• Aucun résultat trouvé

Distinct β-arrestin- and G protein-dependent pathways for parathyroid hormone receptor-stimulated ERK1/2 activation

N/A
N/A
Protected

Academic year: 2021

Partager "Distinct β-arrestin- and G protein-dependent pathways for parathyroid hormone receptor-stimulated ERK1/2 activation"

Copied!
10
0
0

Texte intégral

(1)

HAL Id: hal-02667175

https://hal.inrae.fr/hal-02667175

Submitted on 31 May 2020

HAL is a multi-disciplinary open access archive for the deposit and dissemination of sci- entific research documents, whether they are pub- lished or not. The documents may come from teaching and research institutions in France or abroad, or from public or private research centers.

L’archive ouverte pluridisciplinaire HAL, est destinée au dépôt et à la diffusion de documents scientifiques de niveau recherche, publiés ou non, émanant des établissements d’enseignement et de recherche français ou étrangers, des laboratoires publics ou privés.

Distinct β -arrestin- and G protein-dependent pathways for parathyroid hormone receptor-stimulated ERK1/2

activation

Diane Gesty-Palmer, Minyong Chen, Eric Reiter, Seungkirl Ahn, Christopher D. Nelson, Shuntai Wang, Allen E. Eckhardt, Conrad L. Cowan, Robert F.

Spurney, Louis M. Luttrell, et al.

To cite this version:

Diane Gesty-Palmer, Minyong Chen, Eric Reiter, Seungkirl Ahn, Christopher D. Nelson, et al.. Dis- tinct β-arrestin- and G protein-dependent pathways for parathyroid hormone receptor-stimulated ERK1/2 activation. Journal of Biological Chemistry, American Society for Biochemistry and Molec- ular Biology, 2006, 281 (16), pp.10856-10864. �10.1074/jbc.M513380200�. �hal-02667175�

(2)

Distinct-Arrestin- and G Protein-dependent Pathways for Parathyroid Hormone Receptor-stimulated

ERK1/2 Activation

*

Received for publication, December 15, 2005, and in revised form, February 13, 2006 Published, JBC Papers in Press, February 21, 2006, DOI 10.1074/jbc.M513380200

Diane Gesty-Palmer‡§, Minyong Chen, Eric Reiter, Seungkirl Ahn, Christopher D. Nelson, Shuntai Wang, Allen E. Eckhardt, Conrad L. Cowan, Robert F. Spurney, Louis M. Luttrell**‡‡, and Robert J. Lefkowitz‡1 From theDepartments of Medicine and Biochemistry, Howard Hughes Medical Institute, Duke University, Medical Center, Durham, North Carolina 27710, the§Geriatrics Research, Education and Clinical Center, Durham Veterans Affairs Medical Center, Durham, North Carolina 27705, theInstitut National de la Recherche Agronomique, 37380 Nouzilly, France, theXsira

Pharmaceuticals, Morrisville, North Carolina 27709, the**Departments of Medicine and Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425, and the‡‡Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401

Parathyroid hormone (PTH) regulates calcium homeostasisvia the type I PTH/PTH-related peptide (PTH/PTHrP) receptor (PTH1R). The purpose of the present study was to identify the con- tributions of distinct signaling mechanisms to PTH-stimulated activation of the mitogen-activated protein kinases (MAPK) ERK1/2. In Human embryonic kidney 293 (HEK293) cells tran- siently transfected with hPTH1R, PTH stimulated a robust increase in ERK activity. The time course of ERK1/2 activation was biphasic with an early peak at 10 min and a later sustained ERK1/2 activation persisting for greater than 60 min. Pretreatment of HEK293 cells with the PKA inhibitor H89 or the PKC inhibitor GF109203X, indi- vidually or in combination reduced the early component of PTH- stimulated ERK activity. However, these inhibitors of second mes- senger dependent kinases had little effect on the later phase of PTH- stimulated ERK1/2 phosphorylation. This later phase of ERK1/2 activation at 30 – 60 min was blocked by depletion of cellular-ar- restin 2 and␤-arrestin 1 by small interfering RNA. Furthermore, stimulation of hPTH1R with PTH analogues, [Trp1]PTHrp-(1–36) and [D-Trp12,Tyr34]PTH-(7–34), selectively activated Gs/PKA-me- diated ERK1/2 activation or G protein-independent/-arrestin-de- pendent ERK1/2 activation, respectively. It is concluded that PTH stimulates ERK1/2 through several distinct signal transduction pathways: an early G protein-dependent pathway meditated by PKA and PKC and a late pathway independent of G proteins mediated through-arrestins. These findings imply the existence of distinct active conformations of the hPTH1R responsible for the two path- ways, which can be stimulated by unique ligands. Such ligands may have distinct and valuable therapeutic properties.

The type I PTH/PTH-related peptide receptor (PTH1R),2a seven- transmembrane receptor (7TMR) highly expressed in the kidney and

bone, plays a fundamental role in the regulation of calcium homeostasis, as well as in bone formation and resorption. Ligands for PTH1R includ- ing PTHrp and PTH are involved in the etiology and treatment of dis- ease processes such as hypercalcemia of malignancy and osteoporosis.

The actions of PTH, however, are complex. PTH is known for both anabolic and catabolic effects on bone, which are dependent upon inter- mittent or persistent exposure, respectively (1–3). The mechanistic basis of these effects on bone remodeling are not well understood.

The intracellular signaling pathways activated by PTH and PTHrP via the PTH1R receptor include Gs-mediated activation of adenylate cyclase, resulting in cAMP production and PKA activation, and Gq/11- mediated PLC␤ stimulation, leading to inositol 1,4,5-trisphosphate (IP3) production, calcium mobilization, and PKC activation (4 –7). It has also been demonstrated that PTH activates the Raf-MEK-ERK MAP kinase (MAPK) cascade through both PKA and PKC in a cell-specific and G protein-dependent manner (8 –10). MAPKs activated in response to stimulation by many different classes of cell surface recep- tors, including growth factor receptor tyrosine kinases and 7TMRs, reg- ulate cell growth, division, differentiation, and apoptosis (11). PTH- stimulated activation of MAPK is known to have proliferative effects in kidney and bone (12, 13).

There is growing evidence for novel 7TMR signaling mechanisms that are distinct from the classical G protein second messenger-depend- ent pathways. One such mechanism involves␤-arrestins, a small family of cytosolic proteins initially identified for their central role in 7TMR desensitization.␤-Arrestins are recruited to agonist-occupied 7TMRs that have been phosphorylated by specialized G protein-coupled recep- tor kinases (GRKs) and sterically inhibit receptor-G protein coupling resulting in homologous receptor desensitization. Additionally,␤-ar- restins act as adaptors in clathrin-mediated receptor endocytosis (14, 15). The role of␤-arrestins acting as signal transducers through the formation of scaffolding complexes with accessory effector molecules such as Src, Ras, ERK1/2, JNK3, and MAPK kinase 4 (MKK4) is becom- ing increasingly recognized (16 –20).

The potential signaling diversity of 7TMRs suggests the possible existence of multiple discrete “active” receptor conformations. This implies that specific ligands might direct distinct signaling responses by preferentially stabilizing one or more of these active conformations. In the simple two-state model of receptor activation, agonists are defined as drugs that stabilize the active receptor conformation, which in turn promotes G protein activation. Conversely, an inverse agonist preferen- tially binds to the inactive receptor conformational state thereby reduc-

*This work was supported by National Institutes of Health Grants HL16037, HL70631, DK64353, and HD043446. 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.

1Investigator with the Howard Hughes Medical Institute. To whom correspondence should be addressed: Howard Hughes Medical Inst., Duke University Medical Center, Box 3821, Durham, NC 27710. Tel.: 919-684-2974; Fax: 919-684-8875; E-mail:

lefko001@receptor-biol.duke.edu.

2The abbreviations used are: PTH, parathyroid hormone; PTH1R, type I PTH/PTH-related peptide receptor; 7TMR, seven-transmembrane receptor; GRK, G protein-coupled receptor kinase; GFX, GF109203X; IP3, inositol 1,4,5-trisphosphate; PI, phosphoinosit- ide; PTH-NBR, [Trp1]PTHrp-(1–36); PTH-IA, [D-Trp12,Tyr34]PTH-(7–34); ERK, extracellu- lar signal-regulated kinase; MAP, mitogen-activated protein; MAPK, MAP kinase;

siRNA, small interfering RNA; WT, wild type; ANOVA, analysis of variance.

at INRA Institut National de la Recherche Agronomique, on November 8, 2010www.jbc.orgDownloaded from

(3)

ing G protein signaling (21). Recent observations suggest that some inverse agonists for cAMP generation may none the less be capable of recruiting␤-arrestin to the receptor and inducing biological effects (22).

Agonist stimulation of PTH1R promotes the translocation of both

␤-arrestin 1 and␤-arrestin 2 to the plasma membrane, the association of the receptor with␤-arrestins, and the internalization of the receptor/

␤-arrestin complexes (23–25).␤-Arrestin 2 has been shown to influ- ence bone remodeling and the anabolic effects of intermittent PTH-(1–

34) administration in murine models (26, 27). The mechanistic basis of these physiologic effects has not been established. Accordingly, we set out to determine the roles of G proteins and␤-arrestins in PTH1R- stimulated ERK1/2 activation.

EXPERIMENTAL PROCEDURES

Materials—Human PTH-(1–34) and [D-Trp12,Tyr34]PTH-(7–34) (PTH-IA) were obtained from Bachem (Philadelphia, PA).

[Trp1]PTHrp-(1–36) (PTH-NBR) was obtained from Xsira Pharmaceu- ticals (Research Triangle Park, NC). H89 and GF109203X (GFX) were purchased from Calbiochem (La Jolla, CA). GeneSilencer and FuGENE 6 transfection reagents were from Gene Therapy Systems (San Diego, CA) and Roche Applied Science, respectively.

cDNA Constructs—Previously described expression plasmids encod- ing the wild type human PTH1R and H223R mutant PTH1R were gen- erous gifts from Ernestina Shippani. To facilitate the PTH1R immuno- preciptitation, a FLAG epitope tag was introduced using Exsite PCR- based site-directed mutagenesis (Stratagene). The amino acid sequence EKRLK located in the downstream of the predicted PTH1R signal pep- tide was replaced by the FLAG epitope sequence DYKDDDDK. The expression vectors for His-tagged-␤-arrestin 1 and His-tagged-␤-arres- tin 2 vectors have been described previously (28)

Cell Culture and Transfection—HEK293 cells were grown in Eagle’s minimum essential medium with Earle’s salts supplemented with 10% (v/v) FBS and a 1:100 dilution of a penicillin/streptomycin mixture (Sigma). Cells were transiently transfected with 1 ␮g of pcDNA1-PTH1R using FuGENE 6 reagent (Roche Applied Science) according to the manufacturer’s instructions. Binding studies with radiolabeled human125I-[Nle8,18,Tyr34]PTH-(1–34) amide (Amer- sham Biosciences) show PTH1R receptor expression of 100 –200 fmol/mg of protein.

siRNA Transfection—The double-stranded siRNA sequences indi- vidually targeting␤-arrestin 1 and␤-arrestin 2 are 5⬘-AAAGCCUUC- UGCGCGGAGAAU-3⬘ and 5⬘-AAGGACCGCAAAGUGUUUGUG- 3⬘, corresponding to the positions 439 – 459 and 148 –168 relative to the start codon, respectively. The double-stranded siRNA sequence 5⬘-AC- CUGCGCCUUCCGCUAUG-3⬘ was used to simultaneously target

␤-arrestin 1 and␤-arrestin 2. This sequence corresponds to the posi- tions 172–190 and 175–193 relative to the start codons of␤-arrestin 1 and ␤-arrestin 2, respectively. The chemically synthesized double- stranded siRNAs and a non-silencing control RNA duplex were pur- chased from Xeragon (Germantown, MD) in deprotected and desalted form. HEK293 cells were transfected simultaneously with 2 ␮g of pcDNA1-PTH1R and 1.44 nmol of␤-arrestin 2 siRNA,␤-arrestin 1 siRNA or control siRNA using the GeneSilencer Transfection reagent as previously described (29, 30).

Preparation of Cellular Extracts and Immunoblotting—Transfected HEK293 cells were starved for 12–18 h in serum-free medium prior to stimulation. After stimulation, media was removed, and 100␮l of 2⫻

Laemmli sample buffer was added to each well. Whole cell lysates were sonicated, resolved on 4 –20% (for ERK1/2) or 10% (for␤-arrestins 1 and 2) Tris/glycine polyacrylamide gels (Invitrogen), and transferred to

nitrocellulose membranes for immunoblotting. Phosphorylated ERK1/2, total ERK1/2, and␤-arrestins were detected by immunoblot- ting with rabbit polyclonal anti-phospho-p44/42 MAPK (Cell Signaling, 1:2,000 Beverly, MA), anti-MAP kinase 1/2 (Upstate Technology Inc, 1:10,000, Lake Placid, NY), or rabbit polyclonal anti-␤-arrestin (A1CT, 1:5,000) antibodies, respectively. Chemiluminescent detection was per- formed using the SuperSignal Western Pico reagent (Pierce). Immuno- blots were quantified by densitometry with a Fluor-S MultiImager (Bio-Rad).

Cyclic AMP Determination—PTH-stimulated cAMP levels were determined in HEK293 cells transiently transfected with either the wild type human PTH1R or the constitutively active H223R mutant. Cells were preincubated for 15 min in minimum essential medium supple- mented with 10 mMHEPES (pH 7.4) and 1 mMisobutylmethylxanthine and stimulated for 15 min with PTH-(1–34), PTH-NBR, or PTH-IA.

Forskolin (10M) was used as a positive control. The stimulation was terminated with the addition of 0.125MEDTA, and the samples were boiled for 10 min and clarified by centrifugation for 1 min at 15,000g.

cAMP levels were determined using a 3H-labeled cAMP assay as described previously (31). The results are expressed as a percent maxi- mum stimulation normalized to forskolin.

Coimmunoprecipitation Assays—Transfected HEK293 cells were stimulated with PTH-(1–34), PTH-NBR, or PTH-IA at 37 °C for 5 min.

Cells were subjected to covalent protein cross-linking by using the membrane-permeable, hydrolyzable cross-linker dithiobis(succinimi- dyl propionate) (Pierce) and then solubilized in RIPA lysis buffer (150 mMNaCl, 50 mMTris, 5 mMEDTA, 1% Nonidet P-40, 1 mMsodium orthovanadate, 1 mMsodium fluoride, 1 mMphenylmethylsulfonyl flu- oride, 5␮g/ml leupeptin, 5␮g/ml aprotinin, 1␮g/ml pepstatin A, 100

Mbenzaminidine). Lysates were clarified by centrifugation and immu- noprecipitation was performed using 25␮l of 50% slurry of monoclonal anti-FLAG affinity agarose (Sigma), with constant agitation overnight at 4 °C. Immune complexes were washed three times with lysis buffer and incubated in Laemmli sample buffer at 37 °C for 30 min prior to SDS- PAGE. Coimmunoprecipitated ␤-arrestin with FLAG-PTH1R were detected by immunoblotting with a 1:3,000 dilution of the A1CT rabbit polyclonal anti-␤-arrestin antibody.

RESULTS

Inhibition of PKA and PKC Reveals Distinct Temporal Components of PTH-induced MAP Kinase Activation—HEK293 cells transiently trans- fected to express human PTH1R stimulated with PTH-(1–34) (100 nM) resulted in a time-dependent increase in MAP kinase activation (Fig. 1).

There was an early rapid activation of ERK1/2 at 2–5 min. PTH-(1–34)- stimulated ERK1/2 activation reached a peak at 5 min and remained greater than 8-fold over basal for at least 60 min (Fig. 1A). To determine the contributions of Gs/PKA- and Gq/PKC-dependent mechanisms to MAP kinase activation, cells were pretreated with the PKA inhibitor H89 (20M) or the PKC inhibitor GFX (2.5M), respectively, for 15 min prior to stimulation with PTH-(1–34) (100 nM). Each of these treat- ments inhibited MAP kinase activation. The greatest effect on the time course of ERK phosphorylation was apparent at 2–5 min of stimulation.

In contrast, the sustained ERK1/2 activation at 30 – 60 min after PTH stimulation was significantly less sensitive to H89 or GFX (Fig. 1,Band C). To determine the contribution of a putative MAP kinase signaling pathway independent of both PKA and PKC, cells were pretreated with a combination of H89 (20M) and GFX (2.5M) for 15 min prior to stimulation with PTH-(1–34) (100 nM) (Fig. 1D). These results indicate that PTH stimulation of PTH1R results in MAP kinase activation inde- pendent of PKA and PKC. As illustrated in Fig. 1E, MAP kinase activa-

-Arrestin- and G Protein-mediated ERK1/2 Activation by PTH1R

at INRA Institut National de la Recherche Agronomique, on November 8, 2010www.jbc.orgDownloaded from

(4)

FIGURE 1.Inhibition of PKA or PKC reduces PTH-stimulated ERK1/2 activation.A, phosphorylation of endogenous ERK1/2 in HEK293 cells transiently transfected to express human PTH1R stimulated with PTH-(1–34) (100 nM) for the indicated periods of time in the absence or presence of H89 (20M) or GFX (2.5M) was determined by immunoblotting (IB).B–D, pretreatment with H89, GFX, or combined H89 and GFX alters the time course of PTH-(1–34)-stimulated ERK1/2 activation. Values are expressed as a percent of the ERK1/2 phosphorylation obtained at 5 min of stimulation with PTH-(1–34) in the absence of inhibitors. Data represent the meanS.E. from at least five independent experiments. Statistical comparison of the curves was performed by using a two-way ANOVA between PTH-(1–34)-stimulated cells in the absence and presence of inhibitors (†,p0.0001), and Bonferroni post-tests were used to compare values at each of the time points (***,p0.001; **,p0.01; *,p0.05).E, G protein-dependent and G protein-independent components of ERK1/2 activation (shaded). The G protein-dependent component inEwas generated by subtracting the data obtained from the PTH-(1–34)-stimulated cells that were pretreated with both H89 and GFX (Fig. 1D) from the data points obtained from the PTH-(1–34)-stimulated cells under control conditions. The G protein-independent component of ERK1/2 activation is the time course obtained from the stimulation of PTH1R with PTH-(1–34) in the presence of combined inhibitors for PKA and PKC.DMSO, dimethyl sulfoxide.

at INRA Institut National de la Recherche Agronomique, on November 8, 2010www.jbc.orgDownloaded from

(5)

FIGURE 2.Inhibition of PTH-stimulated ERK1/2 activation by␤-arrestin siRNA.HEK293 cells transiently transfected to express human PTH1R and the indicated siRNAs were stimulated with 100 nMPTH-(1–34) for the indicated periods.A, the effect of siRNA transfection on the expression of␤-arrestin 1 and␤-arrestin 2 was visualized by immunoblot (IB).

B, phosphorylation of endogenous ERK1/2 was determined by immunoblot.C–E, the effect of siRNA transfection targeting␤-arrestin 1,␤-arrestin 2 or combined␤-arrestin 1 and

␤-arrestin 2 on PTH-(1–34)-stimulated ERK/12 activation. The values in the graphs are expressed as percent of the phosphorylation of ERK1/2 obtained at 5 min of stimulation in control (CTL) siRNA-transfected cells. Data represent the meanS.E. from at least five independent experiments. Statistical comparison of the curves was performed by using a two-way ANOVA between␤-arrestin siRNA-transfected and control siRNA-transfected cells (†,p⬍0.0001) and Bonferroni post-tests were used to compare values at each of the time points (***,p0.001; **,p0.01; *,p0.05).F, the time course PTH-stimulated ERK1/2 activation in cells with reduced expression of␤-arrestin 1 and␤-arrestin 2 overlays the predicted G protein-depenedent ERK1/2 activation (shaded) shown in Fig. 1E.

-Arrestin- and G Protein-mediated ERK1/2 Activation by PTH1R

at INRA Institut National de la Recherche Agronomique, on November 8, 2010www.jbc.orgDownloaded from

(6)

tion is comprised of an early G protein-dependent component and a late G protein-independent component. Inhibition with H89 and GFX, indi- vidually and combination, demonstrates that the Gs/PKA- and Gq/PKC- dependent mechanisms contribute primarily to the early time compo- nent of MAP kinase activation. Subtraction of the time course obtained from the stimulation of PTH1R with PTH-(1–34) in the presence of combined inhibitors for PKA and PKC (Fig. 1D) from the curve obtained in the absence of inhibitors predicts the time course for the G protein-dependent activation of ERK1/2. The G protein independent component of MAP kinase activation is the time course obtained from the stimulation of PTH1R with PTH-(1–34) in the presence of com- bined inhibitors for PKA and PKC (Fig. 1E).

The Late Component of ERK1/2 Activation by PTH1R Is Dependent upon␤-Arrestin 1 and␤-Arrestin 2—To determine whether␤-arrestins contribute to PTH-mediated MAP kinase activation, we used RNA interference to reduce the expression of endogenous␤-arrestin 1 or

␤-arrestin 2 in HEK293 cells and measured MAP kinase activation by PTH1R stimulated with PTH-(1–34). Fig. 2Ashows that siRNA target- ing␤-arrestin 1 or ␤-arrestin 2 effectively silences their expression (⬎90%) with little cross-reativity. In control siRNA transfected cells stimulated with PTH-(1–34) (100 nM), ERK1/2 activation reaches max- imal levels rapidly (within 5 min of PTH treatment) and persists, decreasing slowly over 60 min (Fig. 2,BandC). The cotransfection of either␤-arrestin 1 or␤-arrestin 2 siRNA with PTH1R in HEK293 cells stimulated with PTH-(1–34) significantly alters the time course of ERK1/2 activation. PTH-(1–34) stimulation in absence of either␤-ar- restin 1 or␤-arrestin 2 leads to rapid and transient MAP kinase activa- tion, which decreases after 5 min and reaches close to basal levels at 30 min (Fig. 2,B–D). Furthermore, the decreased expression of either␤-ar- restin 1 or␤-arrestin 2 resulted in a decrease in ERK1/2 activity similar to that observed with the decreased expression of both␤-arrestin iso- forms combined (Fig. 2E).

Taken together with the PKA and PKC inhibitor data above, these results suggest that the␤-arrestin-mediated ERK1/2 activation follows a time course that is very different from the G protein-mediated PKA/

PKC-dependent ERK1/2 activation. Specifically, G protein (Gs, Gq)-de- pendent activity is rapid in onset and transient, whereas the majority of

␤-arrestin-dependent activity contributes to the late component of ERK1/2 activation and is more prolonged. Consistent with the idea that the time course of PTH-stimulated ERK1/2 activity is composed of independent G protein-mediated and ␤-arrestin-mediated compo- nents, Fig. 2F shows that the G protein-dependent component of ERK1/2 activation observed in the absence␤-arrestin 1 and␤-arrestin 2 overlays the predicted time course of G protein-dependent ERK1/2 acti- vation illustrated in Fig. 1E.

MAP Kinase Activation by a Gs/PKA-selective PTH1R Ligand—Phar- macologic interest in modifying PTH1R-agonist interactions that con- tribute to␤-arrestin-mediated desensitization has lead to the design of novel PTH1R agonists that uniquely destabilize receptor-␤-arrestin association (32). These PTH analogues have been shown to induce selective stabilization of the active G protein-coupled state of the PTH1R receptor, without causing␤-arrestin recruitment. We exam- ined a new PTH1R agonist, PTH-NBR, and its ability to generate G protein second messengers and to stimulate MAP kinase activation. Fig.

3,AandB, illustrates the effect of PTH-(1–34) and PTH-NBR on cAMP generation and phosphoinositide (PI) hydrolysis in HEK293 cells

FIGURE 3.PTH-NBR activates adenylate cyclase (Gs) but does not activate phospho- lipase C (Gq).cAMP stimulation (A) and PI hydrolysis (B) in response to 100 nMPTH-(1–34) and 100 nMPTH-NBR were measured in HEK293 cells transiently expressing PTH1R. cAMP values were normalized to forskolin-induced levels. PI hydrolysis values were normalized to the maximal response induced by PTH-(1–34). Data correspond to the meanS.E.

from four independent experiments (***,p0.001 compared with the non-stimulated (NS) control values).

FIGURE 4.Inhibition of PKA inhibits PTH-NBR-stimulated ERK1/2 activation.A, phos- phorylation of endogenous ERK1/2 in HEK293 cells transiently transfected to express human PTH1R stimulated with 100 nMPTH-NBR for the indicated periods of time in the absence or presence of H89 (20␮M) or GFX (2.5␮M) was determined by immunoblotting (IB).B, time course of ERK1/2 activation by PTH-NBR compared with that of PTH-(1–34).C, ERK1/2 activation in cells pretreated with H89 or GFX prior to stimulation with 100 nM

PTH-NBR. Values in the graphs are expressed as percent of the phosphorylation of ERK1/2 obtained at 5 min of stimulation with PTH-(1–34) in the absence of inhibitors.

Data represent the meanS.E. from at least five independent experiments. Statistical comparison of the curves was performed by using a two-way ANOVA (†,p⬍0.0001), and a Bonferroni post-test was used to compare values at each of the time points (***,p 0.001; **,p⬍0.01; *,p0.05).DMSO, dimethyl sulfoxide.

at INRA Institut National de la Recherche Agronomique, on November 8, 2010www.jbc.orgDownloaded from

(7)

expressing PTH1R. Stimulation of cells expressing PTH1R with PTH- NBR (100 nM) increased cAMP levels as effectively as PTH-(1–34) (100 nM). There was no measurable generation of IP3in response to PTH- NBR. These experiments demonstrate that PTH-NBR acts as a selective agonist for Gs/PKA activation while having no apparent activity for Gq/PKC stimulation. The effect of PTH-NBR on the time dependence of ERK1/2 activation in HEK293 cells expressing wild type (WT) PTH1R is shown in Fig. 4,AandB. The time course of PTH-NBR- stimulated ERK1/2 activation was significantly different from the time course of PTH-(1–34)-stimulated ERK1/2 activation. PTH-NBR stim- ulated an early and rapid activation of ERK1/2 that reached a peak at 5 min. The peak response of PTH-NBR was 65% of that achieved by PTH- (1–34). PTH-NBR-stimulated ERK1/2, while robust, was not sustained and rapidly returned to basal levels by 30 – 60 min.

To examine the contributions of Gs/PKA- and Gq/PKC-dependent mechanisms to PTH-NBR-stimulated MAP kinase activation, cells were pretreated with the PKA inhibitor H89 (20M) or the PKC inhib- itor GFX (2.5M), respectively, for 15 min prior to stimulation with PTH-NBR (100 nM). Fig. 4Cshows that treatment with H89 completely

inhibited PTH-NBR MAP kinase activation and the time course of MAP kinase activation was not significantly affected by treatment with GFX.

These results indicate that PTH-NBR stimulation of PTH1R results in a time course of MAP kinase activation that is primarily dependent upon a Gs/PKA-mediated pathway. To determine whether␤-arrestins con- tributed to MAP kinase activation, we again used RNA interference to reduce the expression of endogenous␤-arrestin 1 or␤-arrestin 2. PTH- NBR-stimulated ERK1/2 activation was measured in HEK293 cells transfected with PTH1R and siRNA for␤-arrestin 1 or␤-arrestin 2. In control siRNA-transfected cells, ERK1/2 activation reached maximal levels within 5 min of treatment with PTH-(1–34) and then rapidly declined to basal levels by 30 – 60 min, and reduced amounts of either

␤-arrestin 1 or␤-arrestin 2 by siRNA did not significantly alter the time course of PTH-NBR ERK1/2 activation (Fig. 5,AandB). There was no difference in the time course of ERK activation when both␤-arrestin 1 and␤-arrestin 2 are silenced concomitantly (n2; data not shown).

The results obtained from the use of Gs/PKA and Gq/PKC inhibitors taken together with results demonstrating the absence of a␤-arrestin- mediated component contributing to PTH-NBR-stimulated ERK1/2 activation suggest that PTH-NBR exclusively activates ERK1/2 via a G protein-dependent mechanism, namely Gs/PKA.

With the lack of␤-arrestin recruitment by PTH-NBR, one might expect that the desensitization of PTHR would be impaired and that the ERK activation may be extended to some of the later time points. The results presented here suggest that PTH1R must undergo some form(s) of␤-arrestin-independent desensitization when stimulated by PTH- NBR. Desensitization of PTH1R by GRK2 in the absence of receptor phosphorylation has been described previously (33). PKA has also been implicated in the desensitization of PTH1R (34).

MAP Kinase Activation by a␤-Arrestin-selective PTH1R Ligand—It has been previously reported that PTH-IA exhibited inverse agonist activity for cAMP in cells expressing the constitutively active PTH1R H223R mutant (35). The PTH1R point mutation, H223R, constitutively activates adenylate cyclase, but not phospholipase C, and results in Jansens metaphyseal chondrodysplasia (36). The effects of PTH-(1–34) and PTH-IA on cAMP generation in HEK293 cells expressing the WT

FIGURE 5.PTH-NBR-stimulated ERK1/2 activation is unaffected by depletion of

␤-arrestin 1 or␤-arrestin 2.HEK293 cells transiently transfected to express human PTH1R and the indicated siRNAs were stimulated with 100 nMPTH-NBR for the indicated periods.A, phosphorylation of endogenous ERK1/2 was determined by immunoblotting (IB).B, the effect of␤-arrestin 1 siRNA or␤-arrestin 2 siRNA transfection on PTH-NBR- stimulated ERK/12 activation. Values in the graphs are expressed as percent of the phos- phorylation of ERK1/2 obtained at 5 min of stimulation in control (CTL) siRNA-transfected cells. Data represent the meanS.E. from at least four independent experiments. Sta- tistical comparison of the curves was performed by using a two-way ANOVA between

␤-arrestin siRNA-transfected and control siRNA-transfected cells. Bonferroni post-tests were used to compare values at each of the time points. These comparisons showed no significant differences.

FIGURE 6.PTH-IA is an inverse agonist for cAMP accumulation mediated by the constitutively active H223R mutant parathyroid hormone receptor.cAMP stimula- tion in response to PTH-(1–34) and PTH-IA was measured in HEK293 cells transiently expressing either the WT PTH1R or the H223R constitutively active mutant receptor. Cells were treated with 100 nMPTH-(1–34) or 1MPTH-IA. cAMP values were normalized to forskolin-induced levels. Data correspond to the meanS.E. from four independent experiments. (**,p0.01 compared with the non-stimulated (NS) control values for H223R).

FIGURE 7.PTH-IA-stimulated ERK1/2 activation is unaffected by PKA or PKC inhibi- tion.A, phosphorylation of endogenous ERK1/2 in HEK293 cells transiently transfected to express human PTH1R stimulated with 1MPTH-IA for the indicated periods of time in the absence or presence of H89 (20M) or GFX (2.5␮M) was determined by immunoblot- ting (IB).B, time course of ERK1/2 activation by PTH-IA in the absence and presence of H89 and GFX compared with that of PTH-(1–34). The values in the graphs are expressed as percent of the phosphorylation of ERK1/2 obtained at 5 min of stimulation with PTH- (1–34). Data represent the meanS.E. from at least four independent experiments.

Statistical analysis of the curves was performed by using a two-way ANOVA comparison between PTH-IA-stimulated cells in the absence and presence of inhibitors. Bonferroni post-tests were used to compare values at each of the time points. These comparisons showed no significant differences.DMSO, dimethyl sulfoxide.

-Arrestin- and G Protein-mediated ERK1/2 Activation by PTH1R

at INRA Institut National de la Recherche Agronomique, on November 8, 2010www.jbc.orgDownloaded from

(8)

PTH1R or the H223R mutant receptor were confirmed (Fig. 6). Reflect- ing its constitutive activity, the basal cAMP levels in untreated cells expressing H223R were 11-fold greater than basal levels in cells express- ing the WT receptor. Stimulation with PTH-(1–34) (100 nM) increased cAMP levels in cells expressing WT PTH1R or H223R to 15-fold WT receptor basal cAMP levels. Treatment of cells expressing WT PTH1R with PTH-IA (1M) did not activate cAMP. In cells expressing H223R, treatment with PTH-IA (1M) significantly reduced cAMP levels by 50% compared with the cAMP levels in the untreated cells, consistent with inverse agonist activity. PI hydrolysis provoked by PTH-(1–34), and PTH-IA was measured in HEK293 cells transiently expressing WT

PTH1R or H223R. There was no measurable generation of IP3 in response to PTH-IA in cells expressing the wild type PTH1R or the H223R mutant receptor (data not shown). These experiments demon- strate that PTH-IA acts as an inverse agonist for Gs/PKA activation while having no apparent activity for Gq/PKC stimulation.

Despite its inability to activate Gs/PKA or Gq/PKC signaling path- ways, the inverse agonist PTH-IA is able to stimulate MAP kinase acti- vation in HEK293 cells expressing PTH1R. The effect of PTH-IA on the time dependence of MAP kinase activation in HEK293 cells expressing WT PTHR1 is shown in Fig. 7, Aand B. The maximal stimulated ERK1/2 activity by PTH-IA reached 35% of that achieved by PTH-(1–

34). Consistent with the inability of PTH-IA to activate Gs/PKA or Gq/PKC signaling pathways ERK1/2 activation by PTH-IA is unaffected by PKA or PKC inhibition with H89 or GFX, respectively (Fig. 7,Aand B). Moreover, the time course of ERK1/2 activation was prolonged resembling the response to PTH-(1–34) observed in the presence of PKA/PKC inhibitors.

Since PTH-IA is unable to activate Gs/PKA or Gq/PKC signaling pathways, we hypothesized that the ERK1/2 activation provoked by the

“inverse agonist” is mediated by␤-arrestin. To determine whether␤-ar- restin 1 or␤-arrestin 2 are contributing to ERK1/2 activation we used siRNA to silence their expression. Fig. 8,A–C, show that in HEK293 cells cotransfected with control siRNA and PTH1R, PTH-IA stimulates ERK1/2 in a time-dependent manner. The reduced expression of␤-ar- restin 1 or␤-arrestin 2 by siRNA abolished this response at all time points. Taken together with the evidence that PTH1R has the ability to activate ERK1/2 through a␤-arrestin-mediated pathway, which is inde- pendent from the G protein-mediated PKA/PKC-dependent ERK1/2 activation, these results suggest that PTH-IA exclusively activates ERK1/2 via this mechanism. The time course of␤-arrestin-mediated

FIGURE 8.ERK1/2 activation by PTH-IA is␤-arrestin-dependent.HEK293 cells tran- siently transfected to express human PTH1R and the indicated siRNAs were stimulated with 1MPTH-IA for the indicated periods.A, phosphorylation of endogenous ERK1/2 was determined by immunoblotting (IB).BandC, the effect of␤-arrestin 1 siRNA or

␤-arrestin 2 siRNA transfection on PTH-IA-stimulated ERK/12 activation. The values in the graphs are expressed as percent of the phosphorylation of ERK1/2 obtained at 5 min of stimulation in control (CTL) siRNA-transfected cells. Data represent the mean⫾S.E. from at least five independent experiments. Statistical analysis of the curves was performed by using a two-way ANOVA comparison between␤-arrestin siRNA-transfected cells and control siRNA-transfected cells (†,p0.0001), and a Bonferroni post-test was used to compare values at each of the time points (***,p0.001).D, the␤-arrestin-dependent ERK1/2 activation induced by PTH-IA overlies the predicted G protein-independent com- ponent of ERK1/2 activation represented by the data obtained from the PTH-(1–34) in the presence of combined inhibitors for PKA and PKC (shown in Fig. 1E).

FIGURE 9.The association of␤-arrestin with PTH1R is induced by PTH-(1–34) and PTH-IA.A, coimmunoprecipitation of␤-arrestin with FLAG-PTH1R was determined by immunoblot (IB). HEK293 cells transiently transfected with cDNAs encoding FLAG- PTH1R, His-␤-arrestin 1, and His-␤-arrestin 2 were treated with 100 nMPTH-(1–34), 1M PTH-IA, or 100 nMPTH-NBR. Cell lysates were prepared as described and immunoprecipi- tated (IP) with anti-FLAG-agarose.B, quantitated data correspond to the meanS.E.

from four independent experiments (*,p0.05 compared with the non-stimulated (NS) control values).

at INRA Institut National de la Recherche Agronomique, on November 8, 2010www.jbc.orgDownloaded from

(9)

ERK1/2 activation through the PTH1R demonstrated by stimulating PTH1R with PTH-(1–34) in the presence of combined PKA and PKC inhibitors (from Fig. 1E) or by PTH-IA without inhibitors are virtually identical (Fig. 8D).

To examine the interaction of␤-arrestin with PTH1R, HEK239 cells transiently overexpressing FLAG-epitope-tagged PTH1R and␤-arres- tin 1 and␤-arrestin 2 were treated with PTH-(1–34) (100 nM), PTH-IA (1M), or PTH-NBR (100 nM), and the formation of receptor␤-arrestin complexes was measured by coimmunoprecipitation after cross-linking with dithiobis(succinimidyl propionate). As shown in Fig. 9,AandB,

␤-arrestin coimmunoprecipitates with the receptor after stimulation with PTH-(1–34) as well as with PTH-IA. The association of␤-arrestin with FLAG-PTH1R stimulated by PTH-IA was 30% of that stimulated by PTH-(1–34). Coimmunoprecipitaion stimulated by PTH-NBR was not significantly different from the non-stimulated control. These data support the ability of the PTH-IA to induce a receptor conformation that has the ability to recruit␤-arrestin independent of G protein acti- vation, while PTH-NBR, which effectively activates Gs, does not recruit

␤-arrestin.

DISCUSSION

PTH1R has been recognized as a key regulator of calcium homeosta- sis and bone metabolism. Despite the importance in PTH-mediated signals in bone remodeling, our understanding of the mechanistic basis for these effects is incomplete. Our findings indicate that PTH-(1–34) stimulation of the PTH1R activates ERK1/2 MAP kinase by two tempo- rally distinct mechanisms: one, a G protein-dependent pathway, which is rapid in onset and quite transient, and the other␤-arrestin-depend- ent, which is slower in onset but much more persistent. Furthermore, these distinct mechanisms of MAP kinase activation can be selectively stimulated through the use of novel PTH analogues.

Our data suggest that both␤-arrestin 1 and␤-arrestin 2 are required for PKA/PKC-independent stimulation of the ERK MAPK cascade by PTH1R. This differs from the reciprocal effects of␤-arrestin 1 and␤-ar- restin 2 observed for angiotensin II type 1A receptor-mediated ERK1/2 activation (29). For the AT1A receptor, ERK1/2 activation is increased when the cellular level of␤-arrestin 1 is down-regulated by siRNA and ERK1/2 activation is decreased or eliminated when the cellular level of

␤-arrestin 2 is diminished. The requirement for both␤-arrestin iso- forms in PTH1R signaling could reflect a unique requirement for␤-ar- restin heterodimers in the transmission of␤-arrestin-depenedent sig- nals by the PTH1R. Dimerization of ␤-arrestin is supported by structural studies of visual arrestin, a protein analogous to␤-arrestin, that is responsible for rapid desensitization of the GPCR, rhodopsin.

Sedimentation equilibrium analysis of visusal arrestin has provided evi- dence for arrestin dimerization at physiologic concentrations and crys- tallographic studies of visual arrestin reveal the association of two asym- metric dimers (38, 39). It has also recently been demonstrated, using coimmunoprecipitaion and resonance energy transfer (biolumines- cence resonance energy transfer (BRET) and fluorescence resonance energy transfer (FRET)), that ␤-arrestins constitutively form both homo- and hetero-oligimers at physiologic concentrations. Further- more, hetero-dimerization may influence the subcellular distribution of

␤-arrestin 1 and␤-arrestin 2 and their functions (40).

We also show that the two mechanisms of PTH1R ERK1/2 activation (G proteinversus␤-arrestin) can be distinguished by the use of ligands that preferentially activate one or the other pathway. Underlying the complexity of 7TMR signaling diversity are discrete receptor conforma- tions that may be preferentially stabilized as a result of binding to spe- cific receptor ligands. It has been previously demonstrated for PTH1R

that various N-terminal PTH and PTHrp fragments and truncated hPTH-(1–34) analogues are capable of selectively stimulating distinct G protein-mediated signals including adenylate cyclase (Gs) or PKC (Gq) activity (41, 42). Biased agonism-simulating G protein-independent/␤- arrestin-dependent 7TMR signaling to ERK1/2 has been shown in the AT1A angiotensin receptor system using a synthetic angiotensin ago- nist peptide, [Sar1,Ile4,Ile8]SII (43).

Recently, some ligands originally classified as inverse agonists based on their effects on G protein-mediated signaling have been shown to promote scaffold assembly and␤-arrestin-mediated MAPK activation (22). These observations suggest that ␤-arrestin recruitment is not exclusive to “agonist”-stimulated 7TMRs and G protein activation. Fur- thermore, ligands classically classified as inverse agonists with respect to G protein activation may rely on␤-arrestin for their positive signaling activity. This phenomenon is not unique to PTH1R. ICI118551, an inverse agonist for the2-adrenergic receptor, and SR121463B, an inverse agonist for the V2vasopressin receptor-stimulated adenylate cyclase, have also been shown to recruit ␤-arrestin and stimulate ERK1/2 (22). The data presented here are a demonstration of biased agonism for PTH1R, where unique PTH analogues can selectively stim- ulate G protein-dependent signaling (specifically Gs/PKA) or␤-arrestin- dependent signaling mechanisms. Moreover, these findings add to the growing evidence that␤-arrestin scaffolds can mediate 7TMR signaling to ERK1/2 independent of G-protein activation and that␤-arrestin recruitment is not exclusive to classical 7TMR agonists of G-protein- stimulated second messengers.

It is evident that 7TMRs employ multiple mechanisms to activate the ERK1/2 cascade. The signaling mechanisms underlying ERK1/2 activa- tion are complex and may result from activation of classical G protein- regulated effectors such as PKA and PKC, from cross-talk between 7TMRs and receptor tyrosine kinases or focal adhesion complexes or from␤-arrestin scaffolding directly on the 7TMR (11). Depending on receptor and cell type, one mechanism may predominate or multiple mechanisms may be activated simultaneously. The contribution of

␤-arrestin-mediated ERK1/2 activation is widely variable depending upon receptor type. It has been shown in murine embryonic fibroblasts stably expressing the PAR2 receptor that ERK1/2 phosphorylation is predominately mediated through a␤-arrestin-dependent mechanism, whereas in HEK293 cells expressing AT1A angiotensin,␤-arrestin-de- pendent and G protein-dependent mechanisms almost equally contrib- ute to the temporal activation of ERK1/2 (44, 45). Here we show that the temporal contribution␤-arrestin-mediated ERK1/2 activation for the PTH1R is about 35%.

Despite the apparent redundancy of these multiple mechanisms of MAPK activation, emerging data suggest that the different pathways leading to ERK1/2 activation downstream of 7TMRs are not only mech- anistically distinct but also perform different signaling functions.

ERK1/2 activated by G proteins generally accumulates in the nucleus where it phosphorylates and activates various transcription factors (46).

In contrast, ERK1/2 activated by␤-arrestin is largely excluded from the nucleus and is confined to the cytoplasmic compartment where it pre- sumably phosphorylates a distinct set of effectors (47). While␤-arrestin 2 has been shown to influence bone remodeling and the skeletal response to intermittent PTH-(1–34) (26), the contributions of G pro- tein-mediated ERK1/2 activation or␤-arrestin-mediated ERK1/2 acti- vation to cellular responses such as proliferation, differentiation, migra- tion, and apoptosis remain to be determined.

While PTH-(1–34) activates both G protein and␤-arrestin-mediated signaling, PTH-NBR activates only Gs/PKA signaling, and PTH-IA acti- vates only ␤-arrestin signaling (while inhibiting G signaling). Such

-Arrestin- and G Protein-mediated ERK1/2 Activation by PTH1R

at INRA Institut National de la Recherche Agronomique, on November 8, 2010www.jbc.orgDownloaded from

(10)

results directly imply that the receptor conformations initiating the two forms of signaling must be distinct. Over the past several years this idea, variously referred to as “biased agonism” or “ligand-directed signaling”, has been gaining currency (21, 37). This phenomenon is likely to be of significant physiological and therapeutic impact, since the cellular con- sequences of the distinct mechanisms are likely quite different. An important implication of this work is that the receptor can exist in more than one active conformation. Thus, the receptor conformation induced by PTH-NBR selectively activates Gs/PKA but is unable to activate Gq/PKC or recruit ␤-arrestin. The receptor conformation induced by PTH-IA, which is not able to activate G proteins, is able to activate ERK1/2 via␤-arrestin. This demonstration of fundamentally distinct agonist-induced conformations of the PTH1R may have signif- icant implications both for understanding the molecular signaling mechanism of PTH as well as for the development of novel therapeutics for the treatment of various bone pathologies. For example PTH stim- ulation can lead to anabolic or catabolic effects dependent upon inter- mittent or persistent exposure, respectively. The regulatory mecha- nisms invoked in these contrary responses are incompletely understood. The availability of agents such as PTH-NRB and PTH-IA, which allow dissection of previously unappreciated signaling pathways, may ultimately herald the development of entirely new agents, which very specifically target a desired subset of PTH actions.

Acknowledgments—We thank Eric L. Moore (Xsira) and Jeremy L. Rouse (Xsira) for their technical expertise and Carson Loomis (Xsira) for his support and helpful discussions of this project. We also thank Donna Addison and Elizabeth Hall for excellent secretarial assistance.

REFERENCES

1. Tam, C. S., Heersche, J. N., Murray, T. M., and Parsons, J. A. (1982)Endocrinology 110,506 –512

2. Hock, J. M., and Gera, I. (1992)J. Bone Miner. Res.7,65–72

3. Qin, L., Raggatt, L. J., and Partridge, N. C. (2004)Trends Endocrinol. Metab.15, 60 – 65

4. Iida-Klein, A., Guo, J., Takemura, M., Drake, M. T., Potts, J. T., Jr., Abou-Samra, A., Bringhurst, F. R., and Segre, G. V. (1997)J. Biol. Chem.272,6882– 6889

5. Abou-Samra, A. B., Juppner, H., Force, T., Freeman, M. W., Kong, X. F., Schipani, E., Urena, P., Richards, J., Bonventre, J. V., Potts, J. T., Jr., Kronenberg, H. M., and Segre, G. V. (1992)Proc. Natl. Acad. Sci. U. S. A.89,2732–2736

6. Bringhurst, F. R., Juppner, H., Guo, J., Urena, P., Potts, J. T., Jr., Kronenberg, H. M., Abou-Samra, A. B., and Segre, G. V. (1993)Endocrinology132,2090 –2098 7. Juppner, H., Abou-Samra, A. B., Freeman, M., Kong, X. F., Schipani, E., Richards, J.,

Kolakowski, L. F., Jr., Hock, J., Potts, J. T., Jr., Kronenberg, H. M., and Segre, G. V.

(1991)Science254,1024 –1026

8. Verheijen, M. H., and Defize, L. H. (1997)J. Biol. Chem.272,3423–3429 9. Cole, J. A. (1999)Endocrinology140,5771–5779

10. Lederer, E. D., Sohi, S. S., and McLeish, K. R. (2000)J. Am. Soc. Nephrol.11,222–231 11. Gutkind, J. S. (2000)Sci. Stke40,11

12. Swarthout, J. T., Doggett, T. A., Lemker, J. L., and Partridge, N. C. (2001)J. Biol. Chem.

276,7586 –7592

13. Garcia-Ocana, A., Gomez-Casero, E., Penaranda, C., and Esbrit, P. (1998)Life Sci.62, 2267–2273

14. Freedman, N. J., and Lefkowitz, R. J. (1996)Recent Prog. Horm. Res.51,319 –353

15. Ferguson, S. S. (2001)Pharmacol. Rev.53,1–24

16. McDonald, P. H., Chow, C. W., Miller, W. E., Laporte, S. A., Field, M. E., Lin, F. T., Davis, R. J., and Lefkowitz, R. J. (2000)Science290,1574 –1577

17. 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)Science283,655– 661

18. Luttrell, L. M., Roudabush, F. L., Choy, E. W., Miller, W. E., Field, M. E., Pierce, K. L., and Lefkowitz, R. J. (2001)Proc. Natl. Acad. Sci. U. S. A.98,2449 –2454

19. DeFea, K. A., Zalevsky, J., Thoma, M. S., Dery, O., Mullins, R. D., and Bunnett, N. W.

(2000)J. Cell Biol.148,1267–1281

20. Lefkowitz, R. J., and Shenoy, S. K. (2005)Science308,512–517

21. Brzostowski, J. A., and Kimmel, A. R. (2001)Trends Biochem. Sci.26,291–297 22. Azzi, M., Charest, P. G., Angers, S., Rousseau, G., Kohout, T., Bouvier, M., and

Pineyro, G. (2003)Proc. Natl. Acad. Sci. U. S. A.100,11406 –11411 23. Ferrari, S. L., and Bisello, A. (2001)Mol. Endocrinol.15,149 –163

24. Ferrari, S. L., Behar, V., Chorev, M., Rosenblatt, M., and Bisello, A. (1999)J. Biol.

Chem.274,29968 –29975

25. Vilardaga, J. P., Frank, M., Krasel, C., Dees, C., Nissenson, R. A., and Lohse, M. J.

(2001)J. Biol. Chem.276,33435–33443

26. Ferrari, S. L., Pierroz, D. D., Glatt, V., Goddard, D. S., Bianchi, E. N., Lin, F. T., Manen, D., and Bouxsein, M. L. (2005)Endocrinology146,1854 –1862

27. Bouxsein, M. L., Pierroz, D. D., Glatt, V., Goddard, D. S., Cavat, F., Rizzoli, R., and Ferrari, S. L. (2005)J. Bone Miner. Res.20,635– 643

28. Lin, F.-T., Krueger, K. M., Kendall, H. E., Daaka, Y., Fredericks, Z. L., Pitcher, J. A., and Lefkowitz, R. J. (1997)J. Biol. Chem.272,31051–31057

29. Ahn, S., Wei, H., Garrison, T. R., and Lefkowitz, R. J. (2004)J. Biol. Chem.279, 7807–7811

30. Ahn, S., Nelson, C. D., Garrison, T. R., Miller, W. E., and Lefkowitz, R. J. (2003)Proc.

Natl. Acad. Sci. U. S. A.100,1740 –1744

31. Zamah, A. M., Delahunty, M., Luttrell, L. M., and Lefkowitz, R. J. (2002)J. Biol. Chem.

277,31249 –31256

32. Bisello, A., Chorev, M., Rosenblatt, M., Monticelli, L., Mierke, D. F., and Ferrari, S. L.

(2002)J. Biol. Chem.277,38524 –38530

33. Dicker, F., Quitterer, U., Winstel, R., Honold, K., and Lohse, M. J. (1999)Proc. Natl.

Acad. Sci. U. S. A.96,5476 –5481

34. Fukayama, S., Tashjian, A., Jr., and Bringhurst, F. (1992) Endocrinology 131, 1757–1769

35. Gardella, T. J., Luck, M. D., Jensen, G. S., Schipani, E., Potts, J. T., Jr., and Juppner, H.

(1996)Endocrinology137,3936 –3941

36. Schipani, E., Jensen, G. S., Pincus, J., Nissenson, R. A., Gardella, T. J., and Juppner, H.

(1997)Mol. Endocrinol.11,851– 858 37. Kenakin, T. (2001)FASEB J.15,598 – 611

38. Schubert, C., Hirsch, J. A., Gurevich, V. V., Engelman, D. M., Sigler, P. B., and Fleming, K. G. (1999)J. Biol. Chem.274,21186 –21190

39. Granzin, J., Wilden, U., Choe, H.-W., Labahn, J., Krafft, B., and Buldt, G. (1998) Nature391,918 –921

40. Storez, H., Scott, M. G., Issafras, H., Burtey, A., Benmerah, A., Muntaner, O., Piolot, T., Tramier, M., Coppey-Moisan, M., Bouvier, M., Labbe-Jullie, C., and Marullo, S.

(2005)J. Biol. Chem.280,40210 – 40215

41. Azarani, A., Goltzman, D., and Orlowski, J. (1996)J. Biol. Chem.271,14931–14936 42. Takasu, H., Gardella, T. J., Luck, M. D., Potts, J. T., and Bringhurst, F. R. (1999)

Biochemistry38,13453–13460

43. Wei, H., Ahn, S., Shenoy, S. K., Karnik, S. S., Hunyady, L., Luttrell, L. M., and Lefkow- itz, R. J. (2003)Proc. Natl. Acad. Sci. U. S. A.100,10782–10787

44. Stalheim, L., Ding, Y., Gullapalli, A., Paing, M. M., Wolfe, B. L., Morris, D. R., and Trejo, J. (2005)Mol. Pharmacol.67,78 – 87

45. Ahn, S., Shenoy, S. K., Wei, H., and Lefkowitz, R. J. (2004)J. Biol. Chem.279, 35518 –35525

46. Pierce, K. L., Luttrell, L. M., and Lefkowitz, R. J. (2001)Oncogene20,1532–1539 47. Tohgo, A., Pierce, K. L., Choy, E. W., Lefkowitz, R. J., and Luttrell, L. M. (2002)J. Biol.

Chem.277,9429 –9436

at INRA Institut National de la Recherche Agronomique, on November 8, 2010www.jbc.orgDownloaded from

Références

Documents relatifs

In vitro, hepatocytes can also proliferate after growth factor stimulations and in vitro rat hepatocyte cell cycle progression highly mimicked the kinetic of cell proliferation

A Souligne le groupe sujet (GS) et encadre le verbe (V) de chaque phrase. a) Chaque semaine, mon frère et mon père se promènent en forêt. b) Sur la plage, deux mouettes

by Misra and Sudarshan [3], such quantity could not exist in the usual quantum mechanical treatment of the decay processes and could not be related to the “survival probability” for

The effect of mixing calcium and oxalate precursors by diffusion at miscible liquid interfaces on calcium oxalate crystalline phases, and in physiological conditions (concentrations

Contrarily, in the setting of BMS-ISR, the RIBS V (Restenosis Intra-stent: Drug-eluting Balloon vs. Everolimus-eluting Stent) trial 23 , which recently provided the first

Another indicator for the involvement of the olfactory system in Parkinson’s is the finding that α-synuclein aggregates were found across the central olfactory system, including

Quand je demande à benjamin à qui ce personnage lui fait penser, ou de quel modèle il s’est inspiré, ses réponses sont vagues, laissant penser qu’il s’agit d’un

Computational Fluid Dynamics (CFD) simulations of a typical office under both mix- ing and displacement ventilation were performed to study the effects of room