneuve, L. R., Zhu, W. Z., Xiao, R. P., Lakatta, E. G., Bouvier, M., and Hebert, T. E. (2002) J. Biol. Chem. 277, 35402-35410[Abstract/Free Full Text]
  20. Kilts, J. D., Gerhardt, M. A., Richardson, M. D., Sreeram, G., Mackensen, G. B., Grocott, H. P., White, W. D., Davis, R. D., Newman, M. F., Reves, J. G., Schwinn, D. A., and Kwatra, M. M. (2000) Circ. Res. 87, 705-709[Abstract/Free Full Text]
  21. Communal, C., Colucci, W. S., and Singh, K. (2000) J. Biol. Chem. 275, 19395-19400[Abstract/Free Full Text]
  22. Brzostowski, J. A., and Kimmel, A. R. (2001) Trends Biochem. Sci. 26, 291-297[CrossRef][Medline] [Order article via Infotrieve]
  23. Hall, R. A., Premont, R. T., and Lefkowitz, R. J. (1999) J. Cell Biol. 145, 927-932[Free Full Text]
  24. Miller, W. E., and Lefkowitz, R. J. (2001) Curr. Opin. Cell Biol. 13, 139-145[CrossRef][Medline] [Order article via Infotrieve]
  25. Luttrell, L. M., and Lefkowitz, R. J. (2002) J. Cell Sci. 115, 455-465[Abstract/Free Full Text]
  26. McDonald, P. H., and Lefkowitz, R. J. (2001) Cell Signal. 13, 683-689[CrossRef][Medline] [Order article via Infotrieve]
  27. Hall, R. A., Premont, R. T., Chow, C. W., Blitzer, J. T., Pitcher, J. A., Claing, A., Stoffel, R. H., Barak, L. S., Shenolikar, S., Weinman, E. J., Grinstein, S., and Lefkowitz, R. J. (1998) Nature 392, 626-630[CrossRef][Medline] [Order article via Infotrieve]
  28. Hu, L. A., Tang, Y., Miller, W. E., Cong, M., Lau, A. G., Lefkowitz, R. J., and Hall, R. A. (2000) J. Biol. Chem. 275, 38659-38666[Abstract/Free Full Text]
  29. Xu, J., Paquet, M., Lau, A. G., Wood, J. D., Ross, C. A., and Hall, R. A. (2001) J. Biol. Chem. 276, 41310-41317[Abstract/Free Full Text]
  30. Hu, L. A., Chen, W., Premont, R. T., Cong, M., and Lefkowitz, R. J. (2002) J. Biol. Chem. 277, 1607-1613[Abstract/Free Full Text]
  31. Xiang, Y., Devic, E., and Kobilka, B. (2002) J. Biol. Chem. 277, 33783-33790[Abstract/Free Full Text]
  32. Tang, Y., Hu, L. A., Miller, W. E., Ringstad, N., Hall, R. A., Pitcher, J. A., DeCamilli, P., and Lefkowitz, R. J. (1999) Proc. Natl. Acad. Sci. U. S. A. 96, 12559-12564[Abstract/Free Full Text]
  33. 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]
  34. De Vries, L., Lou, X., Zhao, G., Zheng, B., and Farquhar, M. G. (1998) Proc. Natl. Acad. Sci. U. S. A. 95, 12340-12345[Abstract/Free Full Text]
  35. Freedman, N. J., Liggett, S. B., Drachman, D. E., Pei, G., Caron, M. G., and Lefkowitz, R. J. (1995) J. Biol. Chem. 270, 17953-17961[Abstract/Free Full Text]
  36. Wang, L. H., Kalb, R. G., and Strittmatter, S. M. (1999) J. Biol. Chem. 274, 14137-14146[Abstract/Free Full Text]
  37. Cai, H., and Reed, R. R. (1999) J. Neurosci. 19, 6519-6527[Abstract/Free Full Text]
  38. Booth, R. A., Cummings, C., Tiberi, M., and Liu, X. J. (2002) J. Biol. Chem. 277, 6719-6725[Abstract/Free Full Text]
  39. Blobe, G. C., Liu, X., Fang, S. J., How, T., and Lodish, H. F. (2001) J. Biol. Chem. 276, 39608-39617[Abstract/Free Full Text]
  40. Awan, A., Lucic, M. R., Shaw, D. M., Sheppard, F., Westwater, C., Lyons, S. A., and Stern, P. L. (2002) Biochem. Biophys. Res. Commun. 290, 1030-1036[CrossRef][Medline] [Order article via Infotrieve]
  41. Tani, T. T., and Mercurio, A. M. (2001) J. Biol. Chem. 276, 36535-36542[Abstract/Free Full Text]
  42. El Mourabit, H., Poinat, P., Koster, J., Sondermann, H., Wixler, V., Wegener, E., Laplantine, E., Geerts, D., Georges-Labouesse, E., Sonnenberg, A., and Aumailley, M. (2002) Matrix Biol. 21, 207-214[CrossRef][Medline] [Order article via Infotrieve]
  43. Rousset, R., Fabre, S., Desbois, C., Bantignies, F., and Jalinot, P. (1998) Oncogene 16, 643-654[CrossRef][Medline] [Order article via Infotrieve]
  44. Schmitt, J. M., and Stork, P. J. (2000) J. Biol. Chem. 275, 25342-25350[Abstract/Free Full Text]
  45. Zheng, M., Zhang, S. J., Zhu, W. Z., Ziman, B., Kobilka, B. K., and Xiao, R. P. (2000) J. Biol. Chem. 275, 40635-40640[Abstract/Free Full Text]
  46. Lou, X., Yano, H., Lee, F., Chao, M. V., and Farquhar, M. G. (2001) Mol. Biol. Cell 12, 615-627[Abstract/Free Full Text]
  47. Tan, C., Deardorff, M. A., Saint-Jeannet, J. P., Yang, J., Arzoumanian, A., and Klein, P. S. (2001) Development 128, 3665-3674[Abstract/Free Full Text]
  48. Devic, E., Xiang, Y., Gould, D., and Kobilka, B. (2001) Mol. Pharmacol. 60, 577-583[Abstract/Free Full Text]