Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Molecular and Cellular Endocrinology 348 (2012) 247–254

Contents lists available at SciVerse ScienceDirect

Molecular and Cellular Endocrinology


journal homepage: www.elsevier.com/locate/mce

The truncated ghrelin receptor polypeptide (GHS-R1b) is localized


in the endoplasmic reticulum where it forms heterodimers with ghrelin receptors
(GHS-R1a) to attenuate their cell surface expression
Kevin B.S. Chow, Jingxin Sun, Kit Man Chu, Wing Tai Cheung, Christopher H.K. Cheng, Helen Wise ⇑
School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China

a r t i c l e i n f o a b s t r a c t

Article history: The ghrelin receptor (GHS-R1a) is remarkable amongst G-protein-coupled receptors for its high degree of
Received 22 July 2011 constitutive activity, and this agonist-independent activity may be important for its physiological func-
Received in revised form 24 August 2011 tion in the control of food intake and body weight. Ghrelin receptors form heterodimers with the trun-
Accepted 26 August 2011
cated ghrelin receptor polypeptide (GHS-R1b), which has a dominant-negative effect on ghrelin
Available online 31 August 2011
receptor function. Here we show that GHS-R1b has an intracellular localization distinct from ghrelin
receptors, being primarily localized in the endoplasmic reticulum. Immunocytochemical studies suggest
Keywords:
that GHS-R1b decreases the plasma membrane expression of ghrelin receptors, but the overall distribu-
Ghrelin receptor
GHS-R1b
tion profile of ghrelin receptors in isolated subcellular fractions is unaffected by GHS-R1b. Using biolumi-
Dominant-negative mutant nescence resonance energy transfer methods, we have shown that while ghrelin receptor homodimers
Heterodimerization are evenly distributed in all subcellular fractions, GHS-R1a/GHS-R1b heterodimers are concentrated
within the endoplasmic reticulum and these results suggest that GHS-R1b traps ghrelin receptors within
the endoplasmic reticulum by the process of oligomerization. Furthermore, ghrelin receptors constitu-
tively activated extracellular signal-regulated kinases 1/2 in the endoplasmic reticulum, but this small
response was not affected by GHS-R1b and its physiological relevance is uncertain. Taken together, these
results suggest that ghrelin receptors can be retained in the endoplasmic reticulum by heterodimeriza-
tion with GHS-R1b, and constitutive activation of phospholipase C is attenuated due to decreased cell sur-
face expression of ghrelin receptors. However, sufficient ghrelin receptor homodimers can still be
expressed on the cell surface for maximal responses to agonist stimulation.
Ó 2011 Elsevier Ireland Ltd. All rights reserved.

1. Introduction 2009). Nevertheless, studies of mice lacking ghrelin and the ghre-
lin receptor indicate that the primary metabolic function of ghrelin
Ghrelin is a gastrointestinal hormone and is the only circulating in adult mice may be to modulate glucose sensing and insulin sen-
orexigenic hormone secreted from a peripheral organ to act on the sitivity, rather than directly regulate energy intake and energy
hypothalamic arcuate and ventromedial nuclei to regulate appetite expenditure (Sun et al., 2008). Indeed, the role of ghrelin in insulin
(Jia et al., 2008; Kojima and Kangawa, 2010). The ghrelin receptor regulation has been strengthened by observations that mice defi-
(growth hormone secretagogue receptor type 1a: GHS-R1a) is a G- cient in acylated ghrelin experience life-threatening hypoglycae-
protein-coupled receptor (GPCR) with a high degree of constitutive mia which can be prevented by exogenous ghrelin or growth
activity in vitro (Holst et al., 2003; Leung et al., 2007) and this ago- hormone (Zhao et al., 2010).
nist-independent activity may confer some anti-apoptotic proper- The constitutive activity of ghrelin receptors appears to be nec-
ties to cells (Lau et al., 2009). Two novel variants of the GHSR gene essary for effective ghrelin receptor internalization to recycling
have been identified which produced a selective loss of ghrelin compartments (Holliday et al., 2007), and in HEK293 cells tran-
receptor constitutive signalling and defects in growth profiles siently expressing ghrelin receptors, the constitutive activity of
(Pantel et al., 2006). High constitutive signalling from ghrelin ghrelin receptors is dramatically affected by co-expression with
receptors may be important for their in vivo function in the control the truncated ghrelin receptor polypeptide (GHS-R1b) due to de-
of food intake and body weight, as recently demonstrated using a creased cell surface expression (Chan and Cheng, 2004; Chu
potent nverse agonist for the ghrelin receptor (Petersen et al., et al., 2007; Leung et al., 2007). GHS-R1b is a naturally occurring
truncated splice variant consisting of the first five predicted trans-
⇑ Corresponding author. Tel.: +852 2609 6849; fax: +852 2603 5139. membrane regions of the ghrelin receptor; it fails to bind ghrelin
E-mail address: helenwise@cuhk.edu.hk (H. Wise). and has no known signalling activity (Howard et al., 1996). Relative

0303-7207/$ - see front matter Ó 2011 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.mce.2011.08.034
248 K.B.S. Chow et al. / Molecular and Cellular Endocrinology 348 (2012) 247–254

expression levels of ghrelin receptor and GHS-R1b mRNA vary in ERK1/2 and horseradish peroxidase-linked goat anti-rabbit immu-
different tissues (Gnanapavan et al., 2002), and in somatotroph noglobulin (Cell Signaling, Danvers, MA, USA). CellMask Deep Red
adenomas there are significantly higher levels of GHS-R1b mRNA Plasma Membrane stain, BODIPY Golgi TR C5-ceramide stain and
compared with normal pituitary tissues (Korbonits et al., 2001), ER Tracker Red stain were purchased from Invitrogen (Carlsbad,
suggesting that GHS-R1b may be oncogenic in these tissues. In hu- CA, USA). All other compounds were supplied by Invitrogen (Carls-
man carotid artery endothelial cells GHS-R1b mRNA expression is bad, CA, USA) or Sigma Chemical Co. (St. Louis, MO, USA).
significantly greater than in smooth muscle cells (Chow et al.,
2009) which may impact on the anti-inflammatory activity of 2.2. Cell culture and transfection reagents
ghrelin and its involvement in atherosclerosis (Waseem et al.,
2008). In ghrelin receptor-expressing HEK293 cells, the domi- HEK293 cells were purchased from ATCC and were maintained
nant-negative activity of GHS-R1b results in decreased constitutive in Dulbecco’s modified Eagle’s medium (DMEM) containing
activation of phosphatidylinositol-specific phospholipase C (PI- 100 i.u./ml penicillin, 100 lg/ml streptomycin and 10% foetal bo-
PLC) but does not affect ghrelin receptor signalling to extracellular vine serum, and were incubated in a humidified atmosphere of
signal-regulated kinases 1/2 (ERK1/2) (Chu et al., 2007; Leung 5% CO2/95% air at 37 °C. HEK293 cells were transfected at 80% con-
et al., 2007). These results suggested that any up-regulation of fluency using LipofectAMINE 2000 liposome reagent and Opti-
GHS-R1b expression might preferentially attenuate functional mem I reduced serum medium for 5 h, according to the manufac-
activity dependent on the constitutive activity of ghrelin receptors, turer’s instructions. All cells were harvested 48 h post-transfection.
while leaving agonist-stimulated ghrelin receptor signalling unaf- The cDNA constructs (GHS-R1a-Rluc, 2myc-GHS-R1a-GFP2, 3HA-
fected. However, the full functional consequences of GHS-R1b over GHS-R1b, GHS-R1b-Rluc, 3HA-GHS-R1b-GFP2 and CD8-Rluc) were
expression relative to GHS-R1a remain to be determined. prepared as described previously (Leung et al., 2007; Chow et al.,
In co-transfection assays, specific GHS-R1a/GHS-R1b heterodi- 2008). HEK293 cells were co-transfected with Rluc and GFP2 con-
mers were detected by bioluminescence resonance energy transfer structs at a maximal cDNA concentration of 1 lg/ml and at a cDNA
(BRET) and immunoprecipitation assays (Leung et al., 2007). The ratio of 1:5 (0.167 lg/ml Rluc-construct + 0.833 lg/ml GFP2 con-
formation of homodimers and heterodimers results in the produc- struct), to generate a near maximal BRET2 signal.
tion of receptor complexes with altered trafficking, signalling, and
pharmacological properties (Salahpour et al., 2000; Angers et al.,
2002; Milligan, 2004; Maggio et al., 2005; Milligan, 2010). Indeed, 2.3. Immunocytochemistry
GHS-R1b heterodimerizes with the neurotensin receptor (NSR1) to
generate a receptor responsive to neuromedin U (Takahashi et al., HEK293 cells were seeded at a cell density of 1.5  105 cells/
2006). Our original studies confirmed those of Smith et al. (2005) well on poly-D-lysine-coated 22 mm glass coverslips in 6-well tis-
indicating that ghrelin receptors were localized to the plasma sue culture plates. For live cell imaging, cells were stained with
membrane and cytoplasm of HEK293 cells transiently expressing various organelle dyes (CellMask Deep Red Plasma Membrane
ghrelin receptors, while GHS-R1b was in an intracellular compart- stain, BODIPY Golgi TR C5-ceramide stain or ER Tracker Red stain)
ment. Furthermore, the BRET50 values for GHS-R1a homodimers according to the manufacturer’s instructions, and observed using a
and GHS-R1a/GHS-R1b heterodimers were similar, indicating a Leica SP5 laser scanning confocal microscope equipped with Ar-
similar probability of GHS-R1a forming homo- or hetero-dimers gon/DPSS/HeNe lasers (Leica Microsystems GMSGmbH, Germany).
when expressed in the same cell as GHS-R1b (Leung et al., 2007). GFP2 was excited at 476 and 488 nm with a laser power ratio of
Appropriate dimer/oligomer formation appears to be essential for 2:1, and fluorescence was captured at 525 ± 25 nm. The captured
cell surface delivery of class A and class C GPCRs (Milligan, images were processed for publication using Adobe Photoshop
2010), therefore, the dominant-negative activity of GHS-R1b might CS4 (Adobe Systems, CA, USA).
best be explained by the formation of GHS-R1a/GHS-R1b heterodi-
mers within the endoplasmic reticulum (ER). The purpose of the 2.4. Subcellular fractionation
current study was therefore to identify the subcellular localization
of ghrelin receptors in the presence and absence of GHS-R1b, and According to the methods described by Guan et al. (2009), cells
to determine if the dominant-negative effect of GHS-R1b was in- were rinsed with PBS, harvested using a cell scraper, and centri-
deed due to heterodimerization within the ER. fuged at 300g for 5 min. A 10-fold volume of ice-cold hypotonic ly-
sis buffer (20 mM Hepes, pH 7.4; 2 mM EDTA; 2 mM EGTA; 6 mM
2. Materials and methods MgCl2) with protease inhibitors (Complete Tablets, EDTA-free;
Roche Applied Science, Indianapolis, IN, USA) was added to the
2.1. Materials resulting pellet and the cells were allowed to swell for 10 min at
4 °C. Cells were homogenized with 15 strokes of a Dounce homog-
Human GHS-R1 and human GHS-R1b cDNA were purchased enizer then centrifuged at 1000g for 10 min at 4 °C. The post-nucle-
from Guthrie Research Institute (Rolla, MD, USA), and the CD8- ar supernatant (fraction S; approximately 1–3 mg protein) was
Rluc cDNA construct was a gift from Prof. M. Bouvier (Montreal collected, resuspended in 2 M sucrose, and placed in the bottom
University, Canada). Cell culture reagents, including Lipofect- of a 12 ml ultracentrifuge tube for a Beckman SW41 Ti rotor, and
AMINE 2000 reagents, were purchased from Invitrogen (Carlsbad, overlayed with 1.9 M, 1.75 M, 1.5 M, 1.25 M, 1.0 M, 0.75 M and
CA, USA). Hybond nitrocellulose filters were from Pall Life Sciences 0.5 M sucrose. The discontinuous step sucrose gradient was centri-
(Pensacola, FL, USA) and the ECL kit was from GE Medical Systems fuged at 207,000g for 16 h at 4 °C. Aliquots of 1 ml were taken from
Hong Kong Ltd. Micro BCA Protein Assay Kit was from Pierce the top of each tube and BRET2 ratios were determined in duplicate
(Rockford, IL, USA). The codon humanized pRenilla luciferase-N 100 ll samples. To follow the distribution of Rluc-tagged proteins,
vector (pRluc-N2) and the codon humanized green fluorescent luciferase luminescence was also determined in duplicate 100 ll
protein vector (pGFP2-N) were purchased from PerkinElmer samples, using colenterazine H (5 lM) as described previously
(Atlanta, GA, USA). Antibody sources were: a-6F [Na+/K+ ATPase] (Leung et al., 2007). Equal volume samples were also taken from
(Developmental Studies Hybridoma Bank, University of Iowa, each aliquot for Western blotting where the expression of Na+/K+
USA); calnexin (StressGen, Ann Arbor, MI, USA); GHS-R1a and ATPase (a marker for the plasma membrane) and calnexin (a mar-
GHS-R1b (Phoenix Biotech (Beijing) Co. Ltd.); phospho-ERK1/2, ker for the endoplasmic reticulum) was expressed relative to that
K.B.S. Chow et al. / Molecular and Cellular Endocrinology 348 (2012) 247–254 249

in fraction S run on the same gel. Protein content of each sample 3. Results
was determined using Micro BCA Protein Assay Kit.
3.1. The subcellular localization of GHS-R1a ± GHS-R1b in whole cells
2.5. BRET2 assay
We were unable to unambiguously determine the localization
Bioluminescence resonance energy transfer (BRET) assays are of ghrelin receptors and GHS-R1b in native cells due to the lack
proximity-based assays which can provide a quantitative analysis of specificity of commercially available antibodies: anti-GHS-R1a
of receptor dimerization in living cells (Mercier et al., 2002). Because and anti-GHS-R1b antibodies both showed immunoreactivity in
BRET signals arise from the close interaction between Rluc- and pcDNA3.1-transfected HEK293 cells which should be devoid of
GFP2-expressing proteins, it is essential to distinguish specific and these proteins (see Lau et al., 2009) and both antibodies cross-re-
non-specific BRET signals (Mercier et al., 2002; Ayoub and Pfleger, acted in cell lysates run on Western blots (data not shown). There-
2010). We have previously shown that HEK293 cells expressing fore, although over-expression systems may not completely reflect
GHS-R1a/GHS-R1a or GHS-R1a/GHS-R1b displayed BRET2 satura- the situation in native cells, they are a useful starting point for
tion curves (Leung et al., 2007), indicative of a specific interaction these investigative studies. The subcellular localization of these
between GHS-R1a homodimers and GHS-R1a/GHS-R1b heterodi- proteins was therefore determined using live cell microscopy in
mers. In contrast, the interactions between GHS-R1b/GHS-R1b or HEK293 cells transiently transfected with 2myc-GHS-R1a-GFP2 or
CD8/GHS-R1a were non-specific in character because they showed 3HA-GHS-R1b-GFP2 (Fig. 1). We made use of the GFP2 tags to de-
quasi-linear BRET2 saturation curves (Leung et al., 2007; Chow tect the GHS-R constructs and cell stains for intracellular organ-
et al., 2008). Therefore, to demonstrate possible non-specific BRET2 elles. The overall distribution of 2myc-GHS-R1a-GFP2 and 3HA-
in the subcellular fractions, we again used a CD8-Rluc cDNA which GHS-R1b-GFP2, as detected from GFP fluorescence, was similar to
consists of a truncated form of the integrin CD8 fused to Rluc which that of 2myc-GHS-R1a and 3HA-GHS-R1b, as detected by the C-ter-
is reported to exhibit a subcellular distribution similar to that of the minal HA and myc tags (data not shown). 2myc-GHS-R1a-GFP2 co-
GPCRs, but is not expected to interact with them (Galés et al., 2005). localized with the plasma membrane (CellMask Deep Red), the
Samples (100 ll) from each of the 12 sucrose gradient fractions Golgi apparatus (BODIPY TR C5-ceramide) and the ER (ER Tracker
were transferred to two wells of a 96-well microplate (white Opti- Red) (Fig. 1A). In contrast, 3HA-GHS-R1b-GFP2 co-localized with
plate, PerkinElmer, Atlanta, GA, USA). The Rluc substrate Deep- the ER and Golgi apparatus markers but was absent from the plas-
BlueCTM (PerkinElmer, Atlanta, GA, USA) was added to a final ma membrane (Fig. 1B). Next we examined the effect of co-trans-
concentration of 5 lM, and readings were collected immediatedly fecting HEK293 cells with 2myc-GHS-R1a-GFP2 and 3HA-GHS-R1b,
using a FUSION microplate analyser (Packard, Meriden, CT, USA) at cDNA ratios of 1:5 to match the optimum conditions for the
that allows integration of signals detected at 410 nm and BRET2 assay for receptor dimerization (Leung et al., 2007). In the
515 nm, and normalized BRET2 ratios were determined as de- presence of 3HA-GHS-R1b, the expression of 2myc-GHS-R1a-
scribed previously (Leung et al., 2007). The BRET2 ratio is defined GFP2 in the plasma membrane and Golgi apparatus was decreased,
as the BRET2 ratio for the co-expression of the Rluc and GFP2 con- while still abundantly expressed in the ER (Fig. 1C).
structs normalized against the BRET2 ratio for the Rluc expression
construct alone. The normalized BRET2 ratio is defined as (emission
3.2. The localization of GHS-R1a and GHS-R1b in subfractioned cells
at 515 nm of co-transfected cells minus emission at 515 nm of
untransfected cells)/(emission at 410 nm of co-transfected cells
To determine the subcellular localization of GHS-R1a/GHS-R1b
minus emission at 410 nm of untransfected cells) – cf, where cf
heterodimers, we first prepared subcellular fractions of transfected
corresponds to (emission at 515 nm of Rluc-expressing cells minus
HEK293 cells according to Guan et al. (2009). With this method,
emission at 515 nm of untransfected cells)/(emission at 410 nm of
the HEK293 cell membrane fraction was applied to the bottom of
Rluc-expressing cells minus emission at 410 nm of untransfected
a discontinuous sucrose density gradient, and fractions were re-
cells) for the Rluc construct expressed alone in the same experi-
solved by flotation. From the distribution of markers, as assayed
ment. Assays were performed in duplicate at room temperature.
by Western blotting, the plasma membrane fractions were repre-
sented by fractions 1–5, and the ER fractions were in 5–9 (Fig. 2C).
2.6. Western blotting
The distribution of receptor-Rluc constructs was determined by
monitoring luciferase activity in the sucrose gradient fractions. We
The fractions isolated from the sucrose density gradient (25 ll)
identified two peaks of GHS-R1a-Rluc expression, one in plasma
were mixed with sample loading buffer, separated by 10%
membrane fractions and a second peak of luciferase activity in
SDS–polyacrylamide gel electrophoresis, and electrophoretically
the ER fractions (Fig. 2A). This method of analysis did not reveal
transferred onto Hybond nitrocellulose membranes. Membranes
any significant change in the distribution of GHS-R1a-Rluc when
were probed with anti- a-6F for plasma membrane (Na+/K+ ATP-
HEK293 cells were co-transfected with 3HA-GHS-R1b-GFP2
ase; 1:1000) and calnexin for endoplasmic reticulum (1:2000).
(Fig. 2A). 3HA-GHS-R1b-Rluc alone was not detected in the plasma
Phospho-ERK1/2 (1:500) and total ERK1/2 (1:1000) were sequen-
membrane fractions and showed a similar ER distribution profile as
tially detected by specific primary antisera and horseradish perox-
CD8-Rluc (Fig. 2B) which was quite distinct from that of GHS-R1a-
idase-linked goat anti-rabbit immunoglobulin. The immunoblots
Rluc (Fig. 2A).
were visualized by chemiluminescence with an enhanced chemilu-
minescence (ECL) kit, and the band intensities were quantified
using the computer program Image J (NIH, USA). 3.3. The localization of GHS-R1a homodimers and GHS-R1a/GHS-R1b
heterodimers in subfractionated cells
2.7. Data analysis
To determine the precise subcellular localization of BRET2 sig-
Values reported are means ± SEM. Comparisons between groups nals, BRET2 was assayed in all 12 fractions using Rluc- and GFP2-
were made using ANOVA (analysis of variance) with Bonferroni’s tagged receptor constructs. Raw data for fractions 1–9 is presented
post hoc tests, as appropriate, using GraphPad Prism software ver- and shows a steady decline in BRET2 from GHS-R1a homodimers
sion 5.0 (GraphPad Software Inc., San Diego, USA). Statistical signif- (Fig. 3A) and a slight increase in BRET2 in the ER fractions for GHS-
icance was taken as P < 0.05. R1a/GHS-R1b heterodimers (Fig. 3A). BRET2 signals in the bottom
250 K.B.S. Chow et al. / Molecular and Cellular Endocrinology 348 (2012) 247–254

Fig. 1. Ghrelin receptors (GHS-R1a) and the mutant ghrelin receptor polypeptide (GHS-R1b) show different subcellular localization patterns and GHS-R1b traps GHS-R1a in
the ER. HEK293 cells were transfected with GFP2-tagged receptors (green) and stained with subcellular markers (red) of the plasma membrane (CellMask Deep Red), the Golgi
apparatus (BODIPY TR C5-ceramide) and the ER (ER Tracker Red). Live cell confocal microscopy images are shown which are representative of three independent experiments.
(A) 2myc-GHS-R1a-GFP2 co-localized with markers of plasma membrane, Golgi and ER; (B) 3HA-GHS-R1b-GFP2 was only detected in the ER; and (C) less 2myc-GHS-R1a-GFP2
was observed in the plasma membrane and Golgi compartments in cells co-expressing 3HA-GHS-R1b. Images Scale bars = 10 lm. (For interpretation of the references to color
in this figure legend, the reader is referred to the web version of this paper.)

Fig. 2. The expression of Rluc receptor constructs in the sucrose density gradient. HEK293 cell membrane lysates were fractionated as described in Section 2, and Rluc activity
was assayed in duplicate 100 ll fractions. (A) GHS-R1a-Rluc shows a two-phase distribution profile in the absence (s) and presence of 3HA-GHS-R1b-GFP2 (d), with the
major peak in fraction 4 and a second around fraction 7. (B) Both GHS-R1b-Rluc (h) and CD8-Rluc (j) show a single-phase distribution pattern, peaking in fraction 7. Data are
means ± SEM, from 4 independent experiments, with each sample assayed in duplicate. (C) Equal volume fractions were analysed by Western blotting for Na+/K+ ATPase (d;
plasma membrane marker) and calnexin (N; ER marker), and a representative blot from 4 independent experiments is shown.
K.B.S. Chow et al. / Molecular and Cellular Endocrinology 348 (2012) 247–254 251

fractions have been excluded as these were extremely variable. In 2 by GHS-R1a in these fractions, but any ERK1/2 activity was below
living cell BRET2 assays, bystander BRET2 can arise from random col- the level of detection. To account for different protein loading on
lisions of Rluc- and GFP2-tagged receptors promoted by a high the gels, estimates of ERK1/2 activity were determined from the ra-
receptor density (Mercier et al., 2002). It is possible that bystander tio of pERK1/2 to tERK1/2 detected for each fraction. We observed a
BRET2 might also arise from aggregation of proteins in these bottom statistically significant increase in ERK1/2 activity in the ER frac-
fractions of the sucrose density gradient. Because absolute BRET2 tions of cells expressing GHS-R1a when compared with cells trans-
values cannot be used as a quantitative measure of the relative fected with the vector control (P < 0.05; two-way ANOVA), and this
number of homo/heterodimers formed for the different combina- effect remained in cells co-expressing GHS-R1a and GHS-R1b
tions (Ayoub and Pfleger, 2010), we normalized the BRET2 data to (Fig. 4).
the signal from fraction 4 for each pair of BRET2 constructs. This
method allowed us to better identify any relative change in the 4. Discussion
localization of the GHS-R1a homodimers compared with GHS-R1b
homodimers, and with GHS-R1a/GHS-R1b and CD8/GHS-R1a het- In the present study, HEK293 cells expressed the ghrelin recep-
erodimers. This method of analysis resulted in very high variability tor in the plasma membrane, Golgi and ER fractions, which is ex-
in fractions 1 and 2, but it is evident that specific GHS-R1a homodi- pected of a GPCR in heterologous expression systems, and in
mers appear evenly distributed in plasma membrane and ER frac- native cells and tissues (Gobeil et al., 2006). The ghrelin receptor
tions (Fig. 3C). The BRET2 signals from cells expressing GHS-R1b/ possesses a putative nuclear recognition sequence which would al-
GHS-R1b or CD8/GHS-R1a are non-specific in character because low for binding to importins and targeting of the protein to the nu-
they show quasi-linear BRET2 saturation curves (Leung et al., cleus (Christophe et al., 2000), however the present study failed to
2007; Chow et al., 2008), and here they showed no remarkable pat- find any evidence of nuclear translocation of ghrelin receptors even
tern of distribution (Fig. 3C,D). In marked contrast, the BRET2 signals in the presence of GHS-R1b. Contrary to ours (Leung et al., 2007)
from specific GHS-R1a/GHS-R1b heterodimers were significantly and other’s work (Smith et al., 2005), the confocal microscopy
increased in the ER compared with BRET2 from GHS-R1a homodi- images in the present study clearly show that GHS-R1b is not ex-
mers (Fig. 3B). pressed in the nucleus. Indeed, the intracellular localization of
GHS-R1b observed previously using ELISA-based assays (Leung
3.4. Activation of ERK1/2 by GHS-R1a in the endoplasmic reticulum et al., 2007) appears due to concentration of GHS-R1b within the
ER compartment. The ER forms part of the cellular quality control
Constitutive activation of ERK1/2 by ghrelin receptor-express- machinery where functionally inactive mutant GPCRs can be pre-
ing cells was readily detected by Western blotting of cell lysates, vented from expression at the cell surface (Salahpour et al.,
especially in fractions 7–10 (Fig. 4). Protein recovery in fractions 2004). We have previously shown that GHS-R1b acts as a domi-
1–5 was insufficient to detect tERK1/2, but peaked at approxi- nant-negative mutant of the ghrelin receptor and decreases its cell
mately 50 lg/100 ll in fraction 8. Therefore, the lack of data from surface expression (Leung et al., 2007). In the current study, both
fractions 1–6 does not necessarily mean lack of activation of ERK1/ 2myc-GHS-R1a-GFP2 and 3HA-GHS-R1b-GFP2 appeared in the ER,

Fig. 3. GHS-R1a/GHS-R1b heterodimers are concentrated in the ER fractions. HEK293 cell membrane lysates were fractionated as described in Section 2, with fraction 1–5
representing the plasma membrane fractions and 5–9 representing the ER fractions. The absolute BRET2 ratios for GHS-R1a-Rluc + 2myc-GHS-R1a-GFP2 (d) and GHS-R1a-
Rluc + 3HA-GHS-R1b-GFP2 (s) are shown in fractions 1–9 (A). The BRET2 ratios have been normalized to the BRET2 value in fraction 4, to allow comparison between different
BRET pairs: (B) GHS-R1a-Rluc + 2myc-GHS-R1a-GFP2 (d) and GHS-R1a-Rluc + 3HA-GHS-R1b-GFP2 (s); (C) GHS-R1a-Rluc + 2myc-GHS-R1a-GFP2 (d) and GHS-R1b-
Rluc + 3HA-GHS-R1b-GFP2 (4); (D) GHS-R1a-Rluc + 2myc-GHS-R1a-GFP2 (d) and CD8-Rluc + 2myc-GHS-R1a-GFP2 (N). Data are means ± SEM, from 4 independent
experiments, with each sample assayed in duplicate. ⁄P < 0.05, ⁄⁄P < 0.01, ⁄⁄⁄P < 0.001 compared with control cells expressing GHS-R1a-Rluc + 2myc-GHS-R1a-GFP2; 2-way
ANOVA.
252 K.B.S. Chow et al. / Molecular and Cellular Endocrinology 348 (2012) 247–254

ing the view that homodimerization is a prerequisite for receptor


trafficking to the cell surface (Lohse, 2010; Milligan, 2010).
To assess this hypothesis as the mechanism underlying the dom-
inant-negative activity of GHS-R1b (Leung et al., 2007), we used a
sucrose gradient centrifugation method to fractionate membranes
isolated from cells co-expressing Rluc- and GFP2-tagged receptor
constructs in order to determine if the subcellular localization of
ghrelin receptors was affected by the formation of specific hetero-
dimers with GHS-R1b. By following the expression of Rluc-tagged
proteins, we have shown that ghrelin receptors were localized both
in the plasma membrane (those fractions expressing the plasma
membrane marker Na+/K+ ATPase) and the ER (those fractions
expressing the ER marker calnexin), matching a similar subcellular
distribution pattern reported for b2-adrenergic receptors (Salah-
pour et al., 2004). In contrast to the ghrelin receptor, GHS-R1b
was not present in the plasma membrane and localized in the ER
fractions in a similar manner to the integrin CD8. The CD8-Rluc con-
struct used here as the negative control in the BRET2 assays (Chow
et al., 2008) was reported to exhibit a subcellular distribution pro-
file similar to that of GPCRs when visualized by confocal immuno-
fluorescence microscopy (Galés et al., 2005). Nevertheless, in the
present study, CD8-Rluc remained in the ER with a similar subcel-
lular distribution profile as GHS-R1b. Importantly, the presence of
GHS-R1b did not significantly change the distribution profile of
ghrelin receptors in these subcellular fractions. These observations
match those of Leung et al. (2007) where receptor ELISA assays indi-
cated that, despite the decreased cell surface expression of ghrelin
receptors, their relative distribution in extracellular and intracellu-
lar compartments remained similar in the absence and presence of
GHS-R1b. Monitoring the influence of GHS-R1b on the absolute
expression of ghrelin receptors cannot readily be performed by
detecting only Rluc-tagged ghrelin receptors in this experiment. Be-
cause the control HEK293 cells were also transfected with a 5-fold
Fig. 4. The presence of GHS-R1b does not affect the constitutive activation of ERK1/
2 by GHS-R1a in the ER. HEK293 cell membrane lysates were fractionated and the
excess of 2myc-GHS-R1a-GFP2 cDNA, then GHS-R1a-Rluc will make
ratio of pERK1/2 and tERK1/2 expression was determined as described in Materials up less than 17% of total ghrelin receptor expression and any de-
and Methods. Representative blots are shown in (B), (C) and (D). (A) Endogenous crease in plasma membrane expression of GHS-R1a-Rluc will be
ERK1/2 activity was very low in control cells transfected with empty vector hard to detect with this methodology.
(pcDNA3.1; h), but significantly increased in cells expressing GHS-R1a-Rluc (d);
⁄ Using a method of quantitative BRET analysis (Mercier et al.,
P < 0.05 compared with control cells expressing pcDNA3.1 (2-way ANOVA). The
presence of 3HA-GHS-R1b-GFP2 did not affect the activity of GHS-R1a-Rluc (s). 2002), we determined previously that the likelihood of forming
Data are means ± SEM, of 3 independent experiments. GHS-R1a/GHS-R1a homodimers and GHS-R1a/GHS-R1b heterodi-
mers was similar (Leung et al., 2007). Therefore, any ghrelin recep-
tors within the ER compartment would have to compete with GHS-
and the presence of GHS-R1b appeared to diminish co-localization R1b for the formation of GHS-R1a homodimers for trafficking to
of GHS-R1a with plasma membrane and Golgi markers. Our results the cell surface or in the formation of GHS-R1a/GHS-R1b heterodi-
suggest that GHS-R1b might decrease the cell surface expression of mers which would be retained in this intracellular compartment.
ghrelin receptors by hindering their progression from the ER to the The BRET2 signal from GHS-R1a/GHS-R1a homodimers followed a
Golgi apparatus, and thus decreasing their ultimate transport to similar distribution profile between plasma membrane and ER
the plasma membrane. fractions as described for b2-adrenergic receptors (Salahpour
GPCR mutants frequently exert a dominant-negative effect on et al., 2004) and lutropin receptors (Guan et al., 2009), being more
the cell signalling activity of corresponding full length receptors, evenly distributed throughout the subcellular fractions. This is be-
as has been shown for the a1A-adrenergic receptor (Cogé et al., cause BRET is a ratiometric value (Salahpour et al., 2004), so the
1999), a2-adrenergic receptors (Zhou et al., 2006), chemokine proportion of GHS-R1a-Rluc interacting with 2myc-GHS-R1a-
CCR5 and CXCR2 receptors (Benkirane et al., 1997; Trettel et al., GFP2 was similar in all the subcellular fractions.
2003), dopamine D3 receptor (Karpa et al., 2000; Elmhurst et al., As expected from our previous work (Leung et al., 2007), the
2000), gonadotrophin receptors (Grosse et al., 1997), histamine BRET2 signal from HEK293 cell membranes expressing GHS-R1a/
H4 receptors (van Rijn et al., 2008), prostacyclin receptors (Ibrahim GHS-R1a homodimers was much greater than from cells express-
et al., 2010), prostaglandin E2 EP1 receptor subtype (Okuda-Ashi- ing GHS-R1a/GHS-R1b heterodimers. This means that the effi-
taka et al., 1997), relaxin receptors (Kern et al., 2008), secretin ciency of resonance energy transfer between the energy donor
receptors (Ding et al., 2002) and the V2 vasopressin receptor (GHS-R1a-Rluc) and acceptor (2myc-GHS-R1a-GFP2) was greater
(Zhu and Wess, 1998). These dominant-negative mutants can re- than between GHS-R1a-Rluc and 3HA-GHS-R1b-GFP2, but does
sult in physiological outcomes in humans, such as red hair colour not necessarily mean that there were more GHS-R1a homodimers
and skin cancer for loss-of-function melanocortin 1 receptors than GHS-R1a/GHS-R1b heterodimers. To account for the differ-
(MC1R) (Sánchez-Laorden et al., 2009), and accelerated cardiovas- ences in BRET efficiencies, we normalized the BRET2 signals for
cular disease in the case of prostacyclin receptors (IPR212C) (Ibrahim individual donor/acceptor pairs, and found that the proportion of
et al., 2010). In many cases, these mutant GPCRs are proposed to GHS-R1a/GHS-R1b heterodimers was increased within the ER frac-
heterodimerize with the full length GPCRs within the ER, support- tions, whereas no obvious changes were observed for GHS-R1a
K.B.S. Chow et al. / Molecular and Cellular Endocrinology 348 (2012) 247–254 253

homodimers. Interpretation of BRET assays is prone to error when essential for receptor signalling, but it remains to be determined
using single concentrations of Rluc and GFP2 constructs (James if the relatively small changes observed here are physiologically
et al., 2006), but we have previously used BRET2 saturation analysis relevant.
and negative controls to study ghrelin receptor dimerization in In conclusion, these results suggest that ghrelin receptors can
whole cells (Leung et al., 2007; Chow et al., 2008). Thus, in be retained in the endoplasmic reticulum by heterodimerization
HEK293 cells co-transfected with GHS-R1b-Rluc and 3HA-GHS- with GHS-R1b, and constitutive activation of phospholipase C is
R1b-GFP2 or with CD8-Rluc and 2myc-GHS-R1a-GFP2, BRET2 satu- attenuated due to decreased cell surface expression of ghrelin
ration curves were quasi-linear (Leung et al., 2007), indicating that receptors. However, in the presence of GHS-R1b, sufficient ghrelin
the BRET2 signals resulted from random collision events (Mercier receptor homodimers can still be expressed on the cell surface for
et al., 2002; Ayoub and Pfleger, 2010). In the current study, non- maximal responses to agonist stimulation.
specific GHS-R1b homodimers and non-specific CD8/GHS-R1a het-
erodimers were not concentrated in the ER fractions. Together, Acknowledgements
these data support our hypothesis that GHS-R1b retains ghrelin
receptors within the ER through the specific process of heterodi- This work was fully supported by a Grant from the Research
merization, and this could account for the decreased cell surface Grants Council of the Hong Kong Special Administrative Region
expression of ghrelin receptors in the presence of GHS-R1b. De- (GRF475807). The authors would like to thank Dr. Deborah
spite the formation of specific GHS-R1a/GHS-R1b heterodimers in L. Segaloff for invaluable advice on the preparation and use of
the ER, sufficient GHS-R1a/GHS-R1a homodimers must be inserted the sucrose density gradient, and Prof. Micheal Bouvier for the
in the plasma membrane to respond to ghrelin for stimulation of CD8-Rluc construct.
phospholipase C and activation of ERK1/2 assays (Chu et al.,
2007; Leung et al., 2007). Thus, high expression levels of ghrelin
References
receptors are not essential for maximal stimulation of these cell
signalling pathways. Angers, S., Salahpour, A., Bouvier, M., 2002. Dimerization: an emerging concept for G
Ghrelin receptor signalling through Gq-proteins occurs predom- protein-coupled receptor ontogeny and function. Annu. Rev. Pharmacol.
inantly at the plasma membrane (Holliday et al., 2007). Although Toxicol. 42, 409–435.
Ayoub, M.A., Pfleger, K.D., 2010. Recent advances in bioluminescence resonance
ghrelin-stimulated ERK1/2 activity was also dependent on G-pro- energy transfer technologies to study GPCR heteromerization. Curr. Opin.
tein signalling in whole cells (Camina et al., 2007; Chu et al., Pharmacol. 10, 44–52.
2007), the precise localization of this activity was unknown. Nev- Benkirane, M., Jin, D.Y., Chun, R.F., Koup, R.A., Jeang, K.T., 1997. Mechanism of
transdominant inhibition of CCR5-mediated HIV-1 infection by cc5D32. J. Biol.
ertheless, the cell signalling activity of GPCRs is not necessarily
Chem. 272, 30603–30606.
limited to the plasma membrane, with persistent signalling occur- Calebiro, D., Nikolaev, V.O., Persani, L., Lohse, M.J., 2010. Signaling by internalized G-
ring in a G-protein-dependent manner from GPCRs internalized in protein-coupled receptors. Trends Pharmacol. Sci. 31, 221–228.
endosomes (Calebiro et al., 2010). In whole cells transiently Camina, J.P., Lodeiro, M., Ischenko, O., Martini, A.C., Casanueva, F.F., 2007.
Stimulation by ghrelin of p42/p44 nitrogen-activated protein kinase through
expressing ghrelin receptors, ghrelin-stimulated ERK1/2 activity the GHS-R1a receptor: role of G-proteins and b-arrestins. J. Cell. Physiol. 213,
was unaffected by coexpression with GHS-R1b, and data suggested 187–200.
that ERK1/2 activation did not depend on constitutive ghrelin Casar, B., Arozarena, I., Sanz-Moreno, V., Pinto, A., Agudo-Ibáñez, L., Marais, R.,
Lewis, R.E., Berciano, M.T., Crespo, P., 2009. Ras subcellular localization defines
receptor activity (Chu et al., 2007). In the current study, we have extracellular signal-regulated kinase 1 and 2 substrate specificity through
directly compared basal (constitutive) ghrelin receptor activity distinct utilization of scaffold proteins. Mol. Cell. Biol. 29, 1338–1353.
with ERK1/2 activation in HEK293 cells expressing empty vector Chan, C.B., Cheng, C.H.K., 2004. Identification and functional characterization of two
alternatively spliced growth hormone secretagogue receptor transcripts from
(pcDNA3.1), and have detected a small, but statistically significant, the pituitary of black seabream Acanthopagrus schlegeli. Mol. Cell. Endocrinol.
increase in pERK/tERK in the ER fractions of ghrelin receptor- 214, 81–95.
expressing cells which was unaltered by co-expression with Chow, K.B.S., Cheng, C.H.K., Wise, H., 2009. Anti-inflammatory activity of ghrelin in
human carotid artery cells. Inflammation 32, 402–409.
GHS-R1b. In HEK293 cells expressing GHS-R1a homodimers, ago- Chow, K.B.S., Leung, P.K., Cheng, C.H.K., Cheung, W.T., Wise, H., 2008. The
nists can decrease BRET2 but this effect is lost in the presence of constitutive activity of ghrelin receptors is decreased by co-expression with
GHS-R1b (Leung et al., 2007). As the chance for forming GHS- vasoactive prostanoid receptors when over-expressed in human embryonic
kidney 293 cells. Int. J. Biochem. Cell Biol. 40, 2627–2637.
R1a/GHS-R1a homodimers and GHS-R1a/GHS-R1b heterodimers
Christophe, D., Christophe-Hobertus, C., Pichon, B., 2000. Nuclear targeting of
is similar, it is likely that constitutive activation of ERK1/2 involves proteins: how many different signals? Cell. Signal. 12, 337–341.
GHS-R1a/GHS-R1a homodimers. Chu, K.M., Chow, K.B.S., Leung, P.K., Lau, P.N., Chan, C.B., Cheng, C.H.K., Wise, H.,
Because ghrelin-stimulated ERK1/2 was independent of ghrelin 2007. Over-expression of the truncated ghrelin receptor polypeptide attenuates
the constitutive activation of phosphatidylinositol-specific phospholipase C by
receptor internalization (Chu et al., 2007), the detection of pERK1/2 ghrelin receptors but has no effect on ghrelin-stimulated extracellular signal-
in subcellular fractions may not be dependent on prior activation regulated kinase 1/2 activity. Int. J. Biochem. Cell Biol. 39, 752–764.
of cell surface ghrelin receptors and their subsequent internaliza- Cogé, F., Guenin, S.P., Renouard-Try, A., Rique, H., Ouvry, C., Fabry, N., Beauverger, P.,
Nicholas, J.P., Galizzi, J.P., Boutin, J.A., Canet, E., 1999. Truncated isoforms inhibit
tion. Indeed, intracellularly-retained GPCRS, such as the Gs-cou- [3H]prazosin binding and cellular trafficking of native human a1A-
pled vasopressin receptor (V2R) localized in the ER, can still adrenoceptors. Biochem. J. 343, 231–239.
respond to cell permeable agonists (Robben et al., 2009). The cellu- Ding, W.Q., Cheng, Z.J., McElhiney, J., Kuntz, S.M., Miller, L.J., 2002. Silencing of
secretin receptor function by dimerization with a misspliced variant secretin
lar microenvironment will determine which substrates are prefer- receptor in ductal pancreatic adenocarcinoma. Cancer Res. 62, 5223–5229.
entially phosphorylated by activated ERK1/2 in cells expressing Du, Y., Ferro-Novick, S., Novick, P., 2004. Dynamics and inheritance of the
ghrelin receptors (Casar et al., 2009). As the ER is contiguous with endoplasmic reticulum. J. Cell Sci. 117, 2871–2878.
Elmhurst, J.L., Xie, Z., O’Dowd, B.F., George, S.R., 2000. The splice variant D3nf
the nuclear membrane (Du et al., 2004), it is conceivable that ghre- reduces ligand binding to the D3 dopamine receptor: evidence for
lin receptors in the ER may be functionally active. At this time heterooligomerization. Mol. Brain Res. 80, 63–74.
however, the organizational features and spatial orientation of Galés, C., Rebois, R.V., Hogue, M., Trieu, P., Breit, A., Hébert, T.E., Bouvier, M., 2005.
Real-time monitoring of receptor and G-protein interactions in living cells. Nat.
ghrelin receptor homodimers in the ER remains to be determined.
Methods 2, 177–184.
Nuclear glutamate receptors are orientated with their ligand bind- Gnanapavan, S., Kola, B., Bustin, S.A., Morris, D.G., McGee, P., Fairclough, P.,
ing domain inside the nucleoplasmic reticulum, and glutamate can Bhattacharya, S., Carpenter, R., Grossman, A.B., Korbonits, M., 2002. The tissue
access their binding sites by active transport into this subcellular distribution of the mRNA of ghrelin and subtypes of its receptor, GHS-R, in
humans. J. Clin. Endocrinol. Metab. 87, 2988–2991.
compartment (Kumar et al., 2008). Because ghrelin receptors are Gobeil, F., Fortier, A., Zhu, T., Bossolasco, M., Leduc, M., Grandbois, M., Heveker, N.,
constitutively active, ligand access into ER membranes is not Bkaily, G., Chemtob, S., Barbaz, D., 2006. G-protein-coupled receptors signalling
254 K.B.S. Chow et al. / Molecular and Cellular Endocrinology 348 (2012) 247–254

at the cell nucleus: an emerging paradigm. Can. J. Physiol. Pharmacol. 84, 287– Mercier, J.-F., Salahpour, A., Angers, S., Breit, A., Bouvier, M., 2002. Quantitative
297. assessment of b1- and b2-adrenergic receptor homo- and heterodimerization by
Grosse, R., Schöneberg, T., Schultz, G., Gudermann, T., 1997. Inhibition of bioluminescence resonance energy transfer. J. Biol. Chem. 277, 44925–44931.
gonandotropin-releasing hormone receptor signaling by expression of a splice Milligan, G., 2004. G protein-coupled receptor dimerization: function and ligand
variant of the human receptor. Mol. Endocrinol. 11, 1305–1318. pharmacology. Mol. Pharmacol. 66, 1–7.
Guan, R., Feng, X., Wu, X., Zhang, M., Zhang, X., Hébert, T.E., Segaloff, D.L., 2009. Milligan, G., 2010. The role of dimerisation in the cellular trafficking of G-protein-
Bioluminescence resonance energy transfer studies reveal constitutive coupled receptors. Curr. Opin. Pharmacol. 10, 23–29.
dimerization of the human lutropin receptor and a lack of correlation Okuda-Ashitaka, E., Sakamoto, K., Ezashi, T., Miwa, K., Ito, S., Hayaishi, O., 1997.
between receptor activation and the propensity for dimerization. J. Biol. Suppression of prostaglandin E receptor signaling by the variant form of EP1
Chem. 284, 7483–7494. subtype. J. Biol. Chem. 271, 31255–31261.
Holliday, N.D., Holst, B., Rodionova, E.A., Schwartz, T.W., Cox, H.M., 2007. Pantel, J., Legendre, M., Cabrol, S., Hilal, L., Hajaji, Y., Morisset, S., Nivot, S., Vie-Luton,
Importance of constitutive activity and arrestin independent mechanisms for M.P., Grouselle, D., de Kerdanet, M., Kadiri, A., Epelbaum, J., Le Bouc, Y.,
intracellular trafficking of the ghrelin receptor. Mol. Endocrinol. 21, 3100–3112. Amselem, S., 2006. Loss of constitutive activity of the growth hormone
Holst, B., Cygankiewicz, A., Jensen, T.H., Ankersen, M., Schwartz, T.W., 2003. High secretagogue receptor in familial short stature. J. Clin. Invest. 116, 760–768.
constitutive signaling of the ghrelin receptor – identification of a potent inverse Petersen, P.S., Woldbye, D.P.D., Madsen, A.N., Egerod, K.L., Jin, C., Lang, M.,
agonist. Mol. Endocrinol. 17, 2201–2210. Rasmussen, M., Beck-Sickinger, A.G., Holst, B., 2009. In vivo characterization
Howard, A.D., Feighner, S.D., Cully, D.F., Arena, J.P., Liberator, P.A., Rosenblum, C.I., of high basal signaling from the ghrelin receptor. Endocrinology 150, 4920–
Hamelin, M., Hreniuk, D.L., Palyha, O.C., Anderson, J., Paress, P.S., Diaz, C., Chou, 4930.
M., Liu, L.-Y., McKee, K.K., Pong, S.-S., Chaung, L.-Y., Elbrecht, A., Dashkevicz, M., Robben, J.H., Kortenoeven, M.L.A., Sze, M., Yae, C., Milligan, G., Oorschot, V.M.,
Heavens, R., Rigby, M., Sirinathsinghji, D.J.S., Dean, D.C., Melillo, D.G., Patchett, Klumperman, J., Knoers, N.V.A.M., Deen, P.M.T., 2009. Intracellular activation of
A.A., Nargund, R., Griffin, P.R., DeMartino, J.A., Gupta, S.K., Schaeffer, J.M., Smith, vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by
R.G., Van der Ploeg, L.H.T., 1996. A receptor in pituitary and hypothalamus that nonpeptide agonists. Proc. Natl. Acad. Sci. 106, 12195–12200.
functions in growth hormone release. Science 273, 974–977. Salahpour, A., Angers, S., Bouvier, M., 2000. Functional significance of
Ibrahim, S., Tetruashvily, M., Frey, A.J., Wilson, S.J., Stitham, J., Hwa, J., Smyth, E.M., oligomerization of G-protein-coupled receptors. Trends Endocrinol. Metabol.
2010. Dominant negative actions of human prostacyclin receptor variant 11, 163–168.
through dimerization: implications for cardiovascular disease. Arterioscler. Salahpour, A., Angers, S., Mercier, J.F., Lagacé, M., Marullo, S., Bouvier, M., 2004.
Thromb. Vasc. Biol. 30, 1802–1809. Homodimerization of the b2-adrenergic receptor as a prerequisite for cell
James, J.R., Oliveira, M.I., Carmo, A.M., Iaboni, A., Davis, S.J., 2006. A rigorous surface targeting. J. Biol. Chem. 279, 33390–33397.
experimental framework for detecting protein oligomerization using Sánchez-Laorden, B.L., Herraiz, C., Valencia, J.C., Hearing, V.J., Jiménez-Cervantes, C.,
bioluminescence resonance energy transfer. Nat. Methods 3, 1001–1006. García-Borrón, J.C., 2009. Aberrant trafficking of human melanocortin 1 receptor
Jia, Y., Nguyen, T., Desai, M., Ross, M.G., 2008. Programmed alterations in variants associated with red hair and skin cancer: steady-state retention of
hypothalamic neuronal orexigenic responses to ghrelin following gestational mutant forms in the proximal Golgi. J. Cell. Physiol. 220, 640–654.
nutrient restriction. Reprod. Sci. 15, 702–709. Smith, R.G., Jiang, H., Sun, Y., 2005. Developments in ghrelin biology and potential
Karpa, K.D., Lin, R., Kabbani, N., Levenson, R., 2000. The dopamine D3 receptor clinical relevance. Trends Endocrinol. Metabol. 16, 436–442.
interacts with itself and the truncated D3 splice variant D3nf: D3–D3nf Sun, Y., Butte, N.F., Garcia, J.M., Smith, R.G., 2008. Characterization of adult ghrelin
interaction causes mislocalization of D3 receptors. Mol. Pharmacol. 58, 677– and ghrelin receptor knockout mice under positive and negative energy
683. balance. Endocrinology 149, 843–850.
Kern, A., Hubbard, D., Amano, A., Bryant-Greenwood, G.D., 2008. Cloning, Takahashi, K., Furukawa, C., Takano, A., Ishikawa, N., Kato, T., Hayama, S., Suzuki, C.,
expression, and functional characterization of relaxin receptor (leucine-rich Yasui, W., Inai, K., Sone, S., Ito, T., Nishimura, F., Tsuchiya, E., Nakamura, Y.,
repeat-containing G protein-coupled receptor 7) splice variants from human Daigo, Y., 2006. The neuromedin U-growth hormone secretagogue receptor 1b/
fetal membranes. Endocrinology 149, 1277–1294. neurotensin receptor 1 oncogenic signaling pathway as a therapeutic target for
Kojima, M., Kangawa, K., 2010. Ghrelin: more than endogenous growth hormone lung cancer. Cancer Res. 66, 9408–9419.
secretagogue. Ann. N.Y. Acad. Sci. 1200, 140–148. Trettel, F., Bartolomeo, S., Lauro, C., Catalano, M., Ciotti, M.T., Limatola, C., 2003.
Korbonits, M., Bustin, S.A., Kojima, M., Jordan, S., Adams, E.F., Lowe, D.G., Kangawa, Ligand-independent CXCR2 dimerization. J. Biol. Chem. 278, 40980–40988.
K., Grossman, A.B., 2001. The expression of the growth hormone secretagogue van Rijn, R.M., van Marle, A., Chazot, P.L., Langemeijer, E., Qin, Y., Shenton, F.C., Lim,
receptor ligand ghrelin in normal and abnormal human pituitary and other H.D., Zuiderveld, O.P., Sansuk, K., Dy, M., Smit, M.J., Tensen, C.P., Bakker, R.A.,
neuroendocrine tumors. J. Clin. Endocrinol. Metab. 86, 881–887. Leurs, R., 2008. Cloning and characterization of dominant negative splice
Kumar, V., Jong, Y.-J., O’Malley, K.L., 2008. Activated nuclear metabotropic variants of the human histamine H4 receptor. Biochem. J. 414, 121–131.
glutamate receptor mGlu5 couples to nuclear Gq/11 proteins to generate Waseem, T., Duxbury, M., Ito, H., Ashley, S.W., Robinson, M.K., 2008. Exogenous
inositol 1,4,5-trisphosphate-mediated nuclear Ca2+ release. J. Biol. Chem. 283, ghrelin modulates release of pro-inflammatory and anti-inflammatory
14072–14083. cytokines in LPS-stimulated macrophages through distinct signaling
Lau, P.N., Chow, K.B., Chan, C.B., Cheng, C.H., Wise, H., 2009. The constitutive activity pathways. Surgery 143, 334–342.
of the ghrelin receptor attenuates apoptosis via a protein kinase C-dependent Zhao, T.-J., Liang, G., Li, R.L., Xie, X., Sleeman, M.W., Murphy, A.J., Valenzuela, D.M.,
pathway. Mol. Cell. Endocrinol. 299, 232–239. Yancopoulos, G.D., Goldstein, J.L., Brown, M.S., 2010. Ghrelin O-
Leung, P.K., Chow, K.B.S., Lau, P.N., Chu, K.M., Chan, C.B., Cheng, C.H.K., Wise, H., acetlytransferase (GOAT) is essential for growth hormone-mediated survival
2007. The truncated ghrelin receptor polypeptide (GHS-R1b) acts as a of calorie-restricted mice. Proc. Natl. Acad. Sci. USA 107, 7467–7472.
dominant-negative mutant of the ghrelin receptor. Cell. Signal. 19, 1011–1022. Zhou, F., Filipeanu, C.M., Duvernay, M.T., Wu, G., 2006. Cell-surface targeting of a2-
Lohse, M.J., 2010. Dimerization in GPCR mobility and signaling. Curr. Opin. adrenergic receptors–Inhibition by a transport deficient mutant through
Pharmacol. 10, 53–58. dimerization. Cell. Signal. 18, 318–327.
Maggio, R., Novi, F., Scarselli, M., Corsini, G.U., 2005. The impact of G-protein- Zhu, X., Wess, J., 1998. Truncated V2 vasopressin receptors as negative regulators of
coupled receptor hetero-oligomerization on function and pharmacology. FEBS J. wild-type V2 receptor function. Biochemistry 37, 15773–15784.
272, 2939–2946.

You might also like