ArticlePDF Available

Agonism, Antagonism, and Inverse Agonism Bias at the Ghrelin Receptor Signaling

Authors:

Abstract

The G protein-coupled receptor GHS-R1a mediates ghrelin-induced growth hormone secretion, food intake and reward-seeking behaviours. GHS-R1a signals through Gq, Gi/o, G13 and arrestin. Biasing GHS-R1a signaling with specific ligands may lead to the development of more selective drugs to treat obesity or addiction with minimal side effects. To delineate ligand selectivity at GHS-R1a signaling, we analysed in detail the efficacy of a panel of synthetic ligands at activating the different pathways associated to GHS-R1a in HEK293T cells. Besides β-arrestin2 recruitment and ERK1/2 phosphorylation, we monitored activation of a large panel of G protein subtypes using a BRET2-based assay with G protein-activation biosensors. We first found that unlike full agonists, Gq partial agonists were unable to trigger β-arrestin2 recruitment and ERK1/2 phosphorylation. Using G protein-activation biosensors, we then demonstrated that ghrelin promoted activation of Gq, Gi1, Gi2, Gi3, Goa, Gob and G13 but not Gs and G12. Besides, we identified some GHS-R1a ligands that preferentially activated Gq and antagonized ghrelin-mediated Gi /Go activation. Finally, we unambiguously demonstrated that in addition to Gq, GHS-R1a also promoted constitutive activation of G13. Importantly, we identified some ligands that were selective inverse agonists toward Gq but not of G13. This demonstrates that bias at GHS-R1a signaling can occur not only with regard to agonism but also to inverse agonism. Our data, combined to further in vivo studies, may facilitate the design of drugs selectively targeting individual signaling pathways to treat only the therapeutically relevant function.
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
"!
Agonism, antagonism and inverse agonism bias at the Ghrelin receptor
signaling
Céline M’Kadmi
§
, Jean-Philippe Leyris
, Lauriane Onfroy
, Céline Galés
, Aude Saulière
, Didier
Gagne
§
, Marjorie Damian
§
, Sophie Mary
§
, Mathieu Maingot
§
, Séverine Denoyelle
§
, Pascal Verdié
§
,
Jean-Alain Fehrentz
§
, Jean Martinez
§
, Jean-Louis Banères
§
and Jacky Marie
§1
From the
§
Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université
Montpellier-ENSCM, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093
Montpellier cedex 05, France.
Institut des Neurosciences de Montpellier, Hôpital Saint-Eloi, 80 avenue Augustin Fliche, BP 74103,
34091, Montpellier cedex 05, France.
Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la
Recherche Médicale, U1048, Université Toulouse III Paul Sabatier, Centre Hospitalier Universitaire
de Toulouse, 31432, Toulouse, France
Running title: Biased signaling at the ghrelin receptor
To whom correspondence should be addressed: Jacky Marie, Institut des Biomolécules Max
Mousseron (IBMM), UMR 5247 CNRS-Universi Montpellier-ENSCM, Faculté de Pharmacie, 15
avenue Charles Flahault, BP 14491, 34093 Montpellier cedex 05, France. Tel: 33 4 11759753, Fax: 33
4 1175 9760, Email: jacky.marie@univ-montp1.fr
Keywords: G protein-coupled receptor (GPCR), ghrelin, signaling bias, G protein subtypes,
bioluminescence resonance energy transfer (BRET).
Background: GHS-R1a activates multiple
signaling pathways mediating feeding and
addictive behaviours.
Results: Some GHS-R1a ligands activate Gq but
not Gi/o and fail to recruit β-arrestin2, others act
as selective inverse agonists at Gq compared to
G13.
Conclusion: Synthetic ligands can selectively
activate or reverse Gq-dependent signaling at
GHS-R1a.
Significance: Ligand-biased signaling can be
exploited for the development of selective drugs
to treat GHS-R1a-mediated disorders.
ABSTRACT
The G protein-coupled receptor GHS-R1a
mediates ghrelin-induced growth hormone
secretion, food intake and reward-seeking
behaviours. GHS-R1a signals through Gq,
Gi/o, G13 and arrestin. Biasing GHS-R1a
signaling with specific ligands may lead to the
development of more selective drugs to treat
obesity or addiction with minimal side effects.
To delineate ligand selectivity at GHS-R1a
signaling, we analysed in detail the efficacy of
a panel of synthetic ligands at activating the
different pathways associated to GHS-R1a in
HEK293T cells. Besides βarrestin2
recruitment and ERK1/2 phosphorylation, we
monitored activation of a large panel of G
protein subtypes using a BRET
2
-based assay
with G protein-activation biosensors. We first
found that unlike full agonists, Gq partial
agonists were unable to trigger β-arrestin2
recruitment and ERK1/2 phosphorylation.
Using G protein-activation biosensors, we
then demonstrated that ghrelin promoted
activation of Gq, Gi1, Gi2, Gi3, Goa, Gob and
G13 but not Gs and G12. Besides, we
identified some GHS-R1a ligands that
preferentially activated Gq and antagonized
ghrelin-mediated Gi /Go activation. Finally,
we unambiguously demonstrated that in
addition to Gq, GHS-R1a also promoted
constitutive activation of G13. Importantly,
we identified some ligands that were selective
inverse agonists toward Gq but not of G13.
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
#!
This demonstrates that bias at GHS-R1a
signaling can occur not only with regard to
agonism but also to inverse agonism. Our
data, combined to further in vivo studies, may
facilitate the design of drugs selectively
targeting individual signaling pathways to
treat only the therapeutically relevant
function.
!
INTRODUCTION
Ghrelin, a peptide hormone mainly produced by
the stomach (1), has emerged as an important
gut-brain signal to control growth hormone
secretion, food intake and reward seeking
behaviours (2,3). Ghrelin mediates these actions
through the growth hormone secretagogue type
1a receptor (GHS-R1a) receptor, a family A G-
protein coupled receptor (GPCR) (4,5). Because
of its possible implication in several
physiological disorders such as obesity, drug and
alcohol addiction, GHS-R1a represents a major
target for the development of therapeutic
molecules (6). So far, several academic
laboratories and pharmaceutical companies have
developed synthetic molecules that display
agonist, antagonist and inverse agonist properties
toward GHS-R1a intracellular signalling
pathways. Some of these molecules display
interesting properties with regard to food intake
stimulation or inhibition (7-9), addiction to drugs
including alcohol and cocaine (10,11) or growth
hormone secretion (12). Given the pleiotropic
actions of ghrelin, GHS-R1a synthetic ligands
can be useful to block or activate the targeted
physiological effect but can also lead to
undesirable side effects. For instance, synthetic
GHS-R1a antagonists that decrease food intake
and fat storage may be good candidate to treat
obesity but could have side effects due to their
inhibitory action on growth hormone secretion.
In the other way, agonists developed with the
aim to stimulate hormone secretion for treating
postmenopausal osteoporosis may have adverse
effect by increasing body weight (13). Therefore,
development of biased ligands that will
selectively inhibit or activate only one or a
subset of the GHS-R1a dependent physiological
responses could have significant therapeutic
advantages. This aim is certainly now attainable
because during the last decade, many
independent studies on GPCRs have described
biased agonists that are selective of a given
down-stream signaling pathways (14,15). Unlike
the endogenous ligand that usually activates all
signaling pathways the receptor is associated
with (G protein and β-arrestin dependent
pathways), synthetic ligands could thus
selectively activate only some of them (for
instance activation of!β -arrestin with no effect on
G protein or conversely) (16). Although
molecular mechanisms responsible for ligand-
directed functional selectivity are not fully
understood, there are increasing evidences that
biased activity results from selective stabilization
of different receptor conformations that differ
into their ability to couple to different
downstream effectors (17-21). Importantly, in
vivo studies demonstrated that side effects of
classical drugs might be diminished by the use of
biased molecules, suggesting this kind of
molecules could have potential clinical
application (16,22). As for many other GPCR,
rational design of biased ligands of the ghrelin
receptor first requires identification of lead
pathway-selective compounds. To identify such
lead compounds, careful dissection of the
different intracellular down-stream signaling
pathways of GHS-R1a is required. GHS-R1a is
coupled to Gq signaling pathway to trigger
inositol phosphate production and intracellular
calcium release (23).In the context of Gq-
signaling, an interesting particularity of GHS-
R1a is its exceptionally high constitutive
activity. Indeed, high basal levels of inositol
phosphate production were detected in GHS-
R1a-transfected cell lines (24,25). Significant
ligand independent Gq activation and AP2
recruitment were also clearly demonstrated to
occur with the purified GHS-R1a inserted in
lipid disc (26). As many other GPCRs, GHS-R1a
activates other G protein dependent and
independent pathways besides the Gq-associated
one. Indeed, following ghrelin stimulation, GHS-
R1a activates ERK1/2 through β-arrestin
dependent (27,28) and β-arrestin independent
(29) pathways. It also activates PI3 kinase,
PKCε, Src through a Gi/o protein dependent
pathway (28,30).
In this context, we investigated here the
selectivity of a panel of GHS-R1a synthetic
ligands toward arrestin and G protein dependent
pathways. We paid particular attention to the
selectivity of ligands toward activation of several
G protein subtypes and isoforms thanks to the
use of recently developed G protein BRET-based
biosensors that were recently developed (31,32).
Our data suggest that some synthetic GHS-R1a
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
$!
ligands are selective Gq-agonists. We also
identified ligands that displayed potential inverse
agonist selectivity toward Gq compared to G13.
The occurrence of such pathway-selective
ligands questions about the connection that could
exist between the biased behaviour of some
ligands toward intracellular pathways and their
selectivity toward food intake and GH secretion
already demonstrated in vivo.
!
EXPERIMENTAL PROCEDURES
Materials
Ghrelin 1-28 was purchased from polypeptide
group, MK-0677 (33) from Axon MedChem and
[D-Arg1-D-Phe5,D-Trp7,9,Leu11] substance P
(SPA) from Bachem. JMV compounds were
synthesized in our laboratory (IBMM, France).
The pseudopeptide JMV 1843 was previously
described by Guerlavais et al. (34), JMV 2959 by
Moulin et al. (35), JMV 3002, JMV 3018 and
JMV 3011 by Moulin et al. (36) In compound
JMV 4484, a second chiral center was introduced
in position 3 of the 1,2,4-triazole scaffold. This
chiral center contains an amino function, which
was elongated by the Leu-Leu dipeptide and a
lysine residue was then introduced in the N-
terminal part to mimic the peptide core of the
substance P analog. Peptide KwFwLL-NH
2
and
K-(D-1Nal)-FwLL-NH
2
were synthetized at
IBMM as described in (37) and (38)
respectively.
The thromboxane A2 receptor agonist U46619
was purchased from Cayman chemical. Arginine
vasopressin (AVP) was provided by Dr. B.
Mouillac (IGF, Montpellier, France).
Lipofectamine 2000, foetal bovine serum,
antibiotics (penicillin, streptomycin) and DMEM
medium were purchased from Invitrogen.
Coelenterazine 400a (DeepblueC) was
purchased from Interchim. IPone HTRF kit and
benzyl guanine-Tb3
+
-cryptate were provided by
CisBio. BODIPY FL GTPγS was from
Invitrogen.
For experiments in lipid disc, Gαq and Gβ1γ2
subunits were produced in sf9 cells as described
(18). Gα13 was from Kerafast.!
Plasmid constructions.
RLuc-β-arrestin2 cloned in PRK6 vector was a
generous gift of Dr. M.A Ayoub (IGF,
Montpellier, France). Human vasopressin V2
receptor cloned in PRK5 vector was provided by
Dr. T. Durroux (IGF, Montpellier, France).
Human GHS-R1a cloned in pcDNA3.1+
vector
(pcDNA-GHS-R1a) was purchased from the
cDNA resource Center (University of Missouri).
HA-GHS-R1a was generated by PCR using: 1) a
sense oligonucleotide primer containing HindIII
and EcoRI sites followed by a Kozac sequence,
an ATG codon, a HA sequence and nucleotides
1-25 of GHS-R1a; 2) an antisense
oligonucleotide primer containing a BamHI site
followed by a stop codon and nucleotides 1098-
1072 of GHS-R1a. The PCR product was
digested with HindIII and BamHI and cloned in
HindIII-BamHI sites of pcDNA3+ vector
(pcDNA-HA-GHS-R1a). To generate GHS-R1a-
YFP, GHS-R1a sequence was amplified by PCR
with a sense oligonucleotide containing an
EcoRI site and an antisense oligonucleotide
containing a BamHI site. The PCR fragment was
digested with EcoRI and BamHI and inserted
into EcoRI and BamHI sites of PRK6-YFP
vector provided by Dr. M.A Ayoub (IGF,
Montpellier, France).! A204E mutation was
introduced in GHS-R1a (GHS-R1a-A204E) by
PCR using pcDNA-GHS-R1a as a template and
30 mer forward and reverse oligonucleotide
primers in Accuprime Pfx SuperMix solution
(Invitrogen). SNAP-GHS-R1a was already
described (39)
Cell culture.
HEK293T cells were maintained in DMEM
Glutamax (Invitrogen) supplemented with
antibiotics (penicillin 50 µg/ml, streptomycin 50
µg/ml), HEPES 2 mM, 1% non-essential amino
acids and 10% heat inactivated Foetal Calf
Serum.
!
Transfection.
For IP and binding assays, transfections were
performed in 96 well plates using cell density of
50,000 cells per well. Prior to cell plating, wells
were pre-coated with Poly-L-Ornithine (50 µl of
10 mg/ml) for 30 minutes at 37°C. Transfection
mixes were prepared using cDNA encoding
GHS-R1a, GHS-R1a A204E or SNAP-GHS-R1a
(200 ng-300 ng), lipofectamine 2000
(Invitrogen) with a ratio 0.4 for cDNA (µg) /
lipofectamine (µl) in a total volume of 50 µl
optiMEM culture medium per well. Prior to its
addition in plates, the transfection mixture was
pre-incubated for 20 min at room temperature.
Then 100 µl of HEK293T cells at a density of
500,000 cells/ml were plated in each well and
were incubated at 37°C under 5% CO
2
for 48 h.
Transfection condition for HTRF ligand binding
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
%!
was performed as previously described (39).!
Ligand binding assay.
Ki values were determined from binding
competition experiments performed on intact
HEK293T cells expressing the GHS-R1a using a
Homogenous Time Resolved Fluorescence
(HTRF) assay previously described (39). HTRF
signal was collected in a PHERAstar microplate
reader (BMG LABTECH). Ki values were
obtained from binding curves using GraphPad
Prism software (GraphPad Software, Inc., San
Diego, CA).
The expression level (B
max
) of GHS-R1a
expressed in HEK293T cells was determined by
radioactive assay using [
125
I]-His
9
ghrelin as
previously described (39). !
Inositol Phosphate assay.
Inositol phosphate accumulation assay was
carried out 48 h after transfection on adherent
cells in 96-well plate at a density of 50,000
cells/well. IP1 production was measured using
the IP-One HTRF kit (Cisbio Bioassays Ref.
621PAPEC) as previously described (39).
Briefly, cells were stimulated for 30 min at 37°C
with the ligand to be tested in 70 µl of IP1
stimulation buffer. An anti-IP1 antibody labelled
with Lumi4-Tb (15 µl) and an IP1-d2 derivative
(15 µl) were added on cells. The medium was
incubated for 1 h at room temperature. Signals at
665 nm and 620 nm were detected using a
PHERAstar (BMG LABTECH) fluorescence
reader. Values are expressed as F. F
corresponded to: (Ratio 665 nm/620 nm of the
assay - Ratio 665 nm/620 nm of the negative
control)/ Ratio 665nm/620nm of the negative
control.
The negative control corresponded to the Lumi4-
Tb blank and was used as an internal assay
control. Inositol phosphate accumulation was
expressed as the percentage of the maximal
ghrelin response using the formula: (F mock
cells-F receptor transfected cells)/(F mock
cells-F maximal! ghrelin stimulation for
receptor transfected cells).!!
!
ERK1/2 assay.
ERK1/2 assay was carried out 48 h after
transfection on adherent cells in 96-well white
plates (Greiner Bio One) at a density of 50,000
cells/well. ERK1/2 phosphorylation was
measured after 10 min stimulation with ligands
using an HTRF-based Phospho-ERK
(Thr202/Tyr204) cellular assay kit according to
manufacturer instructions (Cisbio Bioassays).
Briefly signal is detected between anti-phospho
ERK antibody labelled with Eu3
+
cryptate donor
and anti ERK1/2 antibody labelled with d2
acceptor. Signals at 665 nm and 620 nm were
measured using a PHERAstar (BMG
LABTECH) fluorescence reader. Values were
expressed as ratio of 665 nm/ 620 nm x 1000.
Arrestin recruitment assay.
The interaction between GHS-R1a-YFP and
Rluc-β-arrestin2 was measured in HEK 293T by
BRET
1
in 96 well white plates (Greiner Bio
One). Briefly cells were transfected by
lipofectamine with 100 ng of GHS-R1aYFP
and 5 ng Rluc-β-arrestin2. 48 h after transfection
cells were washed with PBS and then incubated
for 45 min at 37°C with 50 µ l of ligand in
DMEM, 0.1% BSA. After stimulation, cells were
washed with 100 µl PBS. 50 µl of a 0.5 mM
coelenterazine H (Interchim) solution in PBS
was then added to the cells and the signal
measured with a Mithras LB 940 plate reader
(Berthold Biotechnologies) that allows
sequential integration of luminescence signal (5
cycles of 0.05 sec) with two filter settings (Rluc
filter, 485 ± 20 nm and YFP filter, 530 ± 25 nm).
The BRET ratio was defined as the difference of
the ratio 530 nm/485 nm of the co-transfected
Rluc and YFP proteins and the ratio of the Rluc
protein alone. Results are expressed in mBRET
corresponding to the ratio (530 nm/485 nm)
x1000.
!
G protein activation BRET assay.
G protein activation was measured with the
BRET assay previously described (31,32).
Briefly HEK293T cells grown in 10 cm culture
dishes were co-transfecetd by lipofectamine
2000 with GHS-R1a and G protein subunits
(Rluc8-α, β1 and γ2-GF10). 48 h after
transfection, cells were washed with PBS,
detached with PBS containing 5 mM EDTA, and
resuspended in PBS supplemented with 5 mM
EDTA and 0.1% (w/v) glucose (buffer A) at
room temperature. Cells were then distributed in
a 96-well white plate (300,000 cells per well).
For kinetic analyses, 5 µM of deep blue C
(coelenterazine 400a, Interchim) were added and
the plate was immediately loaded in a Mithras
LB 940 multimode microplate reader (Berthold)
or a PHERAstar microplate reader (BMG
LABTECH). Then, 10 µl of ligand solution (1
µM) was injected after 30 sec of reading and
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
&!
signal was recorded for 90 sec. The BRET signal
was obtained by calculating the ratio of GFP10
emission (515 ± 10 nm) over Rluc8 light
emission (400 ± 10 nm) at 1.6 sec intervals. For
end point measurements, cells (300,000) in 80
µl of buffer A were incubated in a 96 well plate
with 10 ml of ligand solution in buffer A at room
temperature for 3 min to 15 min. Then, 10 µl of
deep blue C solution (50 mM) was added and the
BRET
2
signal was recorded in a PHERAstar
microplate reader (BMG labtech). The BRET
signal was calculated as the ratio of emission
GFP10 (510-530 nm) to RLuc8 (410-480 nm)
recorded 5 times at 0.5 sec intervals.
Quantification of cell surface receptors by
ELISA.
24 h post-transfection with pcDNA3.1 (+)
(control) or vectors encoding N-terminally HA-
tagged GHSR1a either in presence of Gαq-Rluc8
or Gα13-Rluc8, untagged GFP10-Gγ2 and Gβ1
untagged, cells were split into 24-well plates.
Cells were fixed in 4 % paraformaldehyde,
saturated with PBS containing 1 % bovine serum
albumin and incubated with the primary anti-HA
antibody (Clone 16B12, Covance) and then with
HRP-labeled secondary antibody (Sigma). After
washing, cells were incubated with HRP
substrate: TMB (3,39,5,59-
tetramethylbenzidine). The reaction was stopped
with 1N HCl, and the plates were read at 450 nm
in a microplate reader (Varioscan Flash, thermo
electron). The 570-nm optic density
(background) was subtracted.
G-protein activation assay in lipid discs.
The human GHS-R1a receptor was expressed in
E. coli and assembled as a monomer into lipid
discs as described (26). GTPγS binding assays
were carried out by monitoring changes in the
fluorescence emission of bodipyGTPγS (26). In
these assays, ligand concentrations of 1µM and
1:10 receptor:G protein molar ratios with
receptor concentrations in the 20 nM range were
used.
Quantification of ligand bias.
EC
50
and E
max
values were estimated from dose-
response curves using the nonlinear curve fitting
equation (three parameters) in GraphPad Prism
(V5.0).
Ligand bias was quantify by fitting ligand
concentration-response curve using the method
developed by Kenakin et al. (40), which is based
on the operational model of agonism (41). The
transduction coefficient log (τ/Κ
A
) was derived
using the operational model equation in
GraphPad Prism. This transduction coefficient
represents the ability of an agonist to stimulate a
given signaling pathway. τ represents an index
of coupling efficiency of the agonist, K
A
is the
functional equilibrium dissociation constant of
the agonist. To eliminate the impact of the
different sensitivities of the assays used, the [log
(τ/Κ
A
)] value determined for one ligand at a
given pathway was normalized to that
determined for a reference ligand at the same
pathway. In our study, the reference ligand was
ghrelin. By subtracting log (τ/Κ
A
) of ghrelin
from log (τ/Κ
A
) of each compound, a log (t/K
A
)
value was obtained that gave a within-pathway
comparison of transduction efficiency of ligands:
log (τ/K
A
) = log (τ/Κ
A
)
compound
- log (τ/Κ
A
)
ghrelin
Finally, the bias of each ligand between different
signaling pathways was obtained in the form
∆∆log(τ/K
A
) which was:
∆∆log(τ/K
A
)
P1-P2
= log (τ/K
A
)
P1
-log (τ/K
A
)
P2
where P1 is pathway 1 and P2 is pathway 2.
No ligand bias at two different pathways
compared to ghrelin will result in a value of
∆∆log(τ/K
A
)
P1-P2
not significantly different from
0. Statistical analysis was performed using a
two-way unpaired Student’s t test. Difference
were considered significant when p was < 0.05.
Bias factor (BF) (or fold change in bias) was
calculated as follows: BF= 10
∆∆log(
τ
/K
A
)
When a ligand promoted a detectable stimulation
of one pathway (P1) but did not promoted any
detectable stimulation at the other pathway (i.e
P2), a bias factor of this ligand between these
two pathways could not be calculated.
RESULTS
Binding properties of GHS-R1a ligands.
The binding affinities (Ki values) of the GHS-
R1a ligands used all along this study are reported
in Table 1.
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
'!
Efficacy of GHS-R1a ligands toward Inositol
phosphate production.
First, we tested a panel of GHS-R1a ligands for
their efficacy to stimulate Gq/G11 signaling by
measuring inositol phosphate production in
HEK-293T cells transiently expressing GHS-
R1a.
As already published, GHS-R1a expressing cells
displayed a high basal level of inositol phosphate
production compared to un-transfected cells and
this basal activity was reversed in the presence of
the inverse agonist SPA (Fig. 1A). Thus, in the
HEK293T cells used all along this study, the
GHS-R1a displayed high constitutive activity
that amounts 50 to 70 % of the maximal
response promoted by ghrelin (Fig. 1A). The
pseudo-peptide JMV 1843 stimulated inositol
phosphate production to the same extent than
ghrelin, whereas the efficacy of the non-peptide
MK-0677 was slightly higher than that of ghrelin
(150 ± 25 % compared to 100% ghrelin
stimulation over basal) (Fig. 1A, Table 2).
Compared to ghrelin, JMV 1843 and MK-0677
compounds thus behaved as full and super
agonist respectively compared to ghrelin on
inositol phosphate pathway. When tested at a 10
-
6
M maximal dose, JMV 3011 had no effect
whereas JMV 3002, JMV 3018 and JMV 2959
induced an increase of inositol phosphate
production over basal with a partial agonist
effect compared to ghrelin (Fig. 1A, Table 2).
Thus, JMV 3002, JMV 3018 and JMV 2959 can
thus be considered as partial agonists on the IP
pathway (Fig. 1A, Table 2). However, the partial
agonist efficacy of these ligands was sometime
undetectable when the basal IP1 production
reached 60 % of the maximal ghrelin response, a
level often found in these experiments. We thus
postulated that the high level of basal IP1
production might mask this partial agonist effect
of ligands. Therefore, we checked whether we
could decrease the constitutive activity of GHS-
R1a by decreasing the expression level of GHS-
R1a.
The basal level of IP1 production of HEK293T
cells expressing the GHS-R1a represented 55 ± 3
% and 62 ± 4 % of the maximal ghrelin for a
receptor expression level of 2.5 fmol/10
5
cells
and 74 fmol/10
5
cells respectively. Thus,
decreasing the receptor expression level by a
factor 29.6 did not change significantly the level
of constitutive activity expressed as the % of
maximal ghrelin (Fig. 1B). This is due to the fact
that decreasing the receptor expression level
induced a concomitant decrease of the basal
level of IP1 production with a concomitant
decrease of the maximal level promoted by
ghrelin. Decreasing the receptor expression level
did not allow to better visualize the partial
agonist character of the ligands (Fig. 1C). We
therefore decided to evaluate the efficacy of
these ligands to promote IP1 production on a
GHS-R1a mutant (GHS-R1a A204E) with a
decreased basal activity. The A204E mutation
decreased the basal activity of the receptor
without changing its ability to respond to ghrelin
compared to the WT receptor (27,42). As shown
in Fig. 1D, the basal level of IP1 production of
HEK293T cells expressing the GHS-R1a A204E
was highly decreased compared to the WT
receptor, whereas both the efficacy and potency
of full agonists to induce IP1 production were
unaffected. Interestingly, as shown in Fig 1D,
whereas JMV 3011 remained neutral, the partial
agonist activity of JMV 3002, JMV 3018 and
JMV 2959 unambiguously detected with the
GHS-R1a A204E mutant. Although binding
characteristic (Ki) of ligands are quite similar
between the WT and the A204E mutant
receptors (Table 1), we cannot totally exclude,
however, that the A204E mutation induces
discreet changes in binding and signaling
properties of ligands. Nevertheless, our
observation suggested that an extremely high
constitutive activity might hide the partial
agonist character of some ligands. The behaviour
of the different ligands with regard to GHS-R1a
A204 was confirmed by testing them in
competition with ghrelin. In this cas, JMV 3011
fully antagonized the effect of ghrelin at
promoting IP1 production whereas ligands JMV
3002, JMV 3018 and JMV 2959 antagonized
only partially the ghrelin effect (Fig. 3A).
Efficacy of GHS-R1a ligands toward β-
arrestin2 recruitment and ERK1/2 activation.
As previously reported, ghrelin stimulation
promotes β-arrestin2 recruitment to GHS-R1a
(26,27). The level of constitutive β-arrestin2
recruitment was lower than that of IP1
production. SPA reduced almost to zero the basal
level of β-arrestin2 recruitment, suggesting that
this ligand displayed inverse agonist efficacy
toward β-arrestin2 recruitment also (Fig. 2A and
C). As expected, ghrelin, JMV 1843 and MK-
0677 promoted a large increase in β-arrestin2
recruitment to GHS-R1a (Fig. 2A and C). As
observed for IP1 production, MK-0677 behaved
as a super-agonist toward β-arrestin-2
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
(!
recruitment compared to ghrelin and JMV 1843.
In contrast, no β-arrestin2 recruitment was
observed upon stimulation with JMV 3011, JMV
3002, JMV 3018 and JMV 2959 (Fig. 2A and C).
Thus, JMV 3011 was neutral with regard to both
IP1 production and β-arrestin2 recruitment
whereas JMV 3002, JMV 3018 and JMV 2959
were partial agonists on IP1 production but
neutral toward β-arrestin2 recruitment (Fig. 1,
2A and B). Because it was reported that GHS-
R1a activated ERK1/2 through Gq/11, Gi and
arrestin dependent pathways (28,43,44), we
tested the efficacy of our ligands toward ERK1/2
activation. All compounds displayed comparable
efficacy to promote β-arrestin2 recruitment
(Table 2, Fig. 2A and C) and ERK1/2
phosphorylation (Fig. 2B and D). Whereas
ghrelin, MK-0677 and JMV 1843 promoted
ERK1/2 phosphorylation, ligands JMV 3002,
JMV 3018, JMV 2959 and JMV 3011 were
neutral toward ERK1/2 phosphorylation (Fig. 2B
and D). When tested in competition with ghrelin,
JMV 3002, JMV 3018, JMV 2959 and JMV
3011 totally antagonized both β-arrestin2
recruitment (Fig. 3B) and ERK1/2
phosphorylation promoted by ghrelin (Fig. 3C).
Taken together, these data indicate that GHS-
R1a partial agonists of IP1 production are
neutral/antagonists of β-arrestin2 recruitment
and ERK1/2 phosphorylation. Although these
results suggest that these ligands are de facto
biased toward IP1 production over β-arrestin-2
recruitment and ERK1/2 phosphorylation, a bias
factor could not be calculated because no dose
response-curve could be obtained for β -arrestin2
and ERK1/2 (Table 3). Quantification of bias
could be done for JMV 1843 and MK-0677 only
and indicated that these ligands were not
significantly biased toward Gq activation and
IP1 production relative to β-arrestin2 recruitment
and ERK1/2 phosphorylation (Table 3).
Activation of G protein subtypes and isoforms
by GHS-R1a: a study with G protein
activation BRET biosensors.
To test GHS-R1a ligands on the activation of
different G protein subtypes besides Gq, we then
used a BRET
2
based assay that monitors
conformational changes of G proteins upon
activation (31,32). This BRET assay consisted in
measuring a BRET signal between Rluc8 fused
to the α subunit and GFP10 fused to the β2
subunit, in a Rluc8-α, β1, γ2-GFP10 complex
(see experimental procedures). When the three G
protein subunits were transfected in HEK293T
cells, a high BRET basal signal was detected due
to the close proximity of RLuc8 and GFP10 in
the inactive Gαβγ trimer. G-protein activation
then resulted in a large decrease of the BRET
signal due to conformational changes within the
G protein trimer with α subunit moving away
from the βγ complex (31).!!
Using this approach we revisited the GHS-R1a-
G protein coupling. As expected, GHS-R1a
stimulation with either ghrelin or MK-0677
agonists induced a large decrease of the BRET
signal with the Gq biosensor confirming that
GHS-R1a was indeed a typical Gq coupled
GPCR (Fig. 4). A decrease of the BRET signal
was also detected for Gi1, Gi2, Gi3, Goa, Gob
and G13 isoforms, confirming more directly than
in previous studies, that GHS-R1a can couple to
Gi and Go proteins. Importantly, a modest albeit
significant decrease of the BRET signal was
observed for G13, confirming that GHS-R1a
activates G13 also (Fig. 4A and B). In contrast,
no variation of the BRET signal was observed
for G12 and Gs. Because it was reported in the
literature that GHS-R1a activates Gs dependent
pathways in some cell systems, we questioned
whether the absence of Gs activation could result
from a default in the biosensor. To address this
point, we applied our BRET approach to the
detection of Gs coupling to a typical Gs-coupled
receptor control, the vasopressin V2 receptor. As
shown in Fig. 4C, a large decrease of the BRET
signal was observed on HEK293T cells co-
transfected with V2R and the Gs biosensor
following vasopressin stimulation. This
confirmed that the Gs biosensor was an
appropriate tool to detect Gs coupling and
therefore suggested that GHS-R1a was unable to
directly activate Gs, at least in HEK293T cells.
We also confirmed that the absence of G12
stimulation was not due to a non-functioning
G12 biosensor. Indeed, using the same
experimental conditions with HEK293T cells co-
expressing the thromboxane A2 α subtype (TPα)
receptor and the G12 sensor, stimulation with the
agonist U46619 triggered a decrease of BRET
signal was easily detected as already described
(32). This indicates that the G12 biosensor is
also functional, suggesting that GHS-R1a is not
coupled to G12 (Fig. 4D).
Selectivity of GHS-R1a ligands toward Gq
and IP1 production.
Although IP production was routinely used to
measure Gq/11 mediated signaling pathways, it
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
)!
was interesting to compare the behaviour of our
ligands in the Gq activation and IP1 production
assays to assess wether the efficiency of ligands
to activate Gq is transmitted all along the PLCβ-
inositol phosphate pathway. Comparison of
transduction efficiency (log(τ/K
A
)) at Gq
activation and IP1 production was only possible
for ghrelin, MK-0677 and JMV 1843. Indeed, no
robust IP1 production dose-response curves
could be obtained for JMV 2959, JMV 3002 and
JMV 3018. Ghrelin, MK-0677 and JMV 1843
displayed similar potency and efficacy for both
Gq activation and IP1 with MK-0677 displaying
a higher efficacy and being more potent than
JMV 1843 and ghrelin at both pathways (Fig. 5
and Table 2 for EC
50
and E
max
values).
Quantification of bias demonstrated no
selectivity of MK-0677 and JMV 1843 toward
Gq activation and IP1 production compared to
the reference ligand ghrelin (Table 3). As
indicated above, quantification of bias was not
possible for JMV 2959, JMV 3002, JMV 3011
and JMV 3018 (Table 3).
Selectivity of GHS-R1a ligands toward Gq, Gi
and Go activation.
We performed dose-response curves for Gi and
Go isoforms (Fig. 6). EC
50
values reported in
Table 2, showed that ghrelin was equally potent
to stimulate Gq, Gi and Go, whereas MK 0677!
and JMV 1843 were more potent to stimulate Gq
than Gi and Go. As shown in Fig. 6 and Table 2,
MK 0677 displayed a higher potency toward Gq
than toward Gi2, Gi3, Goa and Gob.
Quantification of bias confirms that, MK-0677 is
indeed slightly but significantly biased toward
Gq relative to Gi2, Gi3, Goa and Gob with a bias
factor (BF) of 6.46, 10.11, 12.59 and 18.84
respectively (Fig. 6, Table 3). We then
investigated the selectivity of GHS-R1a ligands
to promote activation of Gq, Gi2 and Gob, three
G protein subtypes for which the highest BRET
variation was observed following ghrelin
stimulation.
As already discussed above, ghrelin, MK-0677
and JMV 1843 behaved as full agonists toward
Gq, Gi2 and Gob activation (Fig 6A, B and C).
JMV 3011, which was classified as neutral and
antagonist based on the IP1 production assay
(Fig. 1 and 3), had indeed no action by its own
on Gq, Gi2 and Gob activation. However, as
expected, it antagonized ghrelin-induced Gq, Gi2
and Gob activation (Fig. 6 and 7). JMV 3011 is
thus neutral and antagonist at Gq, Gi2 and Gob.
In contrast, JMV 2959, JMV 3002, JMV 3018,
which behaved as partial agonists of Gq, were
neutral at Gi2 and Gob-activation (Fig. 6 and 7).
When we tested them at a 10
-6
M maximal dose
in competition with 10
-7
M ghrelin, these
compounds partially inhibited ghrelin-promoted
Gq activation and completely suppressed
ghrelin-evoked Gi2 and Gob activation (Fig. 7).
Taken together, these results suggest that JMV
2959, JMV 3011 and JMV 3018 are de facto
biased agonists toward Gq relative to Gi2 and
Gob. However, one can consider this as an
observational bias. Indeed, no quantification of
bias was possible because these ligands did not
induce any significant signals at Gi2a and Gob.
Selectivity of inverse agonists toward Gq and
G13 activation.
Interestingly, we found that the G-protein BRET
assay was also suitable for detection of inverse
agonists. Indeed, in contrast to agonists, SPA
induced an increase of the BRET signal on cells
co-expressing GHS-R1a and the Gq biosensor
(Fig. 8), confirming that SPA acts as an inverse
agonist toward GHS-R1a-promoted Gq
constitutive activity. To determine if using the
BRET assay could be a general way to identify
inverse agonists, we applied it to a panel of
compounds that had been classified as inverse
agonists based on IP1 production (Fig. 8A, Table
2). All these ligands promoted an increase of the
BRET signal in cells co-expressing GHS-R1a
and the Gq biosensor (Fig. 8B). This confirms
that SPA, KwFwLL-NH
2,
K-(D-1Nal)-FwLL-
NH
2
and JMV 4484 are inverse agonists toward
GHS-R1a-mediated Gq constitutive activity.
Interestingly, for different classes of GHS-R1a
ligands, a close correlation was obtained
between their efficacy to modulate Gq activity
and their efficacy to modulate IP1 production
(Fig 8C).
Beside constitutive activity at the Gq-Inostitol
phosphate signaling pathway, it had been
previously suggested, based on SRE reporter,
that GHS-R1a also constitutively activated
G12/G13-dependent pathways (25). Therefore,
we checked whether the G13 biosensor could be
used to bring a more direct proof of GHS-R1a-
mediated G13 constitutive activation. As was the
case for Gq, the basal level of G13 activity was
strictly dependent on the amount of GHS-R1a
receptors expressed at the surface of the cells.
Indeed, the BRET ratio decreased gradually
upon increasing the amount of GHS-R1a at the
cell surface (Fig. 9 A-F). These results
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
*!
confirmed that GHS-R1a constitutively activated
G13 and, as expected, the level of this
constitutive activity correlated to the receptor
expression level. Based on this result, we
investigated the behaviour of inverse agonists of
Gq on G13 activation (Fig. 9G and H).
Interestingly, compound K-(D-1Nal)-FwLL-NH
2
promoted an increase of the BRET signal at both
Gq and G13 whereas SPA, KwFwLL-NH
2
and
JMV 4484 behaved only inverse agonists on Gq
only (Fig. 9G and H). Although this suggest that
SPA, KwFwLL-NH
2
and JMV 4484 are biased
toward Gq over G13, it is difficult to firmly
conclude at this stage if the analysis. Indeed, the
BRET signal was 10 fold lower for G13 than for
Gq. The observational bias could thus result
from a difference in sensitivity between Gq and
G13 sensors. In order to confirm the results
obtained with the BRET sensors, we then used a
totally different assay. This assay consisted in
measuring GTPγS binding to purified Gq and
G13 proteins, in the presence of purified GHS-
R1a embedded in lipid discs. As expected, the
agonist MK-0677 promoted an increase of
GTPγS binding at both Gq and G13. However,
whereas K-(D-1Nal)-FwLL-NH
2
promoted a
decrease of GTPγS binding for both Gq and G13
compared to the unliganded receptor, SPA,
KwFwLL-NH
2
and JMV 4484 promoted a
decrease of GTPγS binding to Gq only (Fig. 10A
and B). These results were confirmed by kinetic
analyses of GTPγS binding (Fig. 10C and D).
Indeed, stimulation of GHS-R1a with K-(D-
1Nal)-FwLL-NH
2
promoted a decrease of the
Kact value for both Gq and G13 compared to
unliganded receptor,, whereas stimulation with
SPA, KwFwLL-NH
2
and JMV 4484 induced
decrease of the Kact value for Gq only (Fig. 10C
and D). Thus, these data confirmed that SPA,
KwFwLL-NH
2
and JMV 4484 compounds acted
as biased inverse agonists toward Gq relative to
G13. Although the raw fluorescence intensity
increase values upon GTP binding to Gq are two
fold higher than those obtained with G13, both
signal are nevertheless well above the signal to
noise ratio. This suggest that a difference in
sensitivity cannot explain the absence of change
in receptor-catalyzed GTPγS binding to G13
observed for SPA, KwFwLL-NH2 and JMV
4484. Unfortunately, no quantification of bias for
the inverse agonists could be done due to any
detectable effect of these ligands on G13.
!
DISCUSSION
In the present work we revisited the
pharmacological behaviour of ligands targeting
GHS-R1a by deeper exploring their efficacy
toward G protein dependent and independent
signaling pathways. We took a particular
attention to the selectivity of ligands toward a
panel of G-proteins subtypes thanks to G-protein
activation biosensors that were recently
developed (31,32). As already reported, GHS-
R1a displays one of the highest constitutive
activity (24,26) in the GPCR family (45,46). In
HEK293T cells, the model we used in this study,
the basal level of IP1 production represented 50-
70% of the maximal level promoted by ghrelin,
the endogenous ligand of GHS-R1a. In this
particular situation, detection of partial agonists
was problematic because the high basal level of
IP1 production might hide the partial agonist
character of some ligands. This was the case for
JMV 2959, firs reported as an antagonist (35,47)
but which displayed no or partial agonist activity
depending whether the basal activity was higher
or lower than 60 % of the maximal ghrelin
response. Indeed, we unambiguously
highlighted here the partial agonist character of
JMV 2959 and of other compounds (JMV 3002
and JMV 3018) by testing their efficacy on the
GHS-R1a-A204E mutant, which exhibits a low
constitutive activity (42). These results
highlighted the fact that ligands targeting GHS-
R1a might be classified as neutral or partial
agonists depending of the level of constitutive
activity of the receptor. This point is of
importance because constitutive activity of the
GHS-R1a was demonstrated in vivo in rat brain
(48) and in human somatotroph adenomas (49).
However, the exact level of constitutive activity
of the receptor is not ascertained due to the
difficulty to its in vivo quantification.
Furthermore, one can imagine that the level of
constitutive activity of the GHS-R1a varies with
its tissue or cellular localization. Indeed, it had
been repeatedly demonstrated that the level of
constitutive activity of a GPCR depends on the
cell content in various protein partners such as
other GPCRs or intracellular proteins (G
proteins, scaffolding proteins..) (45,46).
Therefore, the lack of knowledge on the ligand
independent activity of GHS-R1a in vivo should
make us cautious about classifying GHS-R1a
antagonists until a deep investigation has been
performed. Interestingly we found that JMV
compounds that behaved as partial agonists on IP
signaling were unable to promote both β-
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
"+!
arrestin2 recruitment and ERK1/2 activation, in
contrast to the full agonists ghrelin, MK-0677
and JMV1843. However, these partial agonists
totally inhibited ghrelin-promoted ERK1/2
phosphorylation and β-arrestin2 recruitment.
Thus, this suggests that JMV 2959, JMV 3018,
JMV 3002 are biased agonists toward IP
pathway compared to arrestin recruitment and
ERK1/2 activation and behave as biased
antagonists of arrestin recruitment and ERK1/2
activation compared to the IP pathway.
However, one can also consider this bias as an
observational bias. Indeed, no quantification of
bias was possible because these ligands did not
induce any detectable IP1 production, β-arrestin2
recruitment and ERK1/2 phosphorylation. We
took advantage here of G protein activation
BRET
biosensors (31,32) that directly report on
the conformational change of G protein upon
activation, to assess whether the partial agonist
behaviour of these ligands toward IP1 production
resulted from their Gq partial agonism. Although
it was reported that GHS-R1a activated G
protein dependent signalling pathways through
Gq, Gi, Go (23,30,50) and G13 (51), these
conclusions were! drawn from studies that
indirectly measured G protein activation. We
monitored here the selective coupling of GHS-
R1a to G protein family using activation
biosensors for a panel of G protein subtypes and
isoforms (Gq, Gi1, Gi2, Gi3, Goa, Gob, Gs, G12,
G13). As expected, GHS-R1a interaction with
either ghrelin, MK 0677 and JMV1843 resulted
in an efficient Gq activation. We also confirmed
that stimulation of GHS-R1a either with ghrelin
or MK-0677 promoted activation of Gi proteins
with a better efficacy for Gi2 and Gi3 than for
Gi1. We also found that ghrelin had the ability to
promote Goa, Gob and G13 activation, but was
unable to promote G12 and Gs activation. Thus,
our data demonstrate that ghrelin-stimulated
GHS-R1a activates Gq, Gi, Go and G13 proteins,
confirming that GHS-R1a signals through
intracellular pathways governed by these G
protein types. Although it was previously
reported that GHS-R1a activated G13 dependent
signaling pathways, these conclusions were
drawn from indirect measurement of SRE
(25,52) and Rho-kinase activation or by the use
of a dominant negative mutant of Gα13 (51).
Our data confirm more directly that ghrelin
stimulates GHS-R1a-mediated G13 activation. In
contrast, no Gs activation was detected,
confirming our previous data that concluded to
the inability of the purified GHS-R1a to couple
to Gs in lipid nanodiscs (26). Taken together our
data suggest that GHS-R1a does not interact with
Gs in contrast to published data indicating that!
ghrelin activated cAMP production in pancreatic
HIT-T25 beta cells (53,54). Although we found
that MK-0677 and JMV 1843 displayed similar
efficacy toward several G protein subtypes and
isoforms, quantification of bias demonstrate that
MK-0677 is slightly biased compared to ghrelin
toward Gq over Gi2, Gi3, Goa and Gob.
Interestingly, the neutral/antagonist character of
JMV 3011 demonstrated on IP1 production,
arrestin and ERK1/2 pathways was also
confirmed on Gq, Gi2 and Gob, making this
ligand a good lead for the design of GHS-R1a
signaling neutral/antagonist. More importantly,
the partial agonist behaviour of JMV 2959, JMV
3002 and JMV 3018 first assessed with the IP1
production assay was further confirmed with the
Gq biosensor. However, and interestingly,
ligands displaying partial agonist efficacy toward
Gq, were silent toward Gi2 and Gob. Moreover,
we clearly demonstrate that these ligands fully
inhibit Gi2 and Gob activation promoted by
ghrelin and MK-0677, whereas they only
partially inhibited Gq activation. Since, JMV
2959, JMV 3002 and JMV 3018 did not promote
any detectable activation of Gi2 and Gob, no
quantification of bias between Gq and Gi/o could
be obtained for these compounds. Nevertheless,
on can consider that up to 10
-6
M, JMV 2959,
JMV 3002 and JMV 3018 behaved as: (i) biased
agonists toward Gq activation and IP1
production relative to Gi2, Gob activation,
arrestin recruitment and ERK1/2 activation; (ii)
biased antagonists toward Gi2, Gob activation,
arrestin recruitment and ERK1/2 activation
relative to Gq activation and IP production. It is
obvious that these results need to be confirmed
in a more physiologically relevant natural system
than HEK293 cells. It also remains to assess
whether the ligand-directed functional selectivity
toward down-stream signaling pathways leads to
a functional selectivity toward physiological
functions controlled by the ghrelin/GHS-
R1aaxis. One can nevertheless mention that in
vivo studies in rat demonstrated that JMV 2959
inhibited food intake, addiction but not GH
secretion promoted by ghrelin (35,47). This
observation is of importance because it suggests
that a potential link may exist between the
selective action of JMV 2959 toward the
signaling pathways activated by GHS-R1a and
its selective antagonist action toward
physiological responses promoted by ghrelin
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
""!
(Fig. 11). Very interestingly, the synthetic ligand
GSK1614343 described as antagonist on calcium
release and IP1 production behaved in vivo as an
antagonist against GH secretion although it
stimulated food intake (55,56). In the same line,
another ghrelin analog, BIM-28163 considered
as a ghrelin receptor antagonist based on in vitro
calcium release assays, behaved in vivo as a
ghrelin-induced GH secretion antagonist, but as
an agonist on stimulation of food intake (57).
Therefore, GSK and BIM compounds behaved in
vivo in an opposite way of JMV 2959. Whether
the in vivo selective action of GSK and BIM
compounds results from a signaling bias opposite
to that of JMV 2959 remains to be determined.
Altogether, these observations suggest that it will
certainly be possible in a near future to design
new selective therapeutic drugs for pathologies
associated to ghrelin/GHS-R1a interactions. In
this context, it could be useful to selectively
block some of the signaling pathways linked to
constitutive activation of the GHS-R1a. Indeed,
GHS-R1a is one of the most constitutively active
GPCR and the potential role of its constitutive
activity in the “snacking” behaviour between
meals is questioned (58). So far, to our
knowledge, inverse agonists that display a
functional selectivity toward signaling pathways
have been only described for the ghrelin
receptor. Indeed it was reported that compound
KwFwLL-NH
2
, a GHS-R1a inverse agonist on
IP production was neutral in a SRE luciferase
assay, suggesting that this ligand is a biased
inverse agonist favouring inhibition of the
constitutive GHS-R1a-mediated Gq-dependent
pathway compared to that of the G13-dependent
pathway (52). Thanks to G-protein BRET
biosensors, we directly demonstrated that GHS-
R1a constitutively activates both Gq and G13.
We found that K-(D-1Nal)-FwLL-NH
2,
which
had been previously characterized as a GHS-R1a
inverse agonist on IP pathway (38), is indeed an
efficient and potent inverse agonist toward Gq
activity but behaves also as an inverse agonist
toward G13. We also found that SPA as well as
KwFwLL-NH
2
and JMV 4484 behave as inverse
agonists toward IP production and Gq activation
but were silent toward G13 activation. However,
it was difficult to interpret these data as resulting
from a real functional selectivity because the
intensity of G13 signal was much lower than that
of Gq. Nevertheless, these results were
confirmed by directly measuring receptor
catalysed GTPγS binding to Gq and G13 with
GHS-R1a reconstituted in lipid discs, an assay
for which Gq and G13 signals were comparable.
Thus, using two different approaches that
directly report on G protein activity, some
inverse agonists appear selective toward Gq over
G13. However, we could not confirm with these
assays that SPA behaves as a modest inverse
agonist at G13 signaling as previously suggested
from data that measured indirectly G13 activity
by recording SRE activity (25). A possibility
would be that this discrepancy results from the
different methods used in the two studies.
Indeed, the method we have employed in the
present study monitors the activity of the G
protein itself whereas the SRE reporter assay
monitors an activity resulting from the activation
of various G proteins, including Gq, G13, Gi and
Gβγ of Gi. Therefore, it may be possible that the
modest inverse agonist activity of SPA observed
in the SRE reporter assay did not result from the
G13 activation but rather from the activation of
other G protein-dependent pathways. In
summary, we have identified in this study, a
series of synthetic ligands that behave as partial
agonists at Gq but are silent toward β-arrestin2
recruitment and Gi/Go activation. One of them,
JMV 2959, appears particularly attractive
because this ligand selectively blocks ghrelin-
evoked food intake and addictions without
altering GH secretion. It should be now of
interest to further explore the in vivo behaviour
of the other molecules identified in this study
that display a bias behaviour similar to that of
JMV 2959. Finally, our data suggest that GHS-
R1a-dependent constitutive activation of Gq and
G13 can be selectively modulated by synthetic
ligands. It would be also important to test in
future studies whether the signaling bias
promoted by some of our ligands can result from
their action on allosteric binding sites and not
through their direct action on the orthosteric site,
an issue that has not been explored in this study.
Indeed, for a given GPCR, allosteric agonists can
promote different signaling profiles compared to
the orthosteric agonist, and there are several
examples of allosteric ligands that change the
coupling preference of the endogenous agonist
(59). Finally, although our study brings new
information on the selectivity of ligands at the
GHS-R1a signaling, the data reported in this
work were obtained in a single cell system
model, HEK293. This cell system is certainly far
from representing the physiological context of
GHS-R1a-dependent signaling. Furthermore, the
selectivity of action of ligands on various
signaling pathways varies depending on the
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
"#!
cellular context (60,61). Therefore, before
drawing any definitive conclusions on the
physiological reality of the signaling selectivity
of our ligands that was only observed so far in
HEK293, further studies should be carried out in
other heterologous cell systems, or even better in
primary cells that endogenously express the
GHS-R1a. It is obvious that the development of
functional selective drugs that could be
therapeutically useful will require further studies
to better understand the contribution of
individual signaling pathways to the diverse
physiological responses controlled by GHS-R1a.
AKNOWLEDGEMENTS
This work was supported by grants from the Institut National de la Santé et de la Recherche Médicale
(INSERM), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier, Agence
Nationale de la Recherche (PCV08-323163).
We thank Eric Trinquet (Cisbio) for providing us materials for HTRF-based assays.
CONFLICT OF INTEREST
The authors declare no conflict of interest.
AUTHOR CONTRIBUTIONS
C.M’K., J.P.L. and J. Marie designed research; C.M’K, J.P.L, L.O., C.G., A.S., D.G., M.D. and S.M.
performed research; M.M., S.D., P.V., J.A.F, J.M. synthetized ligands; C.M’K., J.L.B. and J. Marie
analyzed the data and J. Marie wrote the paper. All the authors reviewed the results and approved the
final version of the manuscript.
REFERENCES
",! -./0123!4,3!5.6.723!5,3!829:3!;,3!<2=2>29.3!4,3!4296?.3!5,3!2@7!-2@A2B23!-,!C"***D!
EFG:H0@! 06! 2 ! AG.B9FIF.G1.@:IG:H:260@A! 2JKH29:7! L:L907:! MG.1! 69.12JF,! Nature!
4023!'&'I''+!
#,! N=210>?3!O,3!2@7!-2@A2B23!-,!C#+"#D!OF:!LFK60.H.A0J2H!60A@0M0J2@J:!2@7!L.9:@902H!
JH0@0J2H!2LLH0J290.@6!.M!AFG:H0@,!European'journal'of'internal'medicine!233!"*(I#+#!
$,! 8:HL.G9:3!P,!C#+"$D!Q9G?J9?G:!2@7!LFK60.H.A0J2H!2J90.@6! .M!AFG:H0@,!Scientifica!20133!
&")*+*!
%,! R2A:G69G.13! 4,! P,3! 2@7! QJF0.9F3! 5,! S,! C#++)D! Q9G?J9?G2H! 70T:G609K! .M! E! LG.9:0@I
J.?LH:7! G:J:L9.G6! 2@7! 60A@0M0J2@J:! M.G! 7G?A! 706J.T:GK,! Nature' reviews.' Drug'
discovery!73!$$*I$&(!
&,! PG?>3! P,! U,3! 2@7! Q109F3! U,! E,! C#++)D! OF:! AG.B9F! F.G1.@:! 6:JG:92A.A?:! G:J:L9.G,!
Vitamins'and'hormones!773!%(I))!
',! P2HH2AF2@3! S,3! 2@7! V?G@:663! W,! S,! C#+"%D! <.T:H! 2@7! J.@T:@90.@2H! G:J:L9.G6! M.G!
AFG:H0@3! 7:62JKHIAFG:H0@3! 2@7! LF2G12J.H.A0J2HHK! G:H29:7! J.1L.?@76,!
Pharmacological'reviews!663!*)%I"++"!
(,! 4.?H0@3!N,3!UK2@3!W,3!42G90@:>3!W,3!2@7!V:FG:@9>3!W,!N,!C#++(D!U:J:@9!7:T:H.L1:@96!0@!
AFG:H0@!G:J:L9.G!H0A2@76,!ChemMedChem!23!"#%#I"#&*!
),! 5.H?X.T23! 4,3! QL.HJ.T23! N,3! 8:102@.T23! Y,3! QK=.G23! 8,3! V:FG:@9>3! W,! N,3! 42G90@:>3! W,3!
Q9.M=.T23!N,3!W?GJ.T0J.T23!W,3!8G2L2H.T23!W,3!R2J0@.T23!Y,3!52H?>0=3 !4,3!Y:H:>@23!S,3!2@7!
42H:90@6=23!R,!C#+"$D!EFG:H0@!2A.@069!W4Z!")%$!0@JG:26:6!M..7!0@92=:3!X.7K!B:0AF9!
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
"$!
2@7! :[LG:660.@! .M! .G:[0A:@0J! @:?G.L:L907:6! 0@! 10J:,! Physiological' research' /'
Academia'Scientiarum'Bohemoslovaca!623!%$&I%%%!
*,! PF.HH:93!P,3!4:K:G3!-,3!2@7!S:J=IQ0J=0@A:G3!N,!E,!C#++*D!EFG:H0@II2!@.T:H!A:@:G290.@!
.M!2@90I.X:609K!7G?A\!7:60A@3!LF2G12J.1.7?H290.@!2@7! X 0.H.A0J2H!2J90T09K!.M!AFG:H0@!
2@2H.A?:6,! Journal' of' peptide' science' :' an' official' publication' of' the' European'
Peptide'Society!153!(""I($+!
"+,! 52@66.@3!P,3!QF0G2>03!U,!5,3!<26H?@73!W,3!Z.A:H3!5,3!<:?X:G3!P,3!5.H13!E,3!N@J=2G629:G3!
5,3!80J=6.@3!Q, !R,3!]G0=66.@3!],3!2@7!Q=0X0J=23!-,!^,!C#+"#D!EFG:H0@!0@MH?:@J:6!@.T:H9K!
6::=0@A!X:F2T0.G!0@!G.7:@96!2@7!1:@,!PloS'one!73!:&+%+*!
"",! 4:@>0:63!W,!U,3!Q=0X0J=23!-,!^,3!R:@A3!E,3 !2@7!80J=6.@3!Q,!R,!C#+"$D!EFG:H0@3!G:B2G7!2@7!
1.90T290.@,!Endocrine'development!253!"+"I"""!
"#,! <2663! U,3! E2KH0@@3! S,! 8,3! 2@7! OF.G@:G3! 4,! _,! C#+""D! OF:! G.H:! .M! AFG:H0@! 0@! E5!
6:JG:90.@!2@7!E5!706.G7:G6,!Molecular'and'cellular'endocrinology!3403!"+I"%!
"$,! 4?GLFK3!4,!E,3!`:0663!Q,3!4JPH?@A3!4,3!QJF@09>:G3!O,3!P:GJF0.3!-,3!P.@@.G3!W,3!-G?L23!8,3!
E:G9>3! S,! W,3! 2 @7! EG.?L3! 4,! -,IN,! Q,! C#++"D! ]MM:J9! .M! 2H:@7G.@29:! 2@7! 4-I'((! C2!
AG.B9F! F.G1.@:! 6:JG:92A.A?:D3! 0@70T07?2HHK! 2@7! 0@! J.1X0@290.@3! .@! 12G=:G6! .M!
X.@:!9?G@.T:G!2@7!X.@:!10@:G2H!7:@609K!0@!L.691:@.L2?62H!.69:.L.G.90J!B.1:@,!
The'Journal'of'clinical'endocrinology'and'metabolism!863!"""'I""#&!
"%,! -:@2=0@3! O,! C#+""D! V?@J90.@2H! 6:H:J90T09K! 2@7! X026:7! G:J:L9.G! 60A@2H0@A,! The'
Journal'of'pharmacology'and'experimental'therapeutics!3363!#*'I$+#!
"&,! 42?76H:K3!Q,3!^29:H3!Q,!N,3!^2G=3!Q,!Q,3!R?99G:HH3!R,!4,3!2@7!42G90@3!S,!C#+"#D!V?@J90.@2H!
60A@2H0@A! X026:6! 0@! E! LG.9:0@IJ.?LH :7! G:J:L9.G6\! E21:! OF:.GK! 2@7! G:J:L9.G!
7K@210J6,!Mini'reviews'in'medicinal'chemistry!123!)$"I)%+!
"',! `F2H:@3! ],! W,3! U2/2A.L2H3! Q,3! 2@7! R:M=.B09>3! U,! W,! C#+""D! OF:G2L:?90J! L.9:@902H! .M!
X:92I2GG:690@I! 2@7! E! LG.9:0@IX026:7! 2A.@0696,! Trends' in' molecular' medicine! 173!
"#'I"$*!
"(,! -.X0H=23! S,! -,3! 2@7! 8:?L03! a,! C#++(D! P.@M.G1290.@2H! J.1LH:[09K! .M! EILG.9:0@I
J.?LH:7!G:J:L9.G6,!Trends'in'pharmacological'sciences!283!$*(I%+'!
"),! 42GK3!Q,3!82102@3!4,3!R.?:93! 4 ,3!VH.b?:93!<,3!V:FG:@9>3!W,!N,3!42G0:3!W,3!42G90@:>3!W,3!2@7!
S2@:G:63! W,! R,! C#+"#D! R0A2@76! 2@7! 60A@2H0@A! LG.9:0@6! A.T:G@! 9F:! J.@M.G1290.@2H!
H2@76J2L:! :[LH.G:7! XK! 2! E! LG.9:0@IJ.?LH:7! G:J:L9.G,! Proceedings' of'the'National'
Academy'of'Sciences'of'the'United'States'of'America!1093!)$+%I)$+*!
"*,! -2F6203! N,! `,3! a02.3! -,3! U2/2A.L2H 3! Q,3! NF@3! Q,3! QF?=H23! N,! -,3! Q?@3! W,3! _263! O,! E,3! 2@7!
R:M=.B09>3! U,! W,! C#+""D! 4?H90LH:! H0A2@7I6L:J0M0J! J.@M.G1290.@6! .M! 9F:! X:92#I
27G:@:GA0J!G:J:L9.G,!Nature'chemical'biology!73!'*#I(++!
#+,! ;2.3!a,!W,3!Z:H:>!U?0>3!E,3!`F.G9.@3!4,!U,3!U261?66:@3!Q ,!E,3!8:ZG::3!S,!O,3!8:?L03!a,3!
Q?@2F2G23! U,! -,3! 2@7! -.X0H=23! S,! C#++*D! OF:! :MM:J9! .M! H0A2@7! :MM0J2JK! .@! 9F:!
M.G1290.@!2@7!69 2X0H 09K! .M! 2! E^PUIE! LG.9:0@!J.1LH:[,!Proceedings'of'the'National'
Academy'of'Sciences'of'the'United'States'of'America!1063!*&+"I*&+'!
#",! 82102@3!4,3!42GK3!Q,3!420@A.93!4,3!4c-27103!P,3!E2A@:3!8,3!R:KG063!W,!^,3!8:@.K:HH:3!Q,3!
E20X:H:93!E,3!E2 T2 G2 3!R,3!E2GJ02!7:!Q.?>2!P.6923!4,3!^:G2F023!8,3!OG0@b?:93!],3!4.?0HH2J3!
S,3! E2H2 @7G0@3! Q,3! E2H:63! P,3! V:FG:@9>3! W,! N,3! VH.b?:93! <,3! 42G90@:>3! W,3! 42G0:3! W,3! 2@7!
S2@:G:63! W,! R,! C#+"&D! EFG:H0@! G:J:L9.G! J.@M.G1290.@2H! 7K@210J6! G:A?H29:! 9F:!
9G2@6090.@!MG.1! 2! LG:266:1XH:7! 9.! 2@! 2J90T:!G:J:L9.G\Eb!J.1LH:[,! Proceedings'of'
the'National'Academy'of'Sciences'of'the'United'States'of'America!1123!"'+"I"'+'!
##,! YF.?3! R, 3! 2@7! S.F@3! R,! 4,! C#+"%D! V?@J90.@2H! 6:H:J90T09K! .M! E^PU! 60A@2H0@A! 0@!
2@012H6,!Current'opinion'in'cell'biology!273!"+#I"+)!
#$,! ;0@3!;,3!R03!;,3!2@7!YF2@A3!`,!C#+"%D!OF:!AG.B9F!F.G1.@:!6:JG:92A.A?:!G:J:L9.G\!096!
0@9G2J:HH?H2G! 60A@2H0@A! 2@7! G:A?H290.@,! International' journal' of' molecular' sciences!
153!%)$(I%)&&!
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
"%!
#%,! 5.H693!S,3!PKA2@=0:B0J>3!N,3!W:@6:@3!O,!5,3!N@=:G6:@3!4,3!2@7!QJFB2G9>3!O,!`,!C#++$D!
50AF! J.@6909?90T:! 60A@2H0@A! .M! 9F:! AFG:H0@! G:J:L9.GII07:@90M0J290.@! .M! 2! L.9:@9!
0@T:G6:!2A.@069,!Molecular'endocrinology!173!##+"I##"+!
#&,! 5.H693! S,3! 5.HH072K3! <,! 8,3! S2JF3! N,3! ]HH0@A3! P,! ],3! P.[3! 5,! 4,3! 2@7! QJFB2G9>3! O,! `,!
C#++%D! P.11.@! 69G?J9?G2H! X2606! M.G! J.@6909?90T:! 2J90T09K! .M! 9F:! AFG:H0@! G:J:L9.G!
M210HK,!The'Journal'of'biological'chemistry!2793!&$)+'I&$)"(!
#',! 82102@3!4,3!42G0:3!W,3!R:KG063!W,!^,3!V:FG:@9>3!W,!N,3!Z:G70:3!^,3!42G90@:>3!W,3!S2@:G:63!W,!
R,3!2@7!42GK3!Q,!C#+"#D!50AF!J.@6909?90T:!2J90T09K!06!2@!0@9G0@60J!M:29?G:!.M!AFG:H0@!
G:J:L9.G! LG.9:0@\! 2! 69?7K! B09F! 2! M?@J90.@2H! 1.@.1:G0J! E5QIU"2! G:J:L9.G!
G:J.@6909?9:7!0@!H0L07!706J6,!The'Journal'of'biological'chemistry!2873!$'$+I$'%"!
#(,! 5.HH072K3!<,!8,3!5.H693! S,3!U.70.@.T23!],!N,3! QJFB2G9>3! O,!`,3!2@7!P.[3! 5,!4,!C#++(D!
d1L.G92@J:! .M! J.@6909?90T:! 2J90T09K! 2@7! 2GG:690@I0@7:L:@7:@9! 1:JF2@0616! M.G!
0@9G2J:HH?H2G!9G2MM0J=0@A!.M!9F:!AFG:H0@!G:J:L9.G,!Molecular'endocrinology!213!$"++I
$""#!
#),! P210@23!W,!^,3! R.7:0G.3!4,3!d6JF:@=.3!_,3!42G90@03!N,!P,3!2@7! P262@?:T23!V,!V,! C#++(D!
Q901?H290.@! XK! AFG:H0@! .M! L%#eL%%! 109.A:@I2J90T29:7! LG.9:0@! =0@26:!9FG.?AF!9F:!
E5QIU"2! G:J:L9.G\! G.H:! .M! EILG.9:0@6! 2@7! X:92I2GG:690@6,! Journal' of' cellular'
physiology!2133!")(I#++!
#*,! 4.?66:2?[3!8,3!R:!E2HH0J3!R,3!UK2@3!W,3!_0GK3!P,3!E2A@:3!8,3!V:FG:@9>3!W,!N,3!E2HH:KG2@73!W,!
P,3! 2@7! 42G9 0@:>3! W,! C#++'D! U:A?H290.@! .M! ]U-"e#! 2J90T09K! XK! AFG:H0@I2J90T29:7!
AG.B9F! F.G1.@:! 6:JG:92A.A?:! G:J:L9.G! "N! 0@T.HT:6! 2! ^RPe^-PT2G:L60H.@!
L29FB2K,!British'journal'of'pharmacology!1483!$&+I$'&!
$+,! ^2>.63!;,3!P262@?:T23!V,!V,3!2@7!P210@23!W,!^,!C#++)D!S260J!26L:J96!.M!AFG:H0@!2J90.@,!
Vitamins'and'hormones!773!)*I""*!
$",! E2H:63!P,3!Z2@!8?G13!W,!W,3!QJF22=3!Q,3!^.@90:G3!Q,3!^:GJF:G2@J0:G3!;,3!N?7:93!4,3!^2G063!
5,3! 2@7! S.?T0:G3! 4,! C#++'D! ^G.X0@A ! 9F:! 2J90T290.@ILG.1.9:7! 69G?J9?G2H!
G:2GG2@A:1:@96!0@!LG:266:1XH:7!G:J:L9.GIE!LG.9:0@!J.1LH:[:6,!Nature'structural'
&'molecular'biology!133!(()I()'!
$#,! Q2?H0:G:3! N,3! S:HH.93! 4,3! ^2G063! 5,3! 8:@063! P,3! V0@2@23! V,3! 52@6:@3! W,! O,3! NH90:3! 4,! V,3!
Q:A?:H263! 4,! 5,3! ^29F2=3! N,3! 52@6:@3! W,! R,3! Q:@2G73! W,! 4,3! 2@7! E2H:63! P,! C#+"#D!
8:J0LF:G0@A!X026:7I2A.@061!J.1LH:[09K!G:T:2H6!2!@:B!2J90T:!NO"!G:J:L9.G!:@909K,!
Nature'chemical'biology!83!'##I'$+!
$$,! ^29JF:993!N,!N,3!<2GA?@73!U,!^,3!O2923!W,!U,3!PF:@3!4,!5,3!S2G2=293!-,!W,3!W.F@69.@3!8,!S,3!
PF:@A3!-,3!PF2@3!`,!`,3!S?9H:G3!S,3!50J=:K3!E,3!2@7!:9!2H,!C"**&D!8:60A@!2@7!X0.H.A0J2H!
2J90T090:6! .M! RI"'$3"*"! C4-I+'((D\! 2! L.9:@93! .G2HHK! 2J90T:! AG.B9F! F.G1.@:!
6:JG:92A.A?:,!Proceedings'of'the'National'Academy'of'Sciences'of'the'United'States'
of'America!923!(++"I(++&!
$%,! E?:GH2T2063!Z,3!S.:AH0@3!8,3!4.?66:2?[3!8,3!_0GK3!P,3! 5:09>3!N,3!8:AF:@AF03!U,3!R.J29:HH03!
Z,3! O.G6:HH.3! N,3! EF:3! P,3! P292L2@.3! V,3! 4?JJ0.H03! E,3! E2HH:KG2@73! W,! P,3! V:FG:@9>3! W,! N,3!
2@7! 42G90@:>3! W,! C#++$D! <:B! 2J90T:! 6:G0:6! .M! AG.B9F! F.G1.@:! 6:JG:92A.A?:6,!
Journal'of'medicinal'chemistry!463!""*"I"#+$!
$&,! 4.?H0@3! N,3! 8:12@A:3! R,3! S:GA:3! E,3! E2A@:3! 8,3! UK2@3! W,3! 4.?66:2?[3! 8,3! 5:09>3! N,3!
^:GG066.?73! 8,3! R.J29:HH03! Z,3! O.G6:HH.3! N,3! E2HH:KG2@73! W,! P,3! V:FG:@9>3! W,! N,3! 2@7!
42G90@:>3!W,!C#++(D!O.B2G7!L.9:@9!AFG:H0@!G:J:L9.G!H0A2@76!X26:7!.@!9G06?X6909?9:7!
"3#3%I9G02>.H:! 69G?J9?G:,! #,! QK@9F:606! 2@7! LF2G12J.H.A0J2H! 0@! T09G.! 2@7! 0@! T0T.!
:T2H?290.@6,!Journal'of'medicinal'chemistry!503!&(*+I&)+'!
$',! 4.?H0@3!N,3! 8:12@A:3! R,3! UK2@3! W,3! 4.?66:2?[3! 8,3! Q2@JF:>3! ^,3! S:GA:3! E,3! E2A@:3! 8,3!
^:GG066.?73! 8,3! R.J29:HH03! Z,3! O.G6:HH.3! N,3! E2HH:KG2@73! W,! P,3! V:FG:@9>3! W,! N,3! 2@7!
42G90@:>3!W,!C#++)D!<:B!9G06?X6909?9:7!"3#3%I9G02>.H:!7:G0T290T:6!26!L.9:@9!AFG:H0@!
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
"&!
G:J:L9.G! 2@92A.@0696,! $,! QK@9F:606! 2@7! LF2G12J.H.A0J2H! 0@! T09G.! 2@7! 0@! T0T.!
:T2H?290.@6,!Journal'of'medicinal'chemistry!513!')*I'*$!
$(,! 5.H693!S,3!R2@A3!4,3!SG2@793!],3!S2JF3!N,3!5.B2G73!N,3!VG01?G:G3!O,!4,3!S:J=IQ0J=0@A:G3!
N,3!2@7!QJFB2G9>3!O,!`,!C#++'D!EFG:H0@!G:J:L9.G!0@T:G6:!2A.@0696\!07:@90M0J290.@!.M!
2@! 2J90T:! L:L907:! J.G:! 2@7! 096! 0@9:G2J90.@! :L09.L:6! .@! 9F:! G:J:L9.G,! Molecular'
pharmacology!703!*$'I*%'!
$),! ]H63! Q,3! QJF0H73! ],3! ^:9:G6:@3! ^,! Q,3! -0H02@3! O,! 4,3! 4.=G.60@6=03! W,3! VG01?G:G3! O,! 4,3!
PF.HH:93!P,3!QJFB2G9>3!O,!`,3!5.H693!S,3!2@7!S:J=IQ0J=0@A:G3!N,!E,!C#+"#D!N@!2G.1290J!
G:A0.@! 9.! 0@7?J:! 2! 6B09JF! X:9B::@! 2A.@061! 2@7! 0@T:G6:! 2A.@061! 29! 9F:! AFG:H0@!
G:J:L9.G,!Journal'of'medicinal'chemistry!553!(%$(I(%%*!
$*,! R:KG063!W,!^,3!U.?[3!O,3!OG0@b?:93!],3!Z:G70:3!^,3!V:FG:@9>3!W,!N,3!_?:6H2903!<,3!8.?>.@3!Q,3!
S.?GG0:G3! ],3! R212Gb?:3! R,3! E2A@:3! 8,3! E2HH:KG2@73! W,! P,3! 4c-27103! P,3! 42G90@:>3! W,3!
42GK3! Q,3! S2@:G:63! W,! R,3! 2@7! 42G0:3! W,! C#+""D! 5.1.A:@:.?6! 901:IG:6.HT:7!
MH?.G:6J:@J:IX26:7! 2662K! 9.! 6JG::@! M.G! H0A2@76! 92GA:90@A! 9F:! AG.B9F! F.G1.@:!
6:JG:92A.A?:!G:J:L9.G!9KL:!"2,!Analytical'biochemistry!4083!#&$I#'#!
%+,! -:@2=0@3!O,3!`296.@3!P,3!4?@0>I4:70@23!Z,3!PFG069.L.?H.63!N,3!2@7!<.T0J=3!Q,!C#+"#D!
N! 601LH:! 1:9 F.7! M.G! b?2@90MK0@A! M?@J90.@2H! 6:H:J90T09K! 2@7! 2A.@069! X026,! ACS'
chemical'neuroscience!33!"*$I#+$!
%",! SH2J=3!W,! `,3! 2@7! R:MM3! ^,! C"*)$D! _L:G290.@2H! 1.7:H6! .M! LF2G12J.H.A0J2H! 2A.@061,!
Proceedings'of' the'Royal'Society' of'London.'Series'B,'Biological'sciences! 2203! "%"I
"'#!
%#,! ^2@9:H3! W,3! R:A:@7G:3! 4,3! P2XG.H3! Q,3! 50H2H3! R,3! 52/2/03! ;,3! 4.G066:93! Q,3! <0T.93! Q,3! Z0:I
R?9.@3!4,!^,3!EG.?6:HH:3!8,3!7:!-:G72@:93!4,3!-270G03!N,3!] L:HX2 ?13!W,3!R:!S.?J3!;,3!2@7!
N16:H:13! Q,! C#++'D! R.66! .M! J.@6909?90T:! 2J90T09K! .M! 9F:! AG.B9F! F.G1.@:!
6:JG:92A.A?:!G:J:L9.G!0@!M210H02H!6F.G9!6929?G:,!The'Journal'of'clinical'investigation!
1163!('+I(')!
%$,! ]TG.@3!O,3!^:9:G6.@3!Q,!4,3!fG63!<,!4,3!S203!;,3!U.JF:HH:3!R,!-,3!P2G.@3!4,!E,3!2@7!S2G2=3!
R,!Q,!C#+"%D!E!^G.9:0@!2@7!X:92I2GG:690@!60A@2H0@A!X026!29!9F:!AFG:H0@!G:J:L9.G,!The'
Journal'of'biological'chemistry!2893!$$%%#I$$%&&!
%%,! Q0T:G96:@3!S,3!5.HH072K3!<,3!4276:@3!N,!<,3!2@7!5.H693!S,!C#+"$D!V?@J90.@2HH K!X026:7!
60A@2HH0@A! L G.L:G90:6! .M! (O4! G:J:L9.G6! I! .LL.G9?@090:6! M.G! 7G?A! 7:T:H.L1:@9! M.G!
9F:!AFG:H0@!G:J:L9.G,!British'journal'of'pharmacology!1703!"$%*I"$'#!
%&,! 40HH0A2@3!E,!C#++$D!P.@6909?90T:!2J90T09K!2@7!0@T:G6:!2A.@0696!.M!E!LG.9:0@IJ.?LH:7!
G:J:L9.G6\!2!J?GG:@9!L:G6L:J90T:,!Molecular'pharmacology!643!"#("I"#('!
%',! <:H6.@3!P,!^,3!2@7!PF2HH0663!U,!N,!C#++(D!g^F:@.9KL0Jg!LF2G12J.H.AK\!9F:!0@MH?:@J:!
.M! J:HH?H2G! :@T0G.@1:@9! .@! E! LG.9:0@IJ.?LH:7! G:J:L9.G! 2@92A.@069! 2@7! 0@T:G6:!
2A.@069!LF2G12J.H.AK,!Biochemical'pharmacology!733!($(I(&"!
%(,! 4.?H0@3!N,3!SG?@:H3!R,3!S.:AH0@3!8,3!8:12@A:3!R,3!UK2@3!W,3!4c-27103!P,3!8:@.K:HH:3!Q,3!
42G90@:>3!W,3!2@7!V:FG:@9>3!W,!N,!C#+"$D!OF:!"3#3%I9G02>.H:!26!2!6J2MM.H7!M.G!9F:!7:60A@!
.M!AFG:H0@!G:J:L9.G!H0A2@76\!7:T:H.L1:@9!.M!W4Z!#*&*3!2!L.9:@9!2@92A.@069,!Amino'
acids!443!$+"I$"%!
%),! ^:9:G6:@3! ^,! Q,3! `.H7XK:3! 8,! ^,3! 4276:@3! N,! <,3! ]A:G.73! -,! R,3! W0@3! P,3! R2@A3! 4,3!
U261?66:@3!4,3!S:J=IQ0J=0@A:G3!N,!E,3!2@7!5.H693!S,!C#++*D!d@!T0T.!JF2G2J9:G0>290.@!
.M!F0AF!S262H!60A@2H0@A!MG.1!9F:!AFG:H0@!G:J:L9.G,!Endocrinology!1503!%*#+I%*$+!
%*,! 4:2G3! ;,3! SH2@JF2G73! 4,! ^,3! 8:M0HH:63! P,3! SG?:3! O,3! V0A2G:HH2ISG2@A:G3! 8,3! EG20HH.@3! O,3!
42@2T:H23! 4,3! S2GH0:G3! N,3! ]@/2HX:G93! N,3! 2@7! OF0G0.@3! Q,! C#+"%D! EFG:H0@! G:J:L9.G!
CE5QIU"2D! 2@7! 096! J.@6909?90T:! 2J90T09K! 0@! 6.129.9G.LF! 27:@.126\! 2! @:B! J.I
92GA:90@A!9F:G2LK!?60@A! E5QIU"2! 0@T:G6:! 2A.@0696! 2@7!6.129.69290@! 2@2H.A6,! The'
Journal'of'clinical'endocrinology'and'metabolism!993!]#%'$I#%("!
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
"'!
&+,! S:@@:993!-,!N,3!R2@A1:273!P,! W,3!`06:3!N,3!2@7!40HH0A2@3!E,!C#++*D!EG.B9F! F.G1.@:!
6:JG:92A.A?:6! 2@7! AG.B9F! F.G1.@:! G:H:260@A! L:L907:6! 2J9! 26! .G9F.69:G0J! 6?L:GI
2A.@0696!X?9!@.9!2HH.69:G0J!G:A?H29.G6!M.G!2J90T290.@!.M!9F:!E!LG.9:0@!E2H LF2 C."D!XK!
9F:!EFG:H0@!G:J:L9.G,!Molecular'pharmacology!763!)+#I)""!
&",! Q0T:G96:@3!S,3!R2@A3!4,3!VG01?G:G3!O,!4,3!5.HH072K3!<,!8,3!S2JF3!N,3!]H63!Q,3!]@A:H69.M93!4,!
Q,3!^:9:G6:@3!^,!Q,3!4276:@3!N,! <,3!QJFB2G9>3!O,!`,3!S:J=IQ0J=0@A:G3!N,!E,3!2@7!5.H693!S,!
C#+""D! f@0b?:! 0@9:G2J90.@! L299:G@! M.G! 2! M?@J90.@2HHK! X026:7! AFG:H0@! G:J:L9.G!
2A.@069,!The'Journal'of'biological'chemistry!2863!#+)%&I#+)'+!
&#,! 5.H693!S,3! 4.=G.60@6=03! W,3! R2@A3! 4,3! SG2@793! ],3! <KA22G73! U,3! VG01?G:G3! O,! 4,3! S:J=I
Q0J=0@A:G3! N,! E,3! 2@7! QJFB2G9>3! O,! `,! C#++(D! d7:@90M0J290.@! .M! 2@! :MM0J2JK! 6B09JF!
G:A0.@!0@! 9F:! AFG:H0@! G:J:L9.G! G:6L.@60XH:! M.G!0@9:GJF2@A:!X:9B::@! 2A.@061! 2@7!
0@T:G6:!2A.@061,!The'Journal'of'biological'chemistry!2823!"&(**I"&)""!
&$,! Q9.G=3! ^,! W,3! 2@7! QJF1099 3! W,! 4,! C#++#D! PG.6692H=! X:9B::@! JN4^! 2@7! 4N^! =0@26:!
60A@2H0@A!0@!9F:!G:A?H290.@!.M!J:HH!LG.H0M:G290.@,!Trends'in'cell'biology!123!#&)I#''!
&%,! EG2@2923!U,3!Q:992@@03!V,3!S02@J.@:3!R,3!OG.T29.3!R,3!<2@.3!U,3!S:G9?>>03!V,3!8:69:M2@063!
Q,3!N@@?@>02923!4,3!42G90@:9903!4,3!P292L2@.3!V,3!EF:3!P,3!d6A22G73!W,3!^2L.9903!4,3!EF0A.3!
],3!2@7!4?JJ0.H03!E,!C#++(D!NJKH29:7!2@7!?@2JKH29:7!AFG:H0@!LG.1.9:!LG.H0M:G290.@!
2@7! 0@F0X09! 2L.L9.606! .M! L2@JG:290J! X:92IJ:HH6! 2@7! F?12@! 06H:96\! 0@T.HT:1:@9! .M!
$c3&cIJKJH0J! 27:@.60@:! 1.@.LF.6LF29:eLG.9:0@! = 0@26:! N3! :[9G2J:HH?H2G! 60A@2HI
G:A?H29:7! =0@26:! "e#3! 2@7! LF.6LF2907KH! 0@.609.H! $I-0@26:eN=9! 60A@2H0@A,!
Endocrinology!1483!&"#I&#*!
&&,! P.692@90@03! Z,! W,3! Z0J:@ 90@03! ],3! Q2XX290@03! V,! 4,3! Z2H:G0.3! ],3! R:L.G:3! Q,3! O:662G03! 4,3!
Q2G9.G03! 4, 3! 40JF0:H0@3! V,3! 4:H.99.3! Q,3! S0M.@:3! N,3! ^0JF3! ],! 4,3! 2@7! P.G603! 4,! C#+""D!
EQ-"'"%$%$3! 2! @.T:H! AFG:H0@! G:J:L9.G! 2@92A.@0693! LG.7?J:6! 2@! ?@:[L:J9:7!
0@JG:26:!.M!M..7!0@92=:!2@7!X.7K!B:0AF9!0@!G.7:@96!2@7!7.A6,!Neuroendocrinology!
943!"&)I"')!
&',! Q2XX290@03! V,! 4,3! 80! V2X0.3! U,3! P.G603! 4,3! P2T2@@03! ^,3! SG.107A:3! Q,! 4,3! Q9I8:@063! ;,3!
8cN721.3! R,3! P.@90@03! Q,3! U0@2H703! 4,3! E?:GK3! Q,3! Q2T.023! P,3! 4?@703! P,3! ^:G0@03! S,3!
P2GL:@9:G3!N,!W,3!82H!V.G@.3!E,3!V2AA0.@03!V,3!O:662G03!4,3!^2T.@:3!V,3!80!VG2@J:6J.3!P,3!
S?6.@3!N,3! 42990.H03!4,3!^:G7.@23! ],3!2@7!4:H.99 .3! Q,!C#+"+D!806J.T:GK! LG.J:66!2@7!
JF2G2J9:G0>290.@! .M! @.T:H! J2GX.FK7G2>07:! 7:G0T290T:6! 26! L.9:@9! 2@7! 6:H:J90T:!
E5QU"2!2@92A.@0696,!ChemMedChem!53!"%&+I"%&&!
&(,! 52H:13! 5,! N,3! O2KH.G3! W,! ],3! 8.@A3! W,! Y,3! QF:@3! ;,3! 829923! U,3! NX0>2073! N,3! 802@.3! Q,3!
5.GT29F3! O,3! Y0>>2G03! ^,3! SH?:9I^2/.93! 4,! O,3! ]L:HX2?13! W,3! 2@7! P?HH:G3! 4,! 8,! C#++%D!
<.T:H!2@2H.A6!.M!AFG:H0@\!LFK60.H.A0J2H!2@ 7!JH0@0J2H!01LH0J290.@6,!European'journal'
of'endocrinology'/'European'Federation'of'Endocrine'Societies!151!Suppl!13!Q("I(&!
&),! 5.H693! S,3! 2@7! QJFB2G9>3! O,! `,! C#++%D! P.@6909?90T:! AFG:H0@! G:J:L9.G! 2J90T09K! 26! 2!
60A@2H0@A! 6:9IL.0@9! 0@! 2LL:909:! G:A?H290.@,! Trends'in'pharmacological'sciences! 253!
""$I""(!
&*,! -:@2=0@3! O,3! 2@7! PFG069.L.?H.63! N,! C#+"$D! Q0A@2HH0@A! X026! 0@! @:B! 7G?A! 706J.T:GK\!
7:9:J90.@3! b?2@90M0J290.@! 2@7! 9F:G2L:?90J! 01L2J9,! Nature' r eviews.' Drug' discovery!
123!#+&I#"'!
'+,! -:@2=0@3! O,! C#++(D! V?@J90.@2H! 6:H:J90T09K! 9FG.?AF! LG.9:2@! 2@7! X026:7! 2A.@061\!
BF.!69::G6!9F:!6F0Lh!Molecular'pharmacology!723!"$*$I"%+"!
'",! R?99G:HH3! R,! 4,3! 42?76H:K3! Q ,3! 2@7! S.F@3! R,! 4,! C#+"&D! V?HM0HH0@A! 9F:! ^G.106:! .M!
gS026:7g!E!^G.9:0@IP.?LH:7!U:J:L9.G! NA.@061,! Molecular'pharmacology!883!&(*I
&))!
!
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
"(!
FOOTNOTES
The abbreviations used are: GHS-R1a, growth hormone secretagogue receptor type 1a; GH, growth
hormone, GPCR, G protein-coupled receptor; PBS, phosphate-buffered saline; IP1, inositol-1-
phosphate; HEK, human embryonic kidney; SRE, serum-responsive element; ERK, extracellular signal-
regulated kinase, BRET, bioluminescence resonance energy transfer; HTRF, homogenous time resolved
fluorescence; GTPγS, guanosine 5’-O-(3-thiotriphosphate); PKC, protein kinase C; PI3K,
phosphatidylinositol 3 kinase; YFP, yellow fluorescent protein; Luc, luciferase.!
FIGURE LEGENDS
Figure 1. Efficacy of GHS-R1a ligands to promote inositol phosphate production. Inositol
phosphate (IP1) production promoted by ligands at maximal dose (10
-6
M) in cells expressing the GHS-
R1a (A) or the A204E GHS-R1a mutant (D). Basal level of IP1 production measured in cells expressing
different amounts of GHS-R1a (B). IP1 production promoted by ligands at maximal dose (10
-6
M), in
cells expressing low and high amount of GHS-R1a (C). IP1 production was measured with an HTRF
assay in HEK293T cells treated with ligands for 30 min at 37°C. Data are expressed as the % of ghrelin
maximal response. Basal represents IP1 production measured in non-stimulated HEK293T cells
expressing GHS-R1a receptors. 0% is defined as the basal of IP1 production of mock-transfected
HEK293T cells (cells transfected with an empty pcDNA3.1 (+) vector). Values are mean ± S.E.M of
three independent experiments performed in triplicate. Statistical significance between stimulated and
non-stimulated cells was assessed using a one-way ANOVA followed by Dunnett’s post-hoc test
(***P<0.001, **P<0.01, *P<0.1).
Figure 2. Efficacy of GHS-R1a ligands to promote β-arrestin2 recruitment and ERK1/2
phosphorylation.
β-arrestin2 recruitment dose-response curves (A) and maximal responses with 10
-6
M of ligand (C). β-
arrestin2 recruitment to GHS-R1a was measured with a BRET
1
assay upon ligand stimulation for 45
min at 37°C in HEK293T cells expressing the GHS-R1a. ERK1/2 phosphorylation dose-response
curves (B) and maximal responses with 10
-6
M of ligand (D). ERK1/2 phosphorylation was measured
with an HTRF assay upon ligand stimulation for 10 min at 37°C in HEK293T cells expressing the
GHS-R1a. All data are expressed as the % of maximal ghrelin-induced stimulation. Dose response
curves (A, B) are representative of three experiments and graphs of maximal responses (C, D). Values
are mean ± SEM of three independent experiments performed in triplicate. 0% represents the basal of
mock-transfected HEK293T cells. Statistical significance between stimulated and non-stimulated cells
was assessed using a one-way ANOVA followed by Dunnett’s post-hoc test (***P<0.001, **P<0.01,
*P<0.1).
Figure 3. Antagonist efficacy of JMV compounds toward ghrelin-promoted IP1 production, β -
arrestin2 recruitment and ERK1/2 phosphorylation. IP1 production (A), β-arrestin2 recruitment (B)
and ERK1/2 phosphorylation (C) measured as described in Figure 1 and 2 and in the experimental
procedures section. HEK293T cells expressing the GHS-R1a were stimulated with ghrelin at 10
-8
M in
the absence or in the presence of 10
-6
M JMV compounds. Data are expressed as the % of maximal
ghrelin-induced stimulation. Bars and error bars represent the mean ± S.E.M of three independent
experiments, each performed in triplicate. Statistical significance between stimulated and non-
stimulated cells was assessed using a one way ANOVA followed by Dunnett’s post-hoc test
(***P<0.001,**P< 0.01, *P<0.1).
Figure 4. Activation of G proteins subtypes and isoforms by GHS-R1a. A, G protein activation
kinetics was measured by BRET
2
using G protein activation biosensors as described in experimental
procedures section. HEK293T cells co-expressing both the GHS-R1a and the G protein biosensor were
stimulated by the GHS-R1a agonist MK-0677 (10
-6
M). Data are representative of three to eight
independent experiments. B, BRET maximal signal promoted by 10
-6
M MK-0677 on HEK293T cells
co-expressing GHS-R1a and G protein biosensors. Results are expressed as the difference in BRET
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
")!
ratio measured in the presence and in the absence of ligand stimulation for each G protein type (mean ±
S.E.M). Statistical significance between stimulated and non-stimulated cells was assessed using a paired
Student’s t-test (**P<0.01, *P<0.05). C, Gs activation by the vasopressin V2 receptor: HEK293T cells
co-expressing the V2 vasopressin receptor and Gs biosensor, were stimulated by 10
-6
M Arg-vasopressin
and the BRET signal was recorded as described in panel A. D, G12 activation by the thromboxane
A2 α type (TPα) receptor: HEK293T cells co-expressing the TPα receptor and G12 biosensor were
stimulated by 10
-6
M U46619 and the BRET signal was recorded at described in panel A.
Figure 5. Efficacy and potency of ghrelin, MK-0677 and JMV 1843 toward Gq activation and
inositol phosphate production (IP1). A, dose-dependent Gq activation measured by BRET
2
in
HEK293T cells co-expressing both the GHS-R1a and Gq biosensor, B, dose-dependent inositol
phosphate production (IP1) measured by HTRF in HEK293T cells expressing the GHS-R1a. The zero
value corresponds to the basal of non-stimulated HEK293T cells expressing the GHS-R1a. Curves are
representative of three independent experiments, each performed in triplicate. Values of EC
50
and E
max
are reported in table 2.
Figure 6. Efficacy and potency of GHS-R1a ligands to trigger activation of Gq, Gi and Go. The
BRET signal was recorded as a function of increasing concentrations of ligands in HEK293T cells co-
expressing GHS-R1a and the G protein sensors. The effect of JMV 2959, JMV 3002, JMV 3018 and
JMV 3011 was tested only on Gq, Gi2 and Gob. Data are representative of three independent
experiments each performed in triplicate. Values of EC
50
and E
max
are reported in Table 2.
Figure 7. Efficacy of JMV 2959, JMV 3002, JMV, 3018 and JMV 3011 at inhibiting ghrelin-
promoted Gq, Gi2 and Gob activation. HEK293T cells expressing GHS-R1a were stimulated for 15
min at 25°C with 10
-6
M of JMV compounds in the presence or absence of ghrelin at 10
-7
M. Results are
expressed as the difference in BRET ratio measured in the presence and in the absence of ligand
stimulation for each G-protein type. Values are mean ± S.E.M of three experiments, each performed in
triplicate. Statistical significance between the signal obtained with ghrelin alone and ghrelin in the
presence of JMV compounds for each G protein was assessed using a paired Student’s t-test
(***P<0.001, **P<0.01, *P<0.05).
Figure 8: Efficacy of inverse agonists toward Gq and IP production. A, Efficacy of ligands (10
-6
M)
at promoting inositol phosphate production (IP1) expressed as the % of basal IP of HEK293T cells
expressing GHS-R1a, where zero represents the basal IP1 production of mock-transfected HEK293T
cells. B, Efficacy of ligands (10
-6
M) at promoting BRET signal increase in HEK293T cells co-
expressing the GHS-R1a and the Gq biosensor. The basal value represents the BRET signal obtained in
the absence of ligand stimulation. Values are mean ± SEM of three experiments, each performed in
triplicate. Statistical significance between stimulated and non-stimulated cells was assessed using a
paired Student’s t-test (***P<0.001, **P<0.01, *P<0.05). C, Correlation between the efficacy of
ligands toward Gq activation and their efficacy toward inositol phosphate production. Variation of the
BRET
2
signal triggered by ligands in HEK293T cells co-expressing GHS-R1a and the Gq sensor are
plotted versus IP1 production promoted by ligands in HEK293T cells expressing GHS-R1a. BRET
2
signal variation is expressed as the difference of the BRET ratio measured in simulated and non-
stimulated cells. IP production is expressed as the % of basal IP production measured in HEK293T
cells expressing the GHS-R1a with basal representing 100%. Values are mean ± SEM of three
experiments. R
2
= 0.94
Figure 9. GHS-R1a-dependent constitutive activity at Gq and G13 and selectivity of agonists and
inverse agonists. A and B: BRET signal measured in HEK293T cells co-expressing either Gαq-Rluc8
(A) or Gα13-Rluc8 (B), GFP10-Gγ2 and Gβ1 in the absence or presence of increasing amounts of HA-
GHS-R1a (vectors encoding N-terminally the HA-tagged GHS-R1a ranging from 0.001 to 4 µg/well)
and in the absence of ligand. Data represent the mean ± S.E.M. of at least three independent
experiments. Statistical significance between cells expressing or not the HA-GHS-R1a was assessed
using a one-way ANOVA followed by a Tukey’s test (*P < 0.05; **P < 0.01, ***P < 0.001).
Each transfection condition was controlled for Gαq-Rluc8 (C) and Gαq-Rluc8 (D) total expression
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
"*!
levels by measuring luminescence intensity (C and D) and cell surface expression of HA-GHS-R1a
quantified by ELISA using an anti-HA antibody (E and F). Results are expressed as the mean ± S.E.M
of at least three independent experiments.
G and H: BRET signal promoted by ligands measured in HEK293T cells co-expressing either Gαq-
Rluc8 (G) or Gα13-Rluc8 (H), GFP10-Gγ2 and Gβ1, in the presence of the HA-GHS-R1a, and
stimulated or not with 10 µM ligands. Results are expressed as the difference in BRET signals
measured in the presence and in the absence of ligand. Values are mean ± S.E.M. of at least four
independent experiments. Statistical significance between stimulated and non-stimulated cells was
assessed using a paired Student’s t-test (***P<0.001, **P < 0.01, *P < 0.05).
Figure 10. GHS-R1a dependent constitutive activity at Gq and G13 proteins and ligand selectivity
with the purified receptor.
The monomeric GHS-R1a in lipid discs was incubated with purified Gαq and Gα13 in the presence of
Gβ 1γ2. The efficacy of ligands to modulate GHS-R1a-promoted Gq (A) and G13 (B) activity was
assessed by monitoring changes in the BODIPY FL GTPγS emission intensity. GTPγS binding is
expressed as raw values of fluorescence emission of bodipyGTPγS . Data are from one representative of
three independent experiments and statistical significance between unliganged and liganded GHS-R1a
was assessed using Student’s t-test (***P<0.001, **P < 0.01, *P < 0.05). Kinetics of GTPγS binding to
Gq (C) and G13 (D) were carried out under the same conditions. GHS-R1a-catalyzed GTPγ S binding is
expressed as the % of maximal MK- 0677 stimulation. E, Kact (min
-1
) values (mean ± S.E.M, n=2)
were calculated from GTPγS binding kinetics using GraphPad Prism software.
Figure 11. Outline of the functional selectivity of GHS-R1a ligands at GHS-R1a signaling.
A: Agonists: Ghrelin, MK-0677 and JMV 1843 are agonists (+) for all pathways whereas JMV 2959,
JMV 3002 and JMV 3018 compounds are partial agonists for Gq and neutral (-) toward the other
pathways. Schematic drawings represent signaling pathways and in vivo effects promoted by ghrelin
upon binding to GHS-R1a. Ghrelin, the endogenous ligand of GHS-R1a activates Gq, Go, Gi and G13-
dependent pathways and induces arrestin recruitment and ERK1/2 phosphorylation, resulting in GH
secretion, food intake and addiction. JMV 2959 antagonizes ghrelin action at Go, Gi, arrestin and ERK
but inhibits only partially Gq resulting in inhibition of addiction and food intake but not to inhibition of
GH secretion. The possible connection between selectivity of JMV 2959 toward signaling pathways and
physiological responses remains to be established.
B: Inverse agonists at Gq and G13: K-(D-1-Nal)-F-wLL-NH
2
is an inverse agonist at both Gq an G13
whereas SPA, JMV 4484, and KwFwLL-NH
2
are inverse agonists for both Gq and G13. Physiological
consequences of the inverse agonism selectivity at Gq and G13 remain to be explored.
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
#+!
Table 1: Structures and binding properties of the GHS-R1a ligands
Structure
Ref.
Ki (nM)
WT A204E
Ghrelin
(1)
11 ± 5
MK 0677
(33)
7.2 ± 3
JMV 1843
(34)
350 ± 110
JMV 2959
(35)
64 ± 19
JMV 3002
(36)
6.9 ± 2.6
JMV 3011
(36)
3.5 ± 1.1
JMV 3018
(36)
9.0 ± 3.7
JMV 4484
-
ND
rPKPfQwFwLL-NH
2
(SPA)
(24)
75 ± 10
192 ± 65
K-(D-1-Nal)-FwLL-NH
2
(38)
ND
KwFwLL-NH
2
(37)
ND
Structures of the GHS-R1a ligands are described in details in references (Ref.)
Ki values were determined in this study from competition binding experiments performed by HTRF-
based ligand binding assay on HEK293T cells expressing WT GHS-R1a or its A204E mutant (see
Materials and Methods. Values are mean ± SEM of at least 3 independent experiments performed in
triplicate. ND: not determined
GSSFLSPEHQRVQQRKESKKPPAKLQPR
O
C-(CH
2
)
6
-CH
3
O
NH
2
O
N
H
O
N
O
N
S
O
O
NH
2
O
N
H
O
H
N
H
N
CHO
H
N
N
H
N
H
N
N
N
HN
O
O
H
2
N
N
H
N
N
N
HN
O
O
O
N
N
H
N
N
N
HN
O
O
O
N
NH
N
H
N
N
N
HN
O
O
N
N
H
O
N
N
N
O
HN
H
2
N
H
2
N
H
N
NH
O
N
H
O
H
N
O
N
H
O
O
H
N
H
2
N
H
2
N
O
N
H
O
H
N
HN
O
N
H
O
NH
2
N
H
O
O
H
N
HN
H
2
N
H
2
N
O
N
H
O
H
N
HN
O
N
H
O
NH
2
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
#"!
Table 2: Potencies and maximal efficacies of GHS-R1a ligands
EC
50
and E
max
were determined from dose response-curves performed in HEK293T cells expressing
GHS-R1a as described in Materials and Methods.
For the agonist and neutral compounds, E
max
is expressed as the % of maximal ghrelin. For the inverse
agonists compounds, E
max
is expressed as the maximal basal inhibition (100% inhibition corresponding
to the basal of mock-transfected HEK293T cells). Values are mean ± SEM of three independent
experiments performed in triplicate.
Agonists and neutrals
Ghrelin
MK 0677
JMV 1843
JMV 2959
JMV 3002
JMV 3018
JMV 3011
EC
50
E
max
EC
50
E
max
EC
50
E
max
EC
50
E
max
EC
50
E
max
EC
50
E
max
EC
50
E
max
nM
% ghrelin
nM
% ghrelin
nM
% ghrelin
nM
% ghrelin
nM
% ghrelin
nM
% ghrelin
nM
% ghrelin
IP1
1.6 ± 1.1
100
0.83 ± 0.08
150 ± 25
0.89 ± 0.10
90 ± 10
NC
8 ± 2.0
24 ± 5
30 ± 5
NC
8 ± 3
NC
0 ± 2
Gq
7.4 ± 1.5
100
0.69 ± 0.2
118 ± 7
1.3 ± 0.3
107 ± 5
50 ± 10
44 ± 10
45 ± 15
38 ± 10
87 ± 20
29 ± 8
NC
1 ± 2
Gi2
17 ± 4
100
7.6 ± 1.4
100 ± 3
8.5 ± 2.9
90 ± 2
NC
3 ± 2
NC
1.5 ± 1
NC
1.7 ± 1
NC
NC
Gi3
15 ± 4
100
7.8 ± 1.5
102 ± 3
9.1 ± 2.6
91 ± 2
ND
ND
ND
ND
ND
ND
ND
ND
Goa
5.2 ± 0.8
100
6.6 ± 0.9
103 ± 2
6.7 ± 2.2
100 ± 3
ND
ND
ND
ND
ND
ND
ND
ND
Gob
7.8 ± 1.9
100
8.3 ± 2.4
99 ± 4
6.3 ± 2.5
85 ± 3
NC
5 ± 3
NC
0.5 ± 1
NC
1.5 ± 1
NC
NC
G13
NC
NC
NC
NC
NC
NC
NC
NC
NC
NC
NC
NC
NC
NC
β-arrestin2
1.0 ± 0.1
100
0.25 ± 0.15
111 ± 9
5.2 ± 1.9
80 ± 5
NC
1 ± 1
NC
0 ± 2
NC
1 ± 2
NC
0 ± 1
ERK1/2
9.9 ± 3.5
100
0.91 ± 0.44
102 ± 6
6.4 ± 2.1
102 ± 4
NC
NC
NC
NC
NC
NC
NC
NC
!
Inverse agonists
!
SPA
JMV 4484
K-(D-1-Nal)-FwLL-NH
2
KwFwLL-NH
2
!
EC
50
E
max
EC
50
E
max
EC
50
E
max
EC
50
E
max
!
nM
% maximal
basal inhibition
nM
% maximal
basal inhibition
nM
% maximal
basal inhibition
nM
% maximal
basal inhibition
IP1
75 ± 15
85 ± 10
53 ± 12
35 ± 5
30 ± 10
66 ± 6
100 ± 20
65 ± 10
Gq
ND
80 ± 2
ND
40 ± 3
ND
46 ± 4
ND
65 ± 3
G13
NC
5 ± 5
NC
5 ± 4
NC
57 ± 4
NC
3 ± 3
ND: not determined, NC: cannot be determined
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
##!
!!
!
!
!
!
!
!
!
!
!
!
!
!!
IP1-ERK1/2
BF
1.00
0.56
1.45
!!
!!
!!
!!
!!
!!
!!
!!
∆∆log(t/K
A
)
0.00 ± 0.47
-0.25 ± 0.46
0.16 ± 046
ERK1/2
log(t/K
A
)
0.00 ± 0.42
0.96 ± 0.35
0.04 ± 0.42
NC
NC
NC
NC
!!
Gq-ERK1/2
BF
1.00
1.48
3.51
log(t/K
A
)
7.89 ± 0.30
8.85 ± 0.25
7.93 ± 0.31
NC
NC
NC
NC
∆∆log(t/K
A
)
0.00 ± 0.43
0.17 ± 0.36
0.55 ± 0.43
β-arrestin2
log(t/K
A
)
0.00 ± 0.12
0.78 ± 0.10
.-0.44 ± 0.39
NC
NC
NC
NC
!!
IP1-βarrestin2
BF
1.00
0.85
4.37
log(t/K
A
)
8.82- ± 0.08
9.60 ± 0.05
8.38 ± 0.38
NC
NC
NC
NC
∆∆log(t/K
A
)
0.00 ± 0.10
-0.07 ± 0.10
0.64 ± 0.43
Ip1
log(t/K
A
)
0.00 ± 0.21
0.71 ± 0.29
0.21 ± 0.17
NC
NC
NC
NC
!!
Gq-βarrestin2
BF
1.00
2.29
10.47
log(t/K
A
)
8.50 ± 0.15
9.21 ± 0.21
8.72 ± 0.12
NC
NC
NC
NC
∆∆log(t/K
A
)
0.00 ± 0.13
0.36 ± 0.12
1.02 ± 0.40
Gob
log(t/K
A
)
0.00 ± 0.14
-0.15 ± 0.19
0.08 ± 0.17
NC
NC
NC
NC
!!
Gq-IP1
BF
1.00
2.63
2.43
log(t/K
A
)
8.09 ± 0.095
7.95 ± 0.16
8.17 ± 0.13
NC
NC
NC
NC
∆∆log(t/K
A
)
0.00 ± 0.22
0.42 ± 0.30
0.39 ± 0.19
Goa
log(t/K
A
)
0.00 ± 0.16
0.03 ± 0.12
.-0.11 ± 0.13
NC
NC
NC
NC
!!
Gq-Gob
BF
1.00
18.84
3.24
log(t/K
A
)
8.21 ± 0.11
8.24 ± 0.04
8.10 ± 0.06
NC
NC
NC
NC
∆∆log(t/K
A
)
0.00 ± 0.16
1.28 ± 0.20**
0.51 ± 0.18
Gi3
log(t/K
A
)
0.00 ± 0.04
0.13 ± 0.19
0.23 ± 0.16
NC
NC
NC
NC
!!
Gq-Goa
BF
1.00
12.59
4.95
log(t/K
A
)
7.85 ± 0.03
7.98 ± 0.19
8.11 ± 0.16
NC
NC
NC
NC
∆∆log(t/K
A
)
0.00 ± 0.18
1.10 ± 0.14**
0.69 ± 0.18
Gi2
log(t/K
A
)
0.00 ± 0.20
0.20 ± 0.14
0.32 ± 0.14
NC
NC
NC
NC
!!
Gq-Gi3
BF
1.00
10.12
2.10
log(t/K
A
)
7.80 ± 0.14
8.06 ± 0.03
8.12 ± 0.01
NC
NC
NC
NC
∆∆log(t/K
A
)
0.00 ± 0.10
1.01 ± 0.20*
0.32 ± 0.18
Gq
log(t/K
A
)
0.00 ± 0.09
1.13 ± 0.06
0.59 ± 0.08
-0.67 ± 0.14
-1.08 ± 0.26
-0.99 ± 0.16
NC
!!
Gq-Gi2
BF
1.00
6.46
1.86
log(t/K
A
)
8.19 ± 0.06
9.32 ± 0.01
8.77 ± 0.04
7.52 ± 0.12
7.11 ± 0.25
7.20 ± 0.15
NC
∆∆log(t/K
A
)
0.00 ± 0.22
0.81 ± 0.16*
0.27 ± 0.16
!
!
Ghrelin
MK 0677
JMV 1843
JMV 2959
JMV 3002
JMV 3018
JMV 3011
!
!
!
Ghrelin
MK 0677
JMV 1843
Table 3: Bias factors of GHS-R1a ligands
Data were analysed using the operational model with ghrelin as the reference ligand as described in Materials and Methods. Quantification of bias was
only possible for ghrelin, MK 0677 and JMV 1843. Values are mean ± SEM of three independent experiments performed in triplicate.
Statistical analysis was performed using a two-way unpaired Student’s t test (**P < 0.01, *P < 0.05). BF= 10
∆∆log(t/KA
NC: cannot be determined
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
#$!
M'KADMI Figure 1
A
B
2.5 12 18 63 74
0
20
40
60
80
100
120
140
GHS-R1a (fmol/ 10
5
cells)
Basal IP1 accumultation (% of maximal ghrelin)
2.5 74
0
20
40
60
80
100
120
140
GHS-R1a (fmol/ 10
5
cells)
IP1 accumulation (% of maximal ghrelin)
Basal
Ghrelin
JMV1843
MK 0677
JMV 3002
JMV 3018
JMV 2959
JMV 3011
SPA
*
*
*
*
*
*
***
***
***
***
***
***
Basal
Ghrelin
JMV 1843
MK 0677
JMV 3002
JMV 3018
JMV 2959
JMV 3011
SPA
0
20
40
60
80
100
120
140
IP1 accumulation (% of max ghrelin)
***
***
*
**
*
*
**
C
D
Basal
Ghrelin
JMV 1843
MK 0677
JMV 3002
JMV 3018
JMV 2959
JMV 3011
SPA
0
20
40
60
80
100
120
140
IP1 accumulation (% of max Ghrelin)
***
***
***
***
**
**
*
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
#%!
-12 -11 -10 -9 -8 -7 -6 -5 -4
0
25
50
75
100
125
150
175
log [ligand] (M)
Ligand promoted β-arrestin2 recruitment
(% of maximal ghrelin)
MK 0677
Ghrelin
JMV 2959
JMV 3002
JMV 3018
JMV 3011
SPA
JMV 1843
M'KADMI Figure 2
-12 -11 -10 -9 -8 -7 -6 -5 -4
0
25
50
75
100
125
150
175
log [ligand] (M)
Ligand promoted ERK1/2 phosphorylation
(% of maximal ghrelin)
MK 0677
Ghrelin
JMV 1843
JMV 2959
JMV 3002
JMV 3018
JMV 3011
SPA
Basal
Ghrelin
JMV 1843
MK 0677
JMV 3002
JMV 3018
JMV 2959
JMV 3011
SPA
0
20
40
60
80
100
120
140
β-arrestin2 recruitment (% of maximal ghrelin)
***
***
***
**
Basal
Ghrelin
JMV 1843
MK 0677
JMV 3002
JMV 3018
JMV 2959
JMV 3011
SPA
0
20
40
60
80
100
120
140
ERK1/2 phosphorylation (% of maximal ghrelin)
**
**
**
*
A
B
C
D
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
#&!
JMV 3002
JMV 3011
JMV 3018
JMV 2959
0
20
40
60
80
100
120
140
arrestin recruitment (% of maximal ghrelin)
Ghrelin 10
-8
M
-
+
+
+
+
+
Compound 10
-6
M
-
-
+
+
+
+
***
A
B
β-arrestin2
M'KADMI Figure 3
0
20
40
60
80
100
120
140
IP1 accumulation (% of maximal ghrelin)
**
*
*
*
Ghrelin 10
-8
M
-
+
+
+
+
Compound 10
-6
M
-
-
+
+
+
+
+
JMV 3002
JMV 3011
JMV 3018
JMV 2959
JMV 3002
JMV 3011
JMV 3018
JMV 2959
0
20
40
60
80
100
120
140
ERK1/2 activation ( % of maximal ghrelin)
Ghrelin 10
-8
M
-
+
+
+
+
+
Compound 10
-6
M
-
-
+
+
+
+
***
C
ERK1/2
IP1
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
#'!
0 20 40 60 80 100 120
0.80
0.85
0.90
0.95
1.00
1.05
Time (sec)
BRET ratio
MK0677
Goa
0 20 40 60 80 100 120
1.3
1.4
1.5
1.6
1.7
1.8
1.9
Time (sec)
BRET ratio
MK0677
Gi2
0 25 50 75 100 125
2.0
2.1
2.2
2.3
2.4
2.5
MK0677
Time (sec)
BRET ratio
Gi1
0 20 40 60 80 100 120
0.0
0.2
0.4
0.6
0.8
1.0
Time (sec)
BRET ratio
MK0677
Gq
0 20 40 60 80 100 120
0.20
0.22
0.24
0.26
Time (sec)
BRET ratio
MK0677
Gs
0 20 40 60 80 100 120
1.5
1.6
1.7
1.8
1.9
2.0
Time (sec)
BRET ratio
MK0677
Gob
0 20 40 60 80 100 120
1.3
1.4
1.5
1.6
1.7
Time (sec)
BRET ratio
MK0677
Gi3
0 20 40 60 80 100 120
0.20
0.25
0.30
0.35
0.40
Time (sec)
BRET ratio
MK0677
G12
0 25 50 75 100 125 150
0.22
0.24
0.26
0.28
0.30
0.32
0.34
Time (sec)
BRET ratio
MK0677
G13
A
Gq Gi1 Gi2 Gi3 Goa Gob G12 G13 Gs
-0.3
-0.2
-0.1
0.0
MK 0677-promoted BRET
**
**
*
*
*
*
*
B
C
M'KADMI Figure 4
0 10 20 30 40 50 60
0.76
0.78
0.80
0.82
Time (sec)
BRET Ratio
AVP
0 10 20 30 40 50 60
0.30
0.35
0.40
0.45
Time (sec)
BRET Ratio
U46619
D
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
#(!
-13 -12 -11 -10 -9 -8 -7 -6 -5
0
20
40
60
80
100
120
140
160
log [ligand] (M)
Ligand-promoted IP1 production
(% of maximal ghrelin)
MK 0677
Ghrelin
JMV 1843
IP1 PRODUCTION
M'KADMI Figure 5
A
B
-12 -11 -10 -9 -8 -7 -6 -5
0
20
40
60
80
100
120
140
160
log [ligand] (M)
Ligand-promoted Gq activation
(% of maximal ghrelin)
JMV 1843
Ghrelin
MK0677
Gq ACTIVATION
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
#)!
Gq
-12 -11 -10 -9 -8 -7 -6 -5
0.00
0.05
0.10
0.15
0.20
log [ligand] (M)
Ligand-promoted BRET decrease
Ghrelin
MK 0677
JMV 1843
JMV 2959
JMV 3002
JMV 3011
JMV 3018
Gi3
-12 -11 -10 -9 -8 -7 -6 -5 -4
0.00
0.05
0.10
0.15
log [ligand] (M)
Ligand promoted BRET decrease
MK 0677
Ghrelin
JMV 1843
Gob
-12 -11 -10 -9 -8 -7 -6 -5 -4
0.00
0.05
0.10
0.15
log [ligand] (M)
Ligand promoted BRET decrease
MK 0677
Ghrelin
JMV 1843
JMV 2959
JMV 3002
JMV 3018
JMV 3011
Gi2
-12 -11 -10 -9 -8 -7 -6 -5 -4
0.00
0.05
0.10
0.15
log [ligand] (M)
Ligand promoted BRET decrease
JMV 1843
Ghrelin
MK 0677
JMV 2959
JMV 3002
JMV 3018
JMV 3011
Goa
-12 -11 -10 -9 -8 -7 -6 -5 -4
0.00
0.05
0.10
0.15
log [ligand] (M)
Ligand promoted BRET decrease
Ghrelin
MK 0677
JMV 1843
M'KADMI Figure 6
A
B
C
D
E
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
#*!
-0.15
-0.10
-0.05
0.00
Ligand-promoted BRET
Gq
Gi2
Gob
Ghrelin (10
-7
M)
+
+
+
+
-
-
-
JMV compound (10
-6
M)
-
+
+
+
+
+
+
+
-
+
+
JMV 2959
JMV 3002
JMV 3018
JMV 3011
JMV 2959
JMV 3002
JMV 3018
JMV 3011
**
***
***
***
***
***
***
***
***
***
***
M'KADMI Figure 7
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
$+!
0 50 100 150 200 250
-0.20
-0.15
-0.10
-0.05
0.00
0.05
0.10
0.15
0.20
Ligand-promoted IP1 (% of basal)
Ligand-promoted Gq activation (BRET signal change)
SPA
K-(D-1-Nal)-FwLL-NH
2
KwFwLL-NH
2
JMV 4484
JMV 3011
JMV 2959
JMV 3002
JMV 3018
Ghrelin
JMV 1843
MK 0677
A
B
C
M'KADMI Figure 8
basal
JMV 4484
K-(D-1Nal)-FwLL-NH
2
KwFwLL-NH
2
SPA
0
20
40
60
80
100
120
140
IP1 production (% of basal)
**
***
***
***
Basal
JMV 4484
K-(D-1-Nal)-F-wLL-NH
2
KwFwLL-NH
2
SPA
0.00
0.02
0.04
0.06
0.08
0.10
Ligand-promoted BRET
*
**
*
**
IP1
Gq
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
$"!
0 0.001 0.01 0.1 1 2 4
0.0
0.5
1.0
1.5
2.0
HA-GHSR1a encoding vector (µg)
Basal BRET ratio
***
***
***
*
0 0.001 0.01 0.1 1 2 4
0.0
0.2
0.4
0.6
0.8
1.0
HA-GHSR1a encoding vector (µg)
Basal BRET ratio
***
***
***
***
***
**
0 0.001 0.01 0.1 1 2 4
0
50 000
100 000
150 000
HA-GHSR1a encoding vector (µg)
Relative expression of
Gα-Rluc8 proteins (Luminescence)
0 0.001 0.01 0.1 1 2 4
0
50 000
100 000
150 000
200 000
HA-GHSR1a encoding vector (µg)
Relative expression of
Gα-Rluc8 proteins (Luminescence)
0 0.001 0.01 0.1 1 4
0.0
0.2
0.4
0.6
0.8
1.0
HA-GHSR1a encoding vector (µg)
Cell surface HA-GHSR1a density
(OD
450nm
- OD
570nm
)
0 0.001 0.01 0.1 1 4
0.0
0.2
0.4
0.6
0.8
1.0
HA-GHSR1a encoding vector (µg)
Cell surface HA-GHSR1a density
(OD
450nm
- OD
570nm
)
-0.020
-0.015
-0.010
-0.005
0.000
0.005
0.010
Gα13 activation
Ligand modulated BRET
**
***
*
*
Ghrelin
JMV 1843
MK 0677
JMV 2959
SPA
JMV 4484
KwFwLL-NH
2
K-(D-1-Nal)-F-wLL-NH
2
JMV 3002
JMV 3018
-0.25
-0.20
-0.15
-0.10
-0.05
0.00
0.05
0.10
Gαq activation
Ligand-promoted BRET
JMV 4484
K-(D-1-Nal)-F-wLL-NH
2
KwFwLL-NH
2
SPA
Ghrelin
**
*
*
**
***
MK 0677
***
***
*
*
*
JMV 1843
JMV 2959
JMV 3002
JMV 3018
A
F
E
D
C
B
H
G
M'KADMI Figure 9
Gq
Gq
Gq
Gq
G13G13
G13
G13
G13
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
$#!
G13
Gq
A
B
M'KADMI Figure 10
0.0 2.5 5.0 7.5 10.0 12.5 15.0 17.5 20.0
0
20
40
60
80
100
120
Time (min)
GTPγS binding
(% of max MK0677 stimulation)
basal
MK 0677
SPA
K-(D-1-Nal)-F-wLL-NH
2
KwFwLL-NH
2
JMV 4484
0.0 2.5 5.0 7.5 10.0 12.5 15.0 17.5 20.0
0
20
40
60
80
100
120
Time (min)
GTPγS binding
(% of max MK0677 stimulation)
basal
MK 0677
SPA
K-(D-1-Nal)-F-wLL-NH
2
KwFwLL-NH
2
JMV 4484
C
D
G13
Gq
Basal MK 0677 SPA
K-(D-1-Nal)-FwLL-NH
2
KwFwLL-NH
2
JMV4484
Gq 0.185 ± 0.003 0.902 ± 0.001 0.019 ± 0.002 0.047 ± 0. 004 0.060 ± 0.014 0.074 ± 0.014
G13 0.101 ± 0.007 0.246 ± 0.002 0.104 ± 0.006 0.051 ± 0.002 0.112 ± 0.014 0.112 ± 0.013
Kact (min
-1
)
E
Basal
MK0677
SPA
K-(D-1-Nal)-F-wLL-NH2
KwFwLL-NH2
JMV4484
0
200
400
600
800
1000
GTPγS binding
fluorescence intensity (a.u.)
***
***
**
***
**
basal
MK0677
SPA
K-(D-1-Nal)-F-wLL-NH2
KwFwLL-NH2
JMV4484
0
200
400
600
800
1000
GTPγS binding
fluorescence intensity (a.u.)
***
***
! !!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!!Biased'signaling'at'the'ghrelin'receptor'
!
!
$$!
!
AGONISTS
Ghrelin
MK 0677
JMV 2959
JMV 3002
JMV 3018
+ + + + +
+ + -
+ +
- -
- - -
JMV 1843
+
+
+
+ + - - - +
+ + - - - +
β-arrestin 2
Gq
Gi2
Gob
ERK1/2
G13
Gq
INVERSE AGONISTS
KwFwLL-NH
2
SPA
JMV 4484
K-(D-1-Nal)-F-wLL-NH
2
+ +
+
+ +
- - -
Gq
Gi
Go
ERK1/2
Food
intake
GH
Addiction
Gq
Gi
Go
ERK1/2
Arr
Arr
+ + +
GH
+
-
-
Food
intake
Addiction
Ghrelin
JMV 2959
Ghrelin
A
B
GHS-R1a
GHS-R1a
G13
G13
G13
+ + +
-
-
-
M'KADMI Figure 11

Supplementary resource (1)

... GHSR-1a is a G-protein coupled receptor (GPCR) that uses two different pathways to send signals. These pathways are called Gαq/11 and β-arrestin signaling, and Gαi/o and Gα12/13 signaling (Damian et al., 2012;M'Kadmi et al., 2015). Gαq/11 increases the intracellular calcium. ...
... Conversely, the GHSR antagonist EP-80317 inhibited pilocarpine-induced seizures and status epilepticus in rats, and suppressed seizures in mice in a model of 6-Hz corneal stimulation (Lucchi et al., 2017). It is well known that some GHSR ligands trigger certain receptor conformations that lead to the activation of specific intracellular pathways, a process named biased signaling (M'Kadmi et al., 2015). For example, YIL781 activates Gαq/11 and Gα12, but does not recruit β-arrestin and so does not induce a rapid termination of Gαq/11 signaling. ...
... The observed inverse agonism in a β-arrestin2 recruitment assay has so far not been reported for ACKR3 ligands and constitutive recruitment of a β-arrestin2 has so far only been incidentally reported for other GPCRs. [41][42][43][44] . CC-BY-NC-ND 4.0 International license perpetuity. ...
Preprint
Full-text available
The atypical chemokine receptor 3 (ACKR3) has emerged as a promising drug target for the treatment of cancer, cardiovascular, and autoimmune diseases. In this study, we present the pharmacological characterization of VUF16840, the first small-molecule inverse agonist of ACKR3. VUF16840 effectively displaces CXCL12 binding to ACKR3 and inhibits chemokine-induced β-arrestin2 recruitment in a concentration-dependent manner. Furthermore, VUF16840 stabilizes the inactive conformation of ACKR3, as demonstrated by its ability to suppress constitutive β-arrestin2 recruitment. This inverse agonism alters ACKR3 constitutive trafficking, leading to receptor enrichment at the plasma membrane and inhibition of intracellular CXCL12 uptake. Importantly, VUF16840 exhibits high selectivity for ACKR3 over a broad panel of human chemokine receptors. These findings establish VUF16840 as a potent and selective ACKR3 inverse agonist capable of modulating constitutive and chemokine-induced signaling and internalization events. As such, VUF16840 represents a valuable pharmacological tool for exploring the molecular and translational roles of ACKR3 in both physiological and pathological contexts.
... It is often called the "hunger hormone" and is secreted into the bloodstream by the stomach to stimulate appetite. The ghrelin receptor, the growth hormone secretagogue receptor, is a typical rhodopsin-like GPCR that interacts with multiple G proteins [33,34]. Ghrelin exerts its orexigenic effect predominantly via activating neurons that co-express NPY and AgRP in the ARC [35]. ...
Article
Full-text available
Endothermic animals expend significant energy to maintain high body temperatures, which offers adaptability to varying environmental conditions. However, this high metabolic rate requires increased food intake. In conditions of low environmental temperature and scarce food resources, some endothermic animals enter a hypometabolic state known as torpor to conserve energy. Torpor involves a marked reduction in body temperature, heart rate, respiratory rate, and locomotor activity, enabling energy conservation. Despite their biological significance and potential medical applications, the neuronal mechanisms regulating torpor still need to be fully understood. Recent studies have focused on fasting‐induced daily torpor in mice due to their suitability for advanced neuroscientific techniques. In this review, we highlight recent advances that extend our understanding of neuronal mechanisms regulating torpor. We also discuss unresolved issues in this research field and future directions.
... Indeed, inverse agonists inhibit the constitutive activity of a receptor, by switching the receptor from an active R* to an inactive R conformational state. Consequently, inverse agonists should be prone to increase the BRET signal between Gαq and Gγ2 BRET biosensors, reflecting Gαq/Gγ2 reassembly (Fig. 1a) [17,25]. This inhibition on P2Y 1 -R constitutive activity was dependent on receptor amount since MRS2179-modulated BRET signal increased with cell surface P2Y 1 -R expression level ( Fig. 1e and Additional File 3: Fig. S3a, left panel), while Gαq protein BRET probe expression remained constant (Additional File 1: Fig. S1c). ...
Article
Full-text available
Background Purinergic P2Y1 and P2Y12 receptors (P2Y1-R and P2Y12-R) are G protein-coupled receptors (GPCR) activated by adenosine diphosphate (ADP) to mediate platelet activation, thereby playing a pivotal role in hemostasis and thrombosis. While P2Y12-R is the major target of antiplatelet drugs, no P2Y1-R antagonist has yet been developed for clinical use. However, accumulating data suggest that P2Y1-R inhibition would ensure efficient platelet inhibition with minimal effects on bleeding. In this context, an accurate characterization of P2Y1-R antagonists constitutes an important preliminary step. Results Here, we investigated the pharmacology of P2Y1-R signaling through Gq and β-arrestin pathways in HEK293T cells and in mouse and human platelets using highly sensitive resonance energy transfer-based technologies (BRET/HTRF). We demonstrated that at basal state, in the absence of agonist ligand, P2Y1-R activates Gq protein signaling in HEK293T cells and in mouse and human platelets, indicating that P2Y1-R is constitutively active in physiological conditions. We showed that P2Y1-R also promotes constitutive recruitment of β-arrestin 2 in HEK293T cells. Moreover, the P2Y1-R antagonists MRS2179, MRS2279 and MRS2500 abolished the receptor dependent-constitutive activation, thus behaving as inverse agonists. Conclusions This study sheds new light on P2Y1-R pharmacology, highlighting for the first time the existence of a constitutively active P2Y1-R population in human platelets. Given the recent interest of P2Y12-R constitutive activity in patients with diabetes, this study suggests that modification of constitutive P2Y1-R signaling might be involved in pathological conditions, including bleeding syndrome or high susceptibility to thrombotic risk. Thus, targeting platelet P2Y1-R constitutive activation might be a promising and powerful strategy for future antiplatelet therapy.
Article
G protein–coupled receptor (GPCR) signaling is a dynamic process involving various conformational intermediates in addition to those captured in static three-dimensional structures. Here, we used newly developed G protein peptidomimetics to characterize the interactions of the ghrelin receptor (GHSR) with G proteins. Coupling to the G protein peptidomimetic not only affected the conformational features of the cytoplasmic regions of the receptor where the G protein binds but also allosterically affected the extracellular ligand-binding pocket. These conformational and allosteric changes increased the affinity of G protein–coupled GHSR for the endogenous agonist ghrelin. In addition, our data identified different complexes along the G protein activation pathway that differed in the engagement of the Gα q C-terminal helix. Given that this helix is the main link between the activated receptor and the Gα nucleotide-binding pocket, these findings suggested a stepwise process involving distinct states in GPCR-catalyzed G protein activation. Collectively, our results provide evidence for the dynamic behavior of GPCR–G protein signaling complexes, with such dynamics most likely contributing to signaling selectivity and/or efficacy.
Article
Ethnopharmacological relevance The dried fruit of Ligustrum lucidum Ait. (FLL), known as Nuzhenzi, is traditionally recognized for its anti-aging properties in Chinese medicine. Nuezhenide, a water-soluble secoiridoid present in FLL, has demonstrated various pharmacological activities including neuroprotection, enhancement of learning and memory, antiosteoporotic, and antiviral activities. These therapeutic benefits align with the anti-aging effects attributed to ghrelin, particularly in the modulation of growth hormone secretagogue receptor type 1a (GHSR-1a) signaling. Aim of the work This study aimed to investigate the potential of FLL extracts, particularly its major compound nuezhenide, as agonists of GHSR-1a, a receptor implicated in anti-aging mechanisms, utilizing a stable GHSR-1a-expressing cell line. Materials and methods HEK293T cells expressing GHSR-1a-mCherry were used to assess the effects of FLL extract and its major compound, nuezhenide, on cell viability and ERK1/2 signaling. Molecular docking simulations predicted the interaction between nuezhenide and the GHSR-1a binding pocket. The impact of nuezhenide on ERK1/2 phosphorylation was evaluated, along with the involvement of phospholipase C and calcium signaling in this process. Results Molecular docking simulations indicated that nuezhenide could interact with the GHSR-1a receptor, similar to teaghrelin, another known ghrelin analog. Experimental data showed that FLL extracts and nuezhenide enhanced cell viability and ERK1/2 activation in GHSR1a-mCherry HEK293T cells. The effect was specifically mediated by GHSR-1a, as confirmed by SP-analog treatment. Further analysis revealed that nuezhenide-induced ERK1/2 activation is likely mediated through a phospholipase C-dependent pathway involving intracellular calcium release. Conclusion This study demonstrated for the first time that nuezhenide acts as a putative GHSR-1a agonist, promoting cell proliferation and activating ERK1/2 signaling via phospholipase C and calcium pathways. These findings support the traditional use of FLL as an anti-aging herbal remedy and suggest that nuezhenide could be developed as a therapeutic agent targeting GHSR-1a-mediated pathways.
Article
Objective Although the metabolic state of an organism affects olfactory function, the precise mechanisms and their impact on behavior and metabolism remain unknown. Here, we assess whether ghrelin receptors (GHSRs) in the olfactory bulb (OB) increase olfactory function and influence foraging behaviors and metabolism. Methods We performed a detailed behavioural and metabolic analysis in mice lacking GHSRs in the OB (OBGHSR deletion). We also analsyed OB scRNA-seq and spatial transcriptomic datasets to assess GHSR+ cells in the main and accessory olfactory bulbs, as well as the anterior olfactory nucleus. Results OBGHSR deletion affected olfactory discrimination and habituation to both food and non-food odors. Anxiety-like and depression-like behaviors were significantly greater after OBGHSR deletion, whereas exploratory behavior was reduced, with the greatest effect under fasted conditions. OBGHSR deletion impacted feeding behavior as evidenced by altered bout number and duration, as well as buried food-seeking. OBGHSR deletion increased body weight and fat mass, spared fat utilisation on a chow diet and impaired glucose metabolism indicating metabolic dysfunction. Cross referenced analysis of OB scRNA-seq and spatial transcriptomic datasets revealed GHSR+ glutamate neurons in the main and accessory olfactory bulbs, as well as the anterior olfactory nucleus. Ablation of glutamate neurons in the OB reduced ghrelin-induced food finding and phenocopied results seen after OBGHSR deletion. Conclusions OBGHSRs help to maintain olfactory function, particularly during hunger, and facilitate behavioral adaptations that optimise food-seeking in anxiogenic environments, priming metabolic pathways in preparation for food consumption.
Article
Central ghrelin signaling seems to play important role in addiction as well as memory processing. Antagonism of the growth hormone secretagogue receptor (GHS-R1A) has been recently proposed as a promising tool for the unsatisfactory drug addiction therapy. However, molecular aspects of GHS-R1A involvement in specific brain regions remain unclear. The present study demonstrated for the first time that acute as well as subchronic (4 days) administration of the experimental GHS-R1A antagonist JMV2959 in usual intraperitoneal doses including 3 mg/kg, had no influence on memory functions tested in the Morris Water Maze in rats as well as no significant effects on the molecular markers linked with memory processing in selected brain areas in rats, specifically on the β-actin, c-Fos, two forms of the calcium/calmodulin-dependent protein kinase II (CaMKII, p-CaMKII) and the cAMP-response element binding protein (CREB, p-CREB), within the medial prefrontal cortex (mPFC), nucleus accumbens (NAc), dorsal striatum, and hippocampus (HIPP). Furthermore, following the methamphetamine intravenous self-administration in rats, the 3 mg/kg JMV2959 pretreatment significantly reduced or prevented the methamphetamine-induced significant decrease of hippocampal β-actin and c-Fos as well as it prevented the significant decrease of CREB in the NAC and mPFC. These results imply, that the GHS-R1A antagonist/JMV2959 might reduce/prevent some of the memory-linked molecular changes elicited by methamphetamine addiction within brain structures associated with memory (HIPP), reward (NAc), and motivation (mPFC), which may contribute to the previously observed significant JMV2959-induced reduction of the methamphetamine self-administration and drug-seeking behavior in the same animals. Further research is necessary to corroborate these results.
Article
Full-text available
Significance G protein-coupled receptors (GPCRs), one of the largest cell surface receptor families, transmit their signals through the coupling of intracellular partners, such as the G proteins. Knowing how this coupling occurs is essential, because it governs the entire signaling process. To address this open question, we used a purified GPCR as a model to which we applied various state-of-the-art biochemical and biophysical approaches. By doing so, we provide direct experimental evidence of a signaling mechanism in which receptor conformational changes are directly linked to a rearrangement of a preassembled complex between the receptor and its cognate Gq protein. This sheds light on the way in which a GPCR interacts with G proteins to trigger signaling.
Article
Full-text available
Context: The ghrelin receptor GHS-R1a is highly expressed in human somatotroph adenomas and exhibits unusually high basal signaling activity. In humans, the suppression of this constitutive activity by mutation induces a short stature. Objective: Using a GHS-R1a inverse agonist, modified substance P (MSP), we explored the role of GHS-R1a constitutive activity in GH hypersecretion from somatotroph adenomas and as a putative therapeutic target. Design: The effects of MSP were assessed on GH secretion from 19 human somatotroph tumors in vitro. Moreover, these effects were compared with those of octreotide (somatostatin receptor subtype 2 [sst2] agonist) and with the combination of both drugs. Expression and localization of GHS-R1a and sst2 were studied. Results: For all tumors, MSP inhibited GH secretion in a dose-dependent manner from 13 to 64%. Moreover, MSP enhanced octreotide-induced GH inhibition. For five tumors, the effects of combined MSP plus octreotide treatment were significantly higher than the sum of effects of each drug alone. MSP increased the membrane localization of GHS-R1a and of microdomains colocalizing sst2-GHS-R1a, highlighting the cooperation between the two drugs. Conclusions: The GHS-R1a inverse agonist could open new therapeutic options for acromegalic patients, particularly patients partially sensitive to octreotide whose GH secretion is not completely controlled by the sst2 agonist.
Article
Full-text available
The G protein-coupled ghrelin receptor GHSR1a is a potential pharmacological target for treating obesity and addiction because of the critical role ghrelin plays in energy homeostasis and dopamine-dependent reward. GHSR1a enhances GH release, appetite, and dopamine signaling through Gq/11, Gi/o and G12/13 as well as β-arrestin based scaffolds. However, the contribution of individual G protein and β-arrestin pathways to the diverse physiological responses mediated by ghrelin remains unknown. To characterize if signaling bias occurs for the GHSR1a, we investigated ghrelin signaling in a number of cell-based assays, including Ca2+ mobilization, SRF-RE, stress fiber formation, ERK1/2 phosphorylation and β-arrestin translocation, utilizing intracellular second loop (ICL2) and C-tail mutants of GHSR1a. We observed that GHSR1a and β-arrestin rapidly form metastable plasma-membrane complexes following exposure to agonist, but replacement of the GHSR1a C-tail by the tail of the vasopressin 2 receptor greatly stabilizes them, producing complexes observable on the plasma membrane and also in endocytic vesicles. Mutations of the contiguous conserved amino acids P148 and L149 in GHSR1a ICL2 generate receptors with strong bias to G protein and β-arrestin, respectively, supporting a role for conformation dependent signaling bias in the wild type receptor. Our results demonstrate more balance in GHSR1a-mediated ERK signaling from G proteins and β-arrestin, but uncover an important role for β-arrestin in RhoA activation and stress fiber formation. These findings suggest an avenue for modulating drug abuse-associated changes in synaptic plasticity via GHSR1a and indicate the development of GHSR1a biased ligands as a promising strategy for selectively targeting downstream signaling events.
Article
Full-text available
The growth hormone secretagogue receptor (GHSR), also known as the ghrelin receptor, is involved in mediating a wide variety of biological effects of ghrelin, including: stimulation of growth hormone release, increase of food intake and body weight, modulation of glucose and lipid metabolism, regulation of gastrointestinal motility and secretion, protection of neuronal and cardiovascular cells, and regulation of immune function. Dependent on the tissues and cells, activation of GHSR may trigger a diversity of signaling mechanisms and subsequent distinct physiological responses. Distinct regulation of GHSR occurs at levels of transcription, receptor interaction and internalization. Here we review the current understanding on the intracellular signaling pathways of GHSR and its modulation. An overview of the molecular structure of GHSR is presented first, followed by the discussion on its signaling mechanisms. Finally, potential mechanisms regulating GHSR are reviewed.
Article
Full-text available
Ghrelin is a gastric peptide hormone, discovered as being the endogenous ligand of growth hormone secretagogue receptor. Ghrelin is a 28 amino acid peptide presenting a unique n-octanoylation modification on its serine in position 3, catalyzed by ghrelin O-acyl transferase. Ghrelin is mainly produced by a subset of stomach cells and also by the hypothalamus, the pituitary, and other tissues. Transcriptional, translational, and posttranslational processes generate ghrelin and ghrelin-related peptides. Homo- and heterodimers of growth hormone secretagogue receptor, and as yet unidentified receptors, are assumed to mediate the biological effects of acyl ghrelin and desacyl ghrelin, respectively. Ghrelin exerts wide physiological actions throughout the body, including growth hormone secretion, appetite and food intake, gastric secretion and gastrointestinal motility, glucose homeostasis, cardiovascular functions, anti-inflammatory functions, reproductive functions, and bone formation. This review focuses on presenting the current understanding of ghrelin and growth hormone secretagogue receptor biology, as well as the main physiological effects of ghrelin.
Article
Results presented in this study indicate that in human embryonic kidney 293 cells (HEK 293), the ghrelin receptor growth hormone secretagogue receptor type 1 a (GHS-R1a) activates the extracellular signal-related kinases 1 and 2 (ERK1/2) via three pathways. One pathway is mediated by the beta-arrestins 1 and 2, and requires entry of the receptor into a multiprotein complex with the beta-arrestins, Src, Raf-1, and ERK1/2. A second pathway is G(q/11)-dependent and involves a Ca(2+)-dependent PKC (PKC alpha/beta) and Src. A third pathway is G(i)-dependent and involves phosphoinositide 3-kinase (PI3K), PKC epsilon, and Src. Our current study reveals that G(i/o) and G(q/11)-proteins are crucially involved in the P-arrestin-mediated ERK1/2 activation. These results thus support the view that the beta-arrestins act as both scaffolding proteins and signal transducers in ERK1/2 activation, as reported for other receptors. The different pathways of ERK1/2 activation suggest that binding to GHS-R1a activates ERK1/2 pools at different locations within the cell, and thus probably with different physiological consequences.
Article
The fact that over 30% of current pharmaceuticals target heptahelical G protein-coupled receptors (GPCRs) attests to their tractability as drug targets. While GPCR drug development has traditionally focused on conventional agonists and antagonists, the growing appreciation that GPCRs mediate physiologically relevant effects via both G protein and non-G protein effectors has prompted the search for ligands that can 'bias' downstream signaling in favor of one or the other process. Biased ligands are novel entities with distinct signaling profiles dictated by ligand structure, and the potential prospect of biased ligands as better drugs has been pleonastically proclaimed. Indeed, preclinical proof-of-concept studies have demonstrated that both G protein and arrestin pathway-selective ligands can promote beneficial effects in vivo while simultaneously antagonizing deleterious ones. But along with opportunity comes added complexity and new challenges for drug discovery. If ligands can be biased, then ligand classification becomes assay dependent, and more nuanced screening approaches are needed to capture ligand efficacy across several dimensions of signaling. Moreover, because the signaling repertoire of biased ligands differs from that of the native agonist, unpredicted responses may arise in vivo as these unbalanced signals propagate. For any given GPCR target, establishing a framework relating in vitro efficacy to in vivo biological response is crucial to biased drug discovery. This review discusses approaches to describing ligand efficacy in vitro, translating ligand bias into biological response, and developing a systems-level understanding of biased agonism in vivo, with the overall goal of overcoming current barriers to developing biased GPCR therapeutics. The American Society for Pharmacology and Experimental Therapeutics.
Article
The only molecularly identified ghrelin receptor is the growth hormone secretagogue receptor GHSR1a. Its natural ligand, ghrelin, is an acylated peptide whose unacylated counterpart (UAG) is almost inactive at GHSR1a. A truncated, nonfunctional receptor, GHSR1b, derives from the same gene. We have critically evaluated evidence for effects of ghrelin receptor ligands that are not consistent with actions at GHSR1a. Effects of ghrelin are observed in cells or tissues where the expression of GHSR1a is not detectable or after the Ghsr gene has been inactivated. In several, effects of ghrelin are mimicked by UAG, and ghrelin binding is competitively reduced by UAG. Effects in the absence of GHSR1a and sites at which ghrelin and UAG have similar potency suggest the presence of novel nonspecific ghrelin receptors (ghrelin receptor-like receptors [GRLRs]). A third class of receptor, the UAG receptors, at which UAG, but not ghrelin, is an agonist has been proposed. None of the novel receptors, with the exception of the glycoprotein CD36, which accounts for ghrelin action at a limited number of sites, have been identified. GHSR1a and GHSR1b combine with other G protein-coupled receptors to form heterodimers, whose pharmacologies differ from their components. Thus, it is feasible some GRLRs and some UAG receptors are heterodimers. Effects mediated through GRLRs or UAG receptors include adipocyte lipid accumulation, myoblast differentiation, osteoblast proliferation, insulin release, cardioprotection, coronary artery constriction, vascular endothelial cell proliferation, and tumor cell proliferation. The molecular identification and pharmacologic characterization of novel ghrelin receptors are thus important objectives.
Article
At one time, G protein-coupled receptors were envisioned to simply relay either inhibitory or stimulatory binary signals through engaging particular G proteins. These receptors are now viewed as complex, multidimensional triggers of a variety of potential signaling cascades. This review will showcase current attempts to elucidate biased signaling and functional selectivity in tissues and organs as well as in the whole animal. In addition, it will emphasize the challenges that are inherent in attributing bias in a living system as well as offer opinions as to the manner in which these problems may be approached.