Categories
Articles

ABBS 2005,37(08):Regulation of EGF-induced ERK/MAPK Activation and EGFR Internalization by G Protein-coupled Receptor Kinase 2

Research

Paper

Pdf file on Synergy

Download Chinese abstract

Acta Biochim Biophys

Sin 2005,37:525-531

doi:10.1111/j.1745-7270.2005.00076.x

Regulation of EGF-induced ERK/MAPK Activation and EGFR

Internalization by G Protein-coupled Receptor Kinase 2

Jingxia GAO1,2*, Jiali LI1,

and Lan MA1*

1

Pharmacology

Research Center, Shanghai Medical College, Fudan University, Shanghai 200032,

China;

2

Department

of Biochemistry, Medical College, Tongji University, Shanghai 200092, China

Received: May 8,

2005

Accepted: May 20,

2005

This work was

supported by the grants from the National Natural Science Foundation of China

(30230130), the Ministry of Science and Technology (2003CB515405,

2005CB522406), the Ministry of Education­ (20020246042), and the Shanghai Municipal

Commissions for Science and Technology and Education (02DJ14020, 02GG01)

*Corresponding

authors:

Lan MA: Tel,

86-21-54237522; Fax, 86-21-54237621; E-mail, [email protected]

Jingxia GAO: Tel,

86-21-65984980; E-mail, [email protected]

Abstract        G protein-coupled

receptor kinases (GRKs) mediate agonist-induced phosphorylation and

desensitization of various G protein-coupled receptors (GPCRs). We investigate

the role of GRK2 on epidermal­ growth factor (EGF) receptor signaling, including

EGF-induced extracellular signal-regulated kinase and mitogen-activated protein

kinase (ERK/MAPK) activation and EGFR internalization. Immunoprecipitation and

immunofluorescence experiments show that EGF stimulates GRK2 binding to EGFR

complex and GRK2 translocating from cytoplasm to the plasma membrane in human

embryonic kidney 293 cells. Western blotting assay shows that EGF-induced

ERK/MAPK phosphorylation increases 1.9-fold, 1.1-fold and 1.5-fold (P<0.05) at time point 30, 60 and 120 min, respectively when the cells were transfected with GRK2, suggesting the regulatory role of GRK2 on EGF-induced ERK/MAPK activation. Flow cytometry experiments­ show that GRK2 overexpression has no effect on EGF-induced EGFR internalization, however, it increases agonist-induced G protein-coupled d opioid receptor internalization by approximately 40% (P<0.01). Overall, these data

suggest that GRK2 has a regulatory role in EGF-induced ERK/MAPK activation, and

that the mechanisms underlying the modulatory role of GRK2 in EGFR and GPCR

signaling pathways are somewhat different at least in receptor internalization.

Key words        G protein-coupled

receptor kinase; receptor tyrosine kinase; epidermal growth factor receptor;

ERK/MAPK; internalization

G protein-coupled receptors (GPCRs) constitute a superfamily­ of

plasma membrane receptors. Members of this family include receptors for many

hormones, neuro­transmitters, chemokines and calcium ion, as well as sensory­

receptors for various odors, and bitter and sweet tastes, so GPCRs play

important roles in a variety of cellular­ functions [1]. Repeated agonist

stimulation triggers­ a negative­ feedback regulatory mechanism that attenuates

GPCR-mediated signal transduction (desensitization). The initial event of GPCR

desensitization­ is the phosphory­lation of GPCR catalyzed by G protein­-coupled

receptor­ kinases (GRKs). GRKs are a family­ of Ser/Thr kinases and can

phosphorylate agonist-activated GPCRs and initiate­ their desensitization and

subsequent­ down-regulation. Thus GRKs are a key modulator­ of GPCR signaling

[2].Receptor tyrosine kinases (RTKs) constitute another family of plasma

membrane receptors. RTKs are primary mediators of physiological cell responses,

such as cell pro­liferation, differentiation, motility and survival [3].

Epidermal­ growth factor receptor (EGFR) belongs to the RTK family. Binding­ of

EGFR with its ligand induces dimerization of EGFR, resulting in

autophosphorylation of their cytoplasmic domains, thus recruiting the Src homo­logy

2 and phosphotyrosine binding domain-containing­ proteins, which subsequently

activates multiple signaling cascades and ultimately induces altered gene

expression in the nucleus [4]. Disregulation of EGFR by overexpression,

mutation or continuous activation of its intrinsic tyrosine kinase is

frequently linked to hyperproliferative diseases such as cancer [5]. Thus EGFR

signaling must be under stringent control. Previous­ studies have demonstrated

that EGFR signaling is modulated­ by tyrosine dephosphorylation [6], receptor

internalization­ and degradation [7,8]. Previous studies have shown that overexpression of GRK2, a member of

GRK family, could attenuate phos­phoinositide hydrolysis, cell chemotaxis and

proliferation evoked via platelet-derived growth factor receptor b (PDGFRb) [9,10], thus

expanding our understanding of the roles GRKs may play. A recent study has

shown that EGF stimulation induces GRK2-EGFR complex formation­ via Gbg– and

Src-dependent mechanisms [11]. But the modulatory role of GRKs on RTK signaling

pathways is limited to signals mediated by PDGFRb to date, moreover, the

mechanisms underlying the modulatory role of GRK2 remains unknown. In the present study, we investigated the regulatory role of GRK2 on

EGF-induced extracellular signal-regulated kinase and mitogen-activated protein

kinase (ERK/MAPK) activation and EGFR internalization.

Materials and Methods

Materials

Human EGF and [D-Pen2,D-Pen5]enkephalin

(DPDPE) were obtained from Sigma Chemical Co. (St. Louis, USA). Modified

Eagle’s medium (MEM) and fetal bovine­ serum­ (FBS) were purchased from Life

Technologies Incorporated­ (Grand Island, USA). Protein A-Sepharose was

obtained­ from Amersham Pharmacia Biotech (Piscataway, USA). Rabbit

anti-phospho and total ERK1/2 were supplied­ by New England Biolabs (Beverley,

USA). Mouse monoclonal­ antibody against GRK2 was kindly provided by Dr. Martin­

OPPERMANN (Georg-August University, G?ttingen, Germany). Mouse monoclonal

antibody­ against DYKDDDDK octapeptide (FLAG) epitope and mouse monoclonal

antibody 12CA5 recognizing influenza­ hemagglutinin­ (HA) epitope were supplied

by Roche Molecular­ Biochemicals (Indianapolis, USA). Fluorescein­

isothiocyanate (FITC)-conjugated goat anti-mouse IgG was purchased from Jackson

Immunoresearch (West Grove, USA).

Cell culture and plasmid transfection

Human embryonic kidney 293 (HEK293) cells were obtained from

American Type Culture Collection (Manassas, USA). HEK293 cells cultured in MEM

containing­ 10% FBS were seeded in 35 mm or 60 mm tissue culture dishes at 0.21?106 cells/dish 20

h before transfection. Plasmids encoding bovine GRK2, GRK2-GFP (green

fluorescence protein), human FLAG-tagged EGFR and mouse HA-tagged d opioid receptor

(DOR, one kind of GPCR) were prepared as described previously. Plasmids­ (13 mg each) were transfected into the HEK293 cells using the calcium

phosphate/DNA co-precipitation method as described previously. Experiments were

performed 4448 h after transfection and

the cells were maintained overnight in FBS-free medium before the experiments.Human embryonic kidney 293 (HEK293) cells were obtained from

American Type Culture Collection (Manassas, USA). HEK293 cells cultured in MEM

containing­ 10% FBS were seeded in 35 mm or 60 mm tissue culture dishes at 0.21?106 cells/dish 20

h before transfection. Plasmids encoding bovine GRK2, GRK2-GFP (green

fluorescence protein), human FLAG-tagged EGFR and mouse HA-tagged d opioid receptor

(DOR, one kind of GPCR) were prepared as described previously. Plasmids­ (13 mg each) were transfected into the HEK293 cells using the calcium

phosphate/DNA co-precipitation method as described previously. Experiments were

performed 4448 h after transfection and

the cells were maintained overnight in FBS-free medium before the experiments.

Co-immunoprecipitation and Western blotting

HEK293 cells were incubated at 37 ?C in the presence or absence of

100 ng/ml EGF for 5 min, then the cells were washed twice with ice-cold

phosphate buffered saline­ and lysed in 800 ml ice-cold NP-40

solubilization buffer (250 mM NaCl, 50 mM HEPES, 0.5% NP-40, 10% glycerol, 2 mM

EDTA, pH 8.0, 1 mM Na3VO4, 10 mg/ml aprotinin,

10 mg/ml

benzamidine and 0.2 mM PMSF) as described previously [12] for 1.5 h. The lysate

was centrifuged, and the supernatant was incubated with 1 mg of anti-FLAG

antibody and 15 ml of 50% slurry of protein A-Sepharose beads at 4 ?C for 16 h. The

beads were subsequently­ washed three times with NP-40 solubilization­ buffer.

The proteins bound to the beads were eluted using­ the SDS-PAGE sample buffer

and separated by SDS-PAGE. The presence of EGFR and GRK2 in the immunocomplexes

was detected in the subsequent Western­ blotting with antibody specifically

against FLAG epitopes and GRK2 respectively. The immunoblots were visualized­

using an enhanced chemiluminescence (ECL) kit (Amersham Biosciences) following

the manufacturer’s suggested protocol. An aliquot (2.5%) of the cell lysate was

analyzed by Western blotting to quantify the expression­ level of the protein

studied.

Laser confocal fluorescence microscopy

HEK293 cells were transfected with plasmid encoding GRK2-GFP. For

real-time fluorescence analysis of GRK2-GFP in living cells, the fluorescence

was observed under a microscope equipped with a temperature controller at 37

?C. The EGF was applied directly over the selected cells. The image scanned

before EGF application represented­ GRK2-GFP distribution in cells. After EGF

treatment, the same cells were scanned again in a time series. Scanning images­

were recorded with a TCS NT laser confocal microscope­ (Leica Microsystems,

Bensheim, Germany).

Quantitation of receptor internalization by fluorescence flow

cytometry assay

Receptor internalization was quantitated using fluo­rescence flow

cytometry assay. Briefly, stably transfected HEK293 cells were chilled on ice

after agonist stimulation and the surface receptors were labeled with

corresponding­ antibody for 1 h at 4 ?C. After sufficient washing, the cells

were incubated with FITC-conjugated goat anti-mouse antibody for 1 h at 4 ?C.

The cells were then collected­ and fixed and the surface receptor staining

intensity­ was analyzed using FACScalibur flow cytometry (Becton Dickenson,

Mountain View, USA). Basal cell fluorescence­ intensity was determined with

cells stained with the secondary­ antibody alone.

Statistical analysis

Data were analyzed using Student’s t-test for comparison of

independent means with pooled estimates of common variances.

Results

EGF stimulates GRK2-EGFR complex formation in HEK293 cells

HEK293 cells expressing FLAG-tagged EGFR and GRK2 or GRK2 alone were

incubated in the presence or absence of 100 ng/ml EGF, then FLAG-EGFR was

immuno­precipitated with specific anti-FLAG antibody. As shown in Fig. 1(A),

there was little GRK2 in the EGFR immunoprecipitation complex before EGF

stimulation. After EGF stimulation there was a large amount of GRK2 detected in

the EGFR immunoprecipitaion complex using GRK-specific antibody. In the cells

expressing GRK2 alone there was no GRK2 detected in the EGFR immunoprecipitation

complex upon EGF stimulation [Fig. 1(A), upper panel]. This result

indicates that the detected GRK2 in the EGFR immunoprecipitation complex was

specific. Direct Western blot analysis of the total cell lysate detecting­

FLAG-tagged EGFR and GRK2 expression [Fig. 1(A), lower panel] was shown

to ensure similar expression levels. These results are in accordance with our

previous study [11], and clearly demonstrate that EGF stimulates GRK2-EGFR

complex formation in HEK293 cells overexpressing GRK2 and EGFR.

To further demonstrate that EGF stimulates GRK2-EGFR complex

formation, we observed the subcellular redistribution of GRK upon EGF

stimulation in HEK293 cells transiently expressing GRK2-GFP using a laser

confocal­ microscope. As shown in Fig. 1(B), the green fluorescence

representing GRK2 mainly resided in the cytoplasm­ before EGF stimulation. The

real-time recording­ of GRK2-GFP fluorescence images in living cells showed that

after EGF stimulation the GRK2-GFP fluorescence increased quickly on the

membrane. At the same time GRK2-GFP fluorescence decreased in the cytoplasm,

and this redistribution was accompanied by changes in membrane­ shape [Fig.

1(B), 3 min and 5 min]. The redistribution­ of GRK2-GFP was restored to the

basal state at 10 min of EGF stimulation [Fig. 1(B), 10 min]. These results demonstrated that EGF stimulation could induce

translocation of GRK2 from cytoplasm to the plasma membrane and form a complex

with EGFR on the membrane in HEK293 cells overexpressing GRK2 and EGFR.

Overexpression of GRK2 enhances EGF-stimulated ERK/MAPK activation

EGFR activation leads to activation of the ERK/MAPK pathway. To demonstrate

whether GRK-EGFR complex formation upon EGF stimulation leads to modulation of

EGFR signaling, we observed the effect of GRK2 on EGF-stimulated ERK/MAPK

phosphorylation. HEK293 cells overexpressing GRK2 or b-Gal were incubated in the

presence­ or absence of 10 ng/ml EGF for a period ranging from 0 min to 120

min. Phospho-ERK/MAPK and total ERK/MAPK were probed employing phospho-specific

and total ERK/MAPK antibody. Western blotting analysis showed that

phospho-ERK/MAPK was detected at 2 min of EGF stimulation and reached its

maximum at 5 min, then it gradually decreased. Total ERK/MAPK did not show

detectable change before or after EGF stimulation. Phospho-ERK/MAPK in GRK2

transfected cells was significantly increased compared with b-Gal transfected

controls, although the time course of phospho-ERK/MAPK was similar [Fig.

2(A)]. The resulting phospho-ERK/MAPK levels from four independent sets of

experiments were quantified nor­malizing with total ERK as a loading control.

The mean values­ are presented graphically in Fig. 1(B).

Phospho-ERK/MAPK was enhanced 1.9-fold, 1.1-fold and 1.5-fold respectively (P<0.05) in GRK2 transfected cells compared with b-Gal transfected controls

at time point 30 min, 60 min and 120 min [Fig. 2(B)].

The effect of GRK2 overexpression on EGF-induced EGFR

internalization and DPDPE-induced DOR internalization­

Agonist-induced EGFR internalization is a critical regulatory

mechanism in EGFR signaling. EGFR internalization induces receptor

down-regulation by decreasing the amount of EGFR present on the plasma

membrane. To determine whether the modulatory role of GRK2 on EGFR signaling is

through affecting EGFR internalization, the following experiments were carried

out. First we constructed a stable HEK293 cell line expressing FLAG-tagged

EGFR, then we used flow cytometry to quantitatively determine­ the

characteristics of EGF-induced EGFR internalization­ and the effect of GRK2

overexpression on the receptor internalization. Cell surface EGFR declined

gradually after EGF stimulation. At 30 min of EGF stimulation there was about

70% of EGFR left on the cell surface­ compared with the cells left untreated,

indicating that about 30% of cell surface EGFR was internalized into cytoplasm

(data not shown). This data was in accordance­ with a previous study [13],

suggesting EGFR was sequestered from plasma membrane in response to EGF

stimulation. Then, overexpression of GRK2 on EGFR and G protein-coupled DOR

internalization was examined. Fig. 3(A) shows representative results

from a set of experiments. Stimulation of cells with indicated agonist led to

the reduction of both the percentage of the positive cells and the mean

fluorescence density. Quantitatively, overexpression of GRK2 in HEK293 cells

stably expressing FLAG-EGFR did not have any significant effect on EGF-induced

EGFR internalization (P=0.96) compared with control cells overexpressing

b-Gal

[Fig. 3(B)]. But in an HEK293 cell line stably expressing DOR, DOR

internalization induced by its agonist DPDPE was enhanced by approximately 40%

(P<0.01) when the cells were transfected with GRK2 compared with control cells transfected with b-Gal [Fig. 3(B)]. To exclude the possible­ effect of high

density of plasma membrane EGFR on EGFR internalization and the possible

saturability of the EGFR internalization pathway [14], we measured the

internalization­ of endogenously expressed EGFR in HEK293 cells and HeLa cells.

GRK2 overexpression had no significant­ effect on EGF-induced EGFR

internalisation either, although the internalization of endogenously expressed­

EGFR upon EGF stimulation was more rapid and to a greater degree (data not

shown).

Discussion

Previous studies have shown that GRK2 plays a role in the negative regulation

of signaling pathways mediated by PDGFRb, besides its classical

role in phosphorylating and desensitizing agonist-activated GPCRs. In the

current study we investigated the role of GRK2 in the EGFR signaling pathway,

including ERK/MAPK activation induced by EGFR activation and the role of GRK2

in EGF-induced EGFR internalization, as well as the possible mechanisms

underlying it. Our results have shown that overexpression of GRK2 enhances

ERK/MAPK activation induced by EGF stimulation. EGF stimulation induces GRK2

translocation from cytoplasm to the plasma membrane and GRK-EGFR complex

formation. But overexpression of GRK2 had no significant effect on EGF-induced

EGFR internalization; however, agonist-induced DOR internalization increased

significantly.Our present and previous studies have shown that EGF stimulates

GRK2-EGFR complex formation. There are several lines of evidence to support

this. First, co-immunoprecipitation­ experiment showed that there was a large

amount of GRK2 in EGFR immunoprecipitation complex­ upon EGF stimulation.

Second, confocal laser micro­scopy experiment showed that GRK2-GFP translocated­

from cytoplasm to plasma membrane in a real-time confocal fluorescence record

for living cells. The mechanisms­ by which GRK activity is regulated can be

divided into three categories: subcellular localization, alterations in

intrinsic kinase activity and alterations in GRK expression­ level [15,16]. Our

results showed that GRK2 translocated­ to plasma membrane upon EGF stimulation,

indicating­ that upon EGFR activation GRK2 was also activated. Previous studies­

have demonstrated that GRK2 exhibits a primarily cytosolic distribution in

unstimulated cells and appear to translocate to the plasma membrane upon GPCR

activation­ [15,16]. Our results are in accordance­ with this, suggesting­ that

GRK2 may exert its modulatory role in EGFR signaling­ via mechanisms similar to

GPCRs.EGFR activation leads to its dimerization and autophosphorylation of

the cytoplasmic domains of EGFR. Adaptor proteins such as SHC bind to the

phospho-tyrosine­ residues and subsequent formation of an SHC-Grb2-Sos complex

and induction of Raf function, thus, the Ras/MAPK pathway was activated. This

cascade couples agonist­ stimulation to gene transcription [4]. The current

study has shown that overexpression of GRK2 enhances EGF-induced ERK/MAPK

activation, suggesting that, in contrast­ to its negative regulation of GRK2 on

PDGFRb signaling, GRK2 may exert a positive regulation on EGF-induced

ERK/MAPK phosphorylation. Phospho-ERK/MAPK can enter the nucleus and

phosphorylate transcription­ factors such as Elk-1 [17], so enhanced ERK/MAPK

activation by GRK2 overexpression may also lead to changes in gene

transcription mediated by EGFR activation.GRK-catalyzed GPCR phosphorylation leads to GPCR desensitization and

subsequent internalization and degradation, thus GRK plays an important role in

GPCR internalization. The present study showed that overexpression of GRK2 had

no effect on EGF-induced EGFR internalization. However, overexpression of GRK2

significantly enhanced agonist-induced DOR internalization. Ligand-induced EGFR

internalization requires­ intrinsic receptor tyrosine kinase activity [18] and

specific sequences in the carboxyl-terminus of the receptor distal to the

kinase domain. Some adaptors, such as the m2 subunit of the AP2

protein recognize the endocytic signals thus involved in receptor endocytosis

and recycling­ [19]. The GRK2 binding domain on EGFR and the possible­

phosphorylation sites on EGFR catalyzed by GRK2 remain­ elusive. We presume

that EGF-induced GRK2 binding to EGFR does not affect the specific sequences

involved in EGFR endocytosis due to the long distance between the binding

domain or phosphorylation sites and the endocytic sequences, thus, EGF-induced

EGFR internalization was unaffected by GRK2 overexpression. Taken together, these data demonstrate in HEK293 cells

overexpressing GRK2 and EGFR that GRK2 has the regulatory­ role in EGF-induced

ERK/MAPK activation. However, EGF-induced EGFR internalization is not affected,

suggesting that the mechanisms underlying the modulatory role of GRK2 in EGFR

and in GPCR signaling pathways is somewhat different, at least in receptor

internalization.

Acknowledgements

We thank Yanqin GAO, Hui GAO and Yalin HUANG for their technical

assistance. We also thank Xiaoqing ZHANG and Min ZHU for their helpful

discussion.

References

 1   Ji TH, Grossmann M, Ji I. G

protein-coupled receptors. I. Diversity of receptor-ligand interactions. J Biol

Chem 1998, 273: 1729917302

 2   Penela P, Ribas C, Mayor F Jr.

Mechanisms of regulation of the expression and function of G protein-coupled

receptor kinases. Cell Signal 2003, 15: 973981

 3   Schlessinger J. Cell signaling by

receptor tyrosine kinases. Cell 2000, 103: 211225

 4   Zwick E, Hackel PO, Prenzel N,

Ullrich A. The EGF receptor as central transducer of heterologous signalling

systems. Trends Pharmacol Sci 1999, 20: 408412

 5   Holbro T, Civenni G, Hynes NE. The

ErbB receptors and their role in cancer progression. Exp Cell Res 2003, 284: 99110

 6   Ostman A, Bohmer FD. Regulation of

receptor tyrosine kinase signaling by protein tyrosine phosphatases. Trends

Cell Biol 2001, 11: 258266

 7   Vieira AV, Lamaze C, Schmid SL.

Control of EGF receptor signaling by clathrin-mediated endocytosis. Science

1996, 274: 20862089

 8   Wiley HS.Trafficking of the ErbB

receptors and its influence on signaling. Exp Cell Res 2003, 284: 7888

 9   Peppel K, Jacobson A, Huang X,

Murray JP, Oppermann M, Freedman NJ. Overexpression of G protein-coupled

receptor kinase-2 in smooth muscle cells attenuates mitogenic signaling via G

protein-coupled and platelet-derived growth factor receptors. Circulation 2000,

102: 793799

10  Peppel K, Zhang L, Huynh TT, Huang X,

Jacobson A, Brian L, Exum ST et al. Overexpression of G protein-coupled

receptor kinase-2 in smooth muscle cells reduces neointimal hyperplasia. J Mol

Cell Cardiol 2002, 34: 13991409

11  Gao J, Li J, Chen Y, Ma L. Activation of

tyrosine kinase of EGFR induces Gbg-dependent GRK-EGFR complex formation.

FEBS Lett 2005, 579: 122126

12  Maudsley S, Pierce KL, Zamah AM, Miller

WE, Ahn S, Daaka Y, Lefkowitz RJ et al. The b2-adrenergic

receptor mediates extracellular signal-regulated kinase activation via assembly

of a multi-receptor complex with the epidermal growth factor receptor. J Biol

Chem 2000, 275: 95729580

13  Kim J, Ahn S, Guo R, Daaka Y. Regulation

of epidermal growth factor receptor internalization by G protein-coupled

receptors. Biochemistry 2003, 42: 28872894

14  Wiley HS. Anomalous binding of epidermal

growth factor to A431 cells is due to the effect of high receptor densities and

a saturable endocytic system. J Cell Biol 1988, 107: 801810

15  Pitcher JA, Freedman NJ, Lefkowitz RJ. G

protein-coupled receptor kinases. Annu Rev Biochem 1998, 67: 653692

16  Penn RB, Pronin AN, Benovic JL.

Regulation of G protein-coupled receptor kinases. Trends Cardiovasc Med 2000,

10: 8189

17  Papavassiliou AG. The role of regulated

phosphorylation in the biological activity of transcription factors SRF and

Elk-1/SAP-1. Anticancer Res 1994, 14: 19231926

18  Wiley HS, Herbst JJ, Walsh BJ,

Lauffenburger DA, Rosenfeld MG, Gill GN. The role of tyrosine kinase activity

in endocytosis, compartmentation, and down-regulation of the epidermal growth

factor receptor. J Biol Chem 1991, 266: 1108311094

19     Wiley HS. Trafficking of the ErbB receptors

and its influence on signaling. Exp Cell Res 2003, 284: 7888