Sunteți pe pagina 1din 38

ARTICLE IN PRESS

Resonance Energy
Transfer-Based
Approaches to Study
GPCRs
Mohammed Akli Ayoub*, x
*Biologie et Bioinformatique des Syste`mes de Signalisation, Institut National de la Recherche
Agronomique, UMR85, Unite Physiologie de la Reproduction et des Comportements; CNRS,
UMR7247, Nouzilly, France
x
LE STUDIUM Loire Valley Institute for Advanced Studies, Orleans, France
E-mail: maayoub@tours.inra.fr

CHAPTER OUTLINE
Introduction ................................................................................................................ 2
1. RET Approaches ..................................................................................................... 3
2. Ligand Binding ....................................................................................................... 4
3. Ligand-Induced Conformational Changes ................................................................. 5
4. GPCReG Protein Interaction.................................................................................... 7
5. Second Messenger Production .............................................................................. 11
6. b-Arrestin Recruitment ......................................................................................... 12
7. Receptor Internalization ....................................................................................... 15
8. Receptor Oligomerization...................................................................................... 16
9. Summary and Outlook........................................................................................... 23
Acknowledgments ..................................................................................................... 23
References ............................................................................................................... 23

Abstract
Since their discovery, G protein-coupled receptors (GPCRs) constitute one of the most
studied proteins leading to important discoveries and perspectives in terms of their biology
and implication in physiology and pathophysiology. This is mostly linked to the remarkable
advances in the development and application of the biophysical resonance energy transfer
(RET)-based approaches, including bioluminescence and fluorescence resonance energy
transfer (BRET and FRET, respectively). Indeed, BRET and FRET have been extensively
applied to study different aspects of GPCR functioning such as their activation and regulation either statically or dynamically, in real-time and intact cells. Consequently, our view
Methods in Cell Biology, Volume 132, ISSN 0091-679X, http://dx.doi.org/10.1016/bs.mcb.2015.10.008
2016 Elsevier Inc. All rights reserved.

ARTICLE IN PRESS

Resonance Energy Transfer

on GPCRs has considerably changed opening new challenges for the study of GPCRs in
their native tissues in the aim to get more knowledge on how these receptors control the
biological responses. Moreover, the technological aspect of this field of research promises
further developments for robust and reliable new RET-based assays that may be compatible
with high-throughput screening as well as drug discovery programs.

INTRODUCTION
G protein-coupled receptors (GPCRs) constitute one of the largest cell surface receptor families mediating cellular signaling and are thereby involved in many physiological responses and pathophysiological situations. GPCRs constitute the site of
action of a large panel of natural mediators such as hormones and neurotransmitters
(Bockaert & Philippe Pin, 1999; Katritch, Cherezov, & Stevens, 2013; Lagerstrom &
Schioth, 2008). Therefore, fundamental research and pharmaceutical companies
have dedicated a particular interest for GPCRs being the target of 30e50% drugs
used to treat human diseases (Lundstrom, 2006; Schlyer & Horuk, 2006; Williams
& Hill, 2009; Wise, Gearing, & Rees, 2002). At the cellular and molecular levels, the
functioning of GPCRs is essentially orchestrated by their physical interaction with
and activation of the key signaling proteins known as heterotrimeric G proteins
(Bockaert, Homburger, & Rouot, 1987; Bourne, 1997; Gether, 2000; Gilman,
1987; Limbird, 1983; Moreira, 2014; Strange, 2008a). The initial model based on
the ternary complex comprising the ligand, the receptor, and the heterotrimeric G
protein can be considered too simplistic when considering all the recent advances
documenting that GPCRs also activate G protein-independent signaling pathways
such as those mediated by b-arrestins, Src, and proteins with PDZ domains. Moreover, recently new challenging concepts have emerged such as receptor oligomerization, allostery, receptor-G protein preassembly, biased signaling, orphan receptors,
and transactivation with receptor tyrosine kinases (Brady & Limbird, 2002;
Fredholm, Hokfelt, & Milligan, 2007; Garland, 2013; Gomes et al., 2016; Hermans,
2003; Katritch et al., 2013; Rajagopal, Rajagopal, & Lefkowitz, 2010; Strange,
2008b; Wootten, Christopoulos, & Sexton, 2013). These advances have been principally achieved through considerable progresses in developing the technologies used
to study GPCRs, in terms of their structure and complexes as well as their activation
and regulation. Among these methods, resonance energy transfer (RET)-based
approaches (bioluminescence and fluorescence resonance energy transfer (BRET
and FRET)) have significantly contributed to the major advances around GPCRs
leading our view toward this family of receptors being significantly evolved. In
fact, GPCRs constitute the research field of reference where RET-based assays are
elegantly used and are the subject of continual development and improvement in
terms of methodology and instrumentation (Ayoub & Pfleger, 2010; Conn, 2013;
Cottet et al., 2012; Hein, Frank, Hoffmann, Lohse, & Bunemann, 2005; Lohse,
Nuber, & Hoffmann, 2012; Nguyen & Daugherty, 2005; Pfleger & Eidne, 2006;
Pin, Ayoub, Maurel, Perroy, & Trinquet, 2008).

ARTICLE IN PRESS

1. RET approaches

In this chapter, the applications of RET-based technologies to study the different


aspects of GPCR function and regulation are described. This covers the process of
the activation of GPCRs followed by their desensitization and regulation going from
ligand binding, receptor conformation changes, receptor-G protein coupling and receptor oligomerization, to b-arrestin recruitment and receptor internalization and
recycling. The potential applications of RET in GPCR drug discovery are also
briefly discussed.

1. RET APPROACHES
RET approaches link the biophysical concepts of spectral overlap, distance, and proximity, in space and time, between an energy donor and an energy acceptor to the biological question of interest according to Forsters law (Forster, 1948; Pin et al., 2008;
Selvin, 2000). Thus, the changes in the distance between and/or orientation of the
donoreacceptor pair within the individual protein structure (intramolecular) or
proteineprotein/proteinesmall molecule complexes (intermolecular) constitute
the basis of the utilization of RET to study proteins in terms of their association/
dissociation as well as their activation, regulation, and trafficking in the cells (Lohse,
Bunemann, Hoffmann, Vilardaga, & Nikolaev, 2007). This becomes possible thanks
to the development of various resonance energy donors and energy acceptors
compatible with multiple BRET and FRET configurations and assays with improved
spectral properties and increased sensitivities. This includes multiple variants of
green fluorescent proteins (GFPs) and a large array of fluorescent dyes conjugated
to protein fragments, enzyme substrates, chemicals, ligands, or antibodies. These
fluorescent dyes are commonly used together for FRET assays or specifically combined with either the rare earth europium and terbium cryptates or chelates for timeresolved FRET (TR-FRET) and homogeneous time-resolved fluorescence (HTRF)
or various luciferases (Luc) and their substrates for BRET assays. In addition, the
development in the chemistry of RET was accompanied by exponential technical
advances leading to robust instrumentations dedicated to measure RET signals
with high sensitivity and constant reproducibility. Together, theses advances led to
get more insights (quantitatively and qualitatively) into different aspects of molecular and cellular biology, both in spatial and temporal terms, as well as in individual
cells or whole tissues (De, Loening, & Gambhir, 2007; Dragulescu-Andrasi, Chan,
De, Massoud, & Gambhir, 2011; Xu et al., 2007). For GPCRs and their functioning,
the utilization of RET enables the assessment of ligandereceptor, receptoreprotein,
or receptorereceptor complexes in both static and dynamic manners, in real-time
and live cells. These different levels of interactions involving GPCRs are described
below including ligand binding, ligand-induced conformational changes, homo- and
heteromerization, coupling with the heterotrimeric G proteins, modulation of intracellular second messengers, recruitment of b-arrestins, and receptor internalization
and recycling (Lohse et al., 2012; Salahpour et al., 2012). Thus, such advances in
RET-based technologies and their experimental advantages (flexibility, rapidity,

ARTICLE IN PRESS

Resonance Energy Transfer

miniaturization, etc.) open new perspectives in terms of their application in highthroughput screening (HTS) and drug discovery programs for the development of
new GPCR ligands and drugs (Cottet, Faklaris, et al., 2011; Degorce et al., 2009;
Heding, 2004; Milligan, 2004a; Robinson, Yang, & Zhang, 2014; Roda, Guardigli,
Pasini, & Mirasoli, 2003; Zwier et al., 2010).

2. LIGAND BINDING
Since the binding of a ligand on its specific receptor constitutes the first step in the
activation of GPCRs and their downstream signaling, the pharmacological characterization of any GPCR as well as the discovery of potential new ligands involves
the analysis of its ligand-binding properties. Thus, the utilization of highly selective
radioligands targeting GPCRs was the assay of choice for many years. However,
there are growing limitations on using radioactivity in the research laboratories
due to the issues of safety and disposal of radioactive waste issues. Therefore, there
is particular interest in the development of alternative nonradioactive approaches for
ligand binding, and consequently RET-based assays have emerged by labeling the
ligands and/or the receptors with luminescence and fluorescence dyes instead of
incorporation of classical isotopes (Cottet, Faklaris, et al., 2011; Stoddart,
Johnstone, et al., 2015; Stoddart, White, Nguyen, Hill, & Pfleger, 2015; Vernall,
Hill, & Kellam, 2013; Zwier et al., 2010). Indeed, many fluorescent GPCR-selective
ligands compatible with RET-based assays have been synthesized (Stoddart,
Johnstone, et al., 2015; Stoddart, White, et al., 2015; Vernall et al., 2013; Zwier
et al., 2010). Consequently, RET-based assays can be used as a readout of ligand
binding, using a RET compatible donoreacceptor pair between the ligand and its
receptor by assessing either a direct binding of the fluorescently labeled ligand to
the receptor or competition of the binding of the fluorescently labeled ligand by
another unlabeled ligand as illustrated in Figure 1.
Early during the emergence of FRET, the possibility of using FRET to investigate ligand binding of cell surface receptors was reported (Louie & Tromberg,
1998). For GPCRs, one of the pioneer RET-based binding assay works was conducted on the muscarinic M1 receptor (M1R) using GFP-fused receptor and nonfluorescent ligands using FRET quenching (Tahtaoui et al., 2005). In this study, binding
properties (Kd) and kinetic parameters (Kon/Koff) of ligand binding were elegantly
determined for some M1R ligands (Tahtaoui et al., 2005). In addition, a homogeneous ligand-binding assay based on TR-FRET was reported on human complement
5a receptor (C5aR) tagged with hemagglutinin (HA) epitope at the N-terminus (Hu,
Zhou, Hamman, & Liu, 2008). In this study, C5a binding was assessed by measuring
TR-FRET signal between C5a labeled with terbium chelate (energy donor) and
HA-C5aR labeled with anti-HA antibody conjugated with Alexa Fluor-488 (energy
acceptor) (Hu et al., 2008). Later, other RET-based ligand-binding assays using
various FRET configurations have been reported on other GPCRs including
b2-adrenergic (b2AR) (Martikkala et al., 2009), cannabinoid CB2 (Sexton et al.,

ARTICLE IN PRESS

3. Ligand-Induced conformational changes

RET
D

D
RET Signal

(A)
L

Binding of
fluorescent ligand

Ligand Concentration

RET

RET decrease

Competition of binding
of fluorescent ligand

RET Signal

(B)

Ligand Concentration

FIGURE 1 LigandeG protein-coupled receptor interaction assessed by resonance energy


transfer (RET)-based assays.
(A) RET signals are measured between the energy donor (D)-fused receptor (R) and the
energy acceptor (A)-conjugated ligand (L). Saturation binding curves are generated by
increasing the concentration of the ligand allowing the determination of Kd values of the
fluorescent ligand. (B) Competition of the RET signals measured between the donor-fused
receptor (DeR) and the acceptor-conjugated ligand (AeL) by an unlabeled ligand (L0 ). RET
decrease is plotted as function of the increasing doses of L0 allowing the determination of Ki
values of the unlabeled ligand.

2011), vasopressin/oxytocin (Albizu et al., 2010), adenosine A3 (Stoddart et al.,


2012), and dopamine D1/3 (Hounsou et al., 2015) receptors. In most studies, RET
data were very consistent with those obtained using the classical radioligand-binding
method. The first study reporting ligandeGPCR interactions using BRET has just
been reported by Stoddart, Johnstone, et al. (2015) where a new generation of
BRET donor (luciferase), NanoLuc, was fused to the N-terminus of various GPCRs
including b2AR, adenosine A1/3, and angiotensin AT1 receptors. The ligand-binding
properties (Kon/Koff, Kd, and Ki) were determined by measuring the BRET increase
between the NanoLuc-tagged receptor and its selective ligand labeled with redshifted probes for the first time at the cell surface (Stoddart, Johnstone, et al.,
2015; Stoddart, White, et al., 2015).

3. LIGAND-INDUCED CONFORMATIONAL CHANGES


As a consequence of ligand binding, conformational changes of the different domains
within the receptor constitute an important aspect of research on GPCRs. To gain insights into structural and conformational modifications upon GPCR activation

ARTICLE IN PRESS

Resonance Energy Transfer

fluorescence- and luminescence-based techniques have been extensively used. The


first studies were based on fluorescence lifetime analysis on purified receptors using
covalent fluorescence labeling of single cysteine residues localized in different environments of the receptor protein. This revealed the existence of different conformations of the b2AR whether the receptor was bound or not to its agonist (Gether, Lin, &
Kobilka, 1995; Ghanouni, Gryczynski, et al., 2001; Ghanouni, Steenhuis, et al.,
2001; Neumann, Wohland, Whelan, Zare, & Kobilka, 2002). Moreover, the changes
in the intrinsic fluorescence of tryptophan residues or covalently incorporated probes
were also used in many occasions as readout of ligandereceptor interaction
(Damian, Mary, Martin, Pin, & Baneres, 2008; El Moustaine et al., 2012; Rahmeh
et al., 2012). Subsequently, RET-based approaches have been adapted to develop
various conformational sensors of GPCRs expressed in cells taking advantage of
the biophysical properties of these techniques based on the changes in the distance
and/or orientation of the energy donor and energy acceptor moieties (Zhang,
Campbell, Ting, & Tsien, 2002). The RET-based assays introduce both the energy
donor and acceptor partners into the same receptor protein as illustrated in
Figure 2(A) to assess intramolecular changes in RET signals as a consequence of
ligand-promoted conformational changes in the receptor. Of course, GPCRs constitute the model of choice for the application of such approaches due to their structure
of seven transmembrane domains linked by three extracellular and three intracellular
loops and ended by a relatively flexible C-terminal tail. Thus, ligand binding is
thought to affect dramatically such surface and molecular organization leading to
receptor activation and its interaction and coupling with the heterotrimeric G proteins as well as other partners such as GPCR kinases (GRKs) and b-arrestins. The
most important studies concern those reported on the a1-adrenergic (a1AR)
(Vilardaga, Bunemann, Krasel, Castro, & Lohse, 2003; Zurn et al., 2009), b2AR
(Granier, Kim, Fung, Bokoch, & Parnot, 2009), parathyroid hormone (PTHR)
(Castro, Nikolaev, Palm, Lohse, & Vilardaga, 2005; Vilardaga et al., 2003), and
adenosine A1 (Hoffmann et al., 2005) receptors using FRET, as well as the
ODR-10 GPCR in Caenorhabditis elegans studied by BRET upon its activation
by femtomolar levels of volatile ligands (Dacres et al., 2011). The study on PTH
revealed a two-step ligand-binding process with a very fast first phase (milliseconds)
followed by a slow second one (seconds) (Castro et al., 2005; Vilardaga et al., 2003).
Interestingly, the RET-based approaches have also been applied in the context of
GPCR homo- and heterodimers where each energy partner is fused to one protomer
at a specific domain within the dimer (Figure 2(B)). Indeed, in addition to their use
to reveal receptor dimerization (see below), RET was also used to detect conformational changes of GPCR dimers upon ligand binding. This is the case with FRET on
a1AR/m-opioid heterodimers (Vilardaga et al., 2008) and mGluR1 homodimers
(Tateyama, Abe, Nakata, Saito, & Kubo, 2004), as well as HTRF technology applied
to mGluR2 homodimers (Doumazane et al., 2013). Furthermore, BRET has been
used to study ligand-induced conformational changes within melatonin (MT1R/
MT2R) (Ayoub et al., 2002; Ayoub, Levoye, Delagrange, & Jockers, 2004; Journe
et al., 2014), vasopressin V2 (V2R) (Granier et al., 2004), and chemokine

ARTICLE IN PRESS

4. GPCReG protein interaction

R
L

Ligand-Induced
conformational changes
within the monomer

RET Signal

(A)

D
Low RET

Basal RET
D

Dimer

RET decrease

A
L

Ligand-Induced
conformational changes
within the dimer

RET Signal

(B)

Ligand Concentration

RET increase

Ligand Concentration

FIGURE 2 Detection of ligand-induced conformational changes on G protein-coupled receptors


(GPCRs) using resonance energy transfer (RET)-based assays.
(A) In the case of GPCR monomers, both donor and acceptor are fused to the receptor at
different positions (here C-terminus and the third intracellular loop), and changes in the
intramolecular RET signals (here shown as an increase) are measured upon ligand binding.
Sigmoidal doseeresponse curves are generated by increasing the concentrations of the
ligand allowing the determination of EC50 values reflecting the potency of the ligand to
activate and induce conformational changes on the receptor. (B) In the case of GPCR dimers,
the donor and acceptor are fused to two different protomers (here shown at the N-terminus),
and changes in the intermolecular RET signals (here shown as a decrease) are measured
upon ligand binding. Sigmoidal doseeresponse curves are generated by increasing the
concentrations of the ligand allowing the determination of EC50 values reflecting the potency
of the ligand to induce conformational changes within the receptor dimer.

CXCR4 (Percherancier et al., 2005) receptor homo- and heterodimers. In these


studies, RET signals specifically measured from dimer species have been shown
to either increase or decrease upon ligand binding allowing the assessment of
ligand-promoted conformational changes on the dimers. In addition, these assays
have been shown to be useful for the profiling of various agonist and antagonist
ligands acting on heterodimers versus homodimers as shown for MT1R/MT2R heterodimers (Ayoub et al., 2004).

4. GPCReG PROTEIN INTERACTION


The question on how GPCReG protein coupling occurs has been widely studied
over the years. The initial model explaining the functioning of the GPCReG protein
complex has evolved considerably (Bourne, 1997; Limbird, 2004; Moreira, 2014;
Strange, 2008b) and new concepts have emerged such as constitutive activity

ARTICLE IN PRESS

Resonance Energy Transfer

(Seifert & Wenzel-Seifert, 2002), precoupling and/or preassembly (Ayoub et al.,


2007; Ayoub, Trinquet, Pfleger, & Pin, 2010; Gales et al., 2006; Leff & Scaramellini,
1998; Nobles, Benians, & Tinker, 2005; Qin, Dong, Wu, & Lambert, 2011), multiple
coupling (Hamm, 1998; Hermans, 2003; Simon, Strathmann, & Gautam, 1991),
functional selectivity (Kenakin, 2012; Rajagopal et al., 2010; Urban et al., 2007),
and intracellular G protein activation (Irannejad et al., 2013; Koelle, 2006). The
physical interaction and the functional coupling of GPCRs with their cognate heterotrimeric G proteins were initially studied using the GTPgS-binding assay
(Senogles et al., 1987; Stadel, Shorr, Limbird, & Lefkowitz, 1981; Tian, Duzic,
Lanier, & Deth, 1994) and biochemical techniques (Neumann et al., 2002; Smith
& Limbird, 1981), and recently through crystallographic analysis (Kobilka &
Schertler, 2008; Moreira, 2014; Rasmussen et al., 2011). However, the detailed analysis of the activation/deactivation processes of GPCReG protein complexes in realtime and in live cells was very limited for a long time, until the development of RET
techniques (Ayoub, Al-Senaidy, & Pin, 2012; Hein et al., 2005; Lohse et al., 2012;
Pin et al., 2008; Vilardaga et al., 2009). RET-based assays enable assessment of the
intimate changes (increase or decrease) in the proximity between GPCRs and G proteins and/or their conformational modifications upon activation. For this, molecular
fusions with energy donors and acceptors are fused to the receptors (mostly at their
C-terminus) and/or the different G protein subunits (Ga and Gbg). These fusion proteins are then transiently coexpressed, and measurements of RET signals carried
out, in real-time and live cells, before and upon stimulation with the GPCRselective agonist of interest as described in Figure 3 and previously described
(Ayoub, Al-Senaidy, et al., 2012). The first studies were based on FRET measurements between the different Ga and Gbg subunits upon GPCR activation using
different variants of GFPs as FRET donors and acceptors. Indeed, the pioneer
work was reported in Dictyostelium discoideum measuring in real time the dissociation between Ga and Gbg directly in live cells upon chemoattractant application
(Janetopoulos & Devreotes, 2002; Janetopoulos, Jin, & Devreotes, 2001). Then
the following RET studies on the assessment of G protein activation through the
measurements of BRET and FRET signals between the Ga and Gbg subunits
were reported in various mammalian cell lines and yeast coexpressing unmodified
GPCRs (Figure 3(A)) (Ayoub et al., 2009; Ayoub, Landomiel, et al., 2015; Azpiazu
& Gautam, 2004; Bellot et al., 2015; Digby, Lober, Sethi, & Lambert, 2006; Gibson
& Gilman, 2006; Yi, Kitano, & Simon, 2003). As a result, the idea of nondissociation of the Gabg trimer upon GPCR activation was proposed and supported by the
evidence that such activation induces conformational changes of the complexes that
translate into either an increase or decrease in RET signals between G protein subunits (Ayoub, Al-Senaidy, et al., 2012; Azpiazu & Gautam, 2004; Breton, Lagace, &
Bouvier, 2010; Digby et al., 2006; Gales et al., 2006; Gibson & Gilman, 2006;
Yi et al., 2003). The next advances in the application of RET to investigate
GPCReG protein interaction and coupling are characterized by the ability to measure FRET and BRET signals directly between the GPCRs themselves and their
cognate G proteins (Ga and Gbg) as well as the dynamics of their complexes

ARTICLE IN PRESS

4. GPCReG protein interaction

(A)

L
L
G G/
D

Dissociation
of G protein complex

G/

RET decrease

RET

(B)

L
L
G protein

Formation of
R/G protein complex

G protein

D
RET

(C)

L
L

G protein

Low RET

Conformational changes
within R/G protein complex

D
RET increase

FIGURE 3 G protein-coupled receptor (GPCR)eG protein interaction assessed by resonance


energy transfer (RET)-based assays.
(A) The changes in the basal intermolecular RET signals (here shown as a decrease)
measured between the different G protein subunits fused to the donor (Ga-D) and receptor
(Gb/g-A) are assessed as a consequence of their physical dissociation upon ligand-induced
activation of the unmodified GPCR. (B) The increase in the intermolecular RET signals
between the donor-fused receptor (ReD) and the acceptor-fused G protein (either a, b, or
g subunits) (G protein-A) is measured upon ligand binding and GPCR activation. (C) The
changes in the basal RET signals (here shown as an increase) measured between the donorfused receptor (ReD) and the acceptor-fused G protein (G protein-A), either a, b, or g
subunits, are assessed reflecting the ligand-induced conformational changes within the
preassembled GPCReG protein complexes.

(Figure 3(B) and (C)) (Ayoub, Al-Senaidy, et al., 2012; Ayoub et al., 2007, 2010;
Ayoub, Zhang, et al., 2015; Bellot et al., 2015; Gales et al., 2005, 2006; Hasbi
et al., 2007; Hein et al., 2005; Nobles et al., 2005; Qin et al., 2011). Real-time
kinetics of the activation of GPCReG protein complexes were reported in live cells

ARTICLE IN PRESS

10

Resonance Energy Transfer

showing a rapid and transient activation in some cases and slow and stable physical
association in others as previously discussed (Ayoub, Al-Senaidy, et al., 2012; Lohse
et al., 2012). Moreover, constitutive receptoreG protein association was reported in
many situations while agonist-induced interaction was shown for some GPCReG
protein pairs (Figure 3(C)) (Ayoub, Al-Senaidy, et al., 2012; Lohse et al., 2012).
Consequently, the RET-based studies opened an interesting debate on the physical
and functional GPCReG protein interaction and its dynamics analyzed by RETbased approaches challenging the classical model of agonist-inducing GPCReG
protein interaction and coupling (Ayoub, Al-Senaidy, et al., 2012; Bourne, 1997;
Leff, 1995; Strange, 2008b). Indeed, the model of free collision is contrary
to the concepts of precoupling and preassembly (Ayoub, Al-Senaidy, et al.,
2012; Ayoub et al., 2007, 2010; Gales et al., 2005, 2006; Hasbi et al., 2007; Hein
et al., 2005; Nobles et al., 2005; Philip, Sengupta, & Scarlata, 2007; Qin et al.,
2011). In this context, some studies reported an increase in BRET or FRET signals
between the receptors and G proteins exclusively upon agonist stimulation as shown
for a2AR-G1/g2 (Kuravi, Lan, Barik, & Lambert, 2010), a1AR-Gai (Hein et al.,
2005), ghrelin-Gaq (Damian et al., 2015), and protease-activated receptor
(PAR1/2)-Ga12 complexes (Ayoub & Pin, 2013; Ayoub et al., 2010). However, in
other situations significant and specific basal RET signals between the receptors
and G proteins were measured even in the absence of receptor activation as shown
for a1AR (Nobles et al., 2005), adenosine A2 (Nobles et al., 2005), muscarinic M3
(Qin et al., 2011), thrombin PAR1 (Ayoub, Al-Senaidy, et al., 2012; Ayoub et al.,
2007, 2010), trypsin PAR2 (Ayoub & Pin, 2013), a2AR (Breton et al., 2010; Gales
et al., 2005, 2006; Nobles et al., 2005), d-opioid (Audet et al., 2008), CXCR4 and
CXCR7 (Levoye, Balabanian, Baleux, Bachelerie, & Lagane, 2009), ghrelin
(Damian et al., 2015), and bradykinin (Philip et al., 2007) receptors and their
different cognate G proteins. Receptor activation was shown to either increase or
decrease the basal RET signals between the GPCRs and their cognate G proteins.
In some cases, the basal RET signals measured were correlated to the constitutive
activity of the receptors as shown for the preassembly between the ghrelin receptor
and Gaq protein (Damian et al., 2015). However, the basal RET signals measured
from the preassembled GPCReG protein complexes were mostly independent of
any constitutive activity of the receptors or basal activation of the G proteins studied.
For instance, treatments with GPCR-selective antagonists as well as pertussis toxin
(on receptoreGai/o complexes) have been shown to completely abolish the agonistinduced BRET increase between the receptor and Ga subunit, while such treatments
had no effect on the basal BRET signals (Ayoub et al., 2007, 2010; Gales et al.,
2006). Moreover, the agonist-promoted RET changes have been shown to be sensitive to the position where the energy donor and acceptor were inserted into the receptor and/or the G protein (Ayoub et al., 2007, 2010; Gales et al., 2006).
Together, these observations support the notion of agonist-induced conformational
changes within the preassembled GPCReG protein complexes as previously discussed (Ayoub, Al-Senaidy, et al., 2012; Lohse et al., 2012). These observations
are consistent with the old biochemical studies reporting the stable association

ARTICLE IN PRESS

5. Second messenger production

between various GPCRs and G proteins independently of the activation state of the
receptors (Lachance, Ethier, Wolbring, Schnetkamp, & Hebert, 1999; Roka et al.,
1999; Seifert & Wenzel-Seifert, 2002; Senogles et al., 1987). The discordances
raised regarding the preassembly or not between GPCRs and G proteins may depend
on the cellular system and the RET-based assay used. More importantly, the preassembly may constitute a mechanism of regulation of the multiple coupling and
signaling of GPCRs with the occurrence and circumstances of preassembly depending on the Ga protein considered. Indeed, PAR1 has been shown to form preassembled complexes with Gai1/o but not Ga12 (Ayoub et al., 2009, 2010), and ghrelin
receptor has been reported to preassemble with Gaq but not Gai (Damian et al.,
2015), where both assembly was assessed by BRET and FRET techniques. Similarly,
a given G protein may have different association modes with different GPCRs as
nicely demonstrated for Ga12 which interacted with PAR1 and PAR2 in an
agonist-dependent manner while being able to exist in preassembled complexes
with other GPCRs such as vasopressin V1a and muscarinic M3 receptors (Ayoub
& Pin, 2013; Ayoub et al., 2010). Therefore, the population of PAR1/2 preassembled
with Gai1 may not be able to couple with Ga12 and vice versa. Moreover, the desensitization properties may be different depending upon whether the receptor is preassembled or not as shown with PAR1, where the preassembled PAR1-Gai1 complexes
recruit b-arrestins but the induced PAR1-Ga12 complexes did not. In native tissues,
the preassembly or not between GPCRs and their cognate G proteins may be determined by their expression levels and compartmentalization in the cells, as well as
their interactions with other membrane or cytosolic partners such GPCR oligomerization. Conceptually, these RET-based studies challenge the classical dogma of
GPCReG protein coupling and revive the interesting debate regarding the nature
as well as the properties of such molecular interaction and its role in GPCR signaling
and regulation. The concept of preassembly raises intriguing questions regarding
GPCR functioning and illustrates the complexity of GPCReG protein coupling,
signaling, and regulation. Therefore, further investigations on this aspect should
consider other new concepts such as GPCR heterodimerization and their biased
signaling, with the aim to generating new evidence for the reality of such preassembly
and its impact on physiology and pathophysiology involving GPCRs.

5. SECOND MESSENGER PRODUCTION


The development of RET-based assays to investigate GPCR signaling constitutes
another important advance in the field, being used as alternative approaches to the
classical biochemical and radioactive-based assays. Indeed, many BRET and FRET
assays and sensors have been developed to assess the production of intracellular
second messengers, such as cyclic adenosine monophosphate (cAMP), inositol
phosphate (IP), and calcium (Ca2), and to detect the activation of important downstream signaling proteins such as the extracellular signal-regulated kinases (ERK1/2)
(Klarenbeek & Jalink, 2014; Robinson et al., 2014; Salahpour et al., 2012; Xu et al.,

11

ARTICLE IN PRESS

12

Resonance Energy Transfer

2013; Zhang et al., 2002). The first category of these assays comprises the molecular
RET sensors where the second messenger sensor is genetically double fused to energy donor and acceptor (either BRET or FRET), and transfected in cells, and intramolecular RET changes (increase or decrease) are then assessed in real-time and live
cells as a consequence of the changes in the intracellular concentrations and/or binding of the second messenger on the sensor following GPCR activation (Figure 4(A)).
This has been mostly used for the measurement of cAMP by FRET (DiPilato, Cheng,
& Zhang, 2004; Klarenbeek, Goedhart, van Batenburg, Groenewald, & Jalink, 2015;
Klarenbeek & Jalink, 2014) and BRET (Ayoub, Landomiel, et al., 2015; Barak et al.,
2008; Comps-Agrar, Kniazeff, Brock, Trinquet, & Pin, 2012; Jiang et al., 2007;
Monnier et al., 2011), Ca2 release (Ayoub, Landomiel, et al., 2015; Gorokhovatsky
et al., 2004; Naumann, Kampff, Prober, Schier, & Engert, 2010; Whitaker, 2010),
ERK1/2 phosphorylation (Harvey et al., 2008; Xu et al., 2013), and Rho-GTPase
activation (Nakamura, Aoki, & Matsuda, 2005; Nakamura, Kurokawa, Kiyokawa,
& Matsuda, 2006). The second category of RET assays is mostly based on HTRF
technology, where intermolecular FRET signals are measured between energy donor
and acceptor using various combinations of fluorescently labeled antibodies and/or
purified second messengers compatible with HTRF technology (Figure 4(B))
(Degorce et al., 2009; Gabriel et al., 2003; Norskov-Lauritsen, Thomsen, &
Brauner-Osborne, 2014; Pin et al., 2008). Thus, for cAMP and IP1 assays the
changes in the production of second messengers upon GPCR activation results in
the variation of the amplitude of RET signals between the purified donor- and
acceptor-conjugated antibodies and reagents added into the cell samples. In contrast,
ERK1/2 activation is assessed by an increase of the RET signals between the donorand acceptor-conjugated antibodies recognizing different motifs of the active
ERK1/2 as described in Figure 4(B) (Ayoub et al., 2014). Thus, various HTRF-based
kits have been developed and commercialized by CisBio Bioassays company for the
investigation of GPCR signaling in terms of cAMP (Armstrong, Seeber, Ayoub,
Feldman, & Pfleger, 2013; Ayoub et al., 2009; Ayoub, Landomiel, et al., 2015;
Ayoub et al., 2007; Norskov-Lauritsen et al., 2014), IP (Ayoub, Angelicheva,
et al., 2012; Ayoub et al., 2009; Trinquet, Bouhelal, & Dietz, 2011; Trinquet
et al., 2006), and ERK1/2 signaling pathways (Ayoub et al., 2014; Jia, Quinn, Gagnon, & Talanian, 2006). These assays are now widely used and many other scientific
publications reported their utilization to study GPCR pharmacology and signaling.
The rapidity of the RET-based assays and their miniaturization in 96-, 384-, and
1536-well plates constitute the big asset for their application in both fundamental
research and HTS programs to screen new ligands and pharmacologically profile
GPCRs and their multiple signaling pathways.

6. b-ARRESTIN RECRUITMENT
It is well accepted that b-arrestins play a major role in not only GPCR desensitization and internalization but also their signaling (Carman & Benovic, 1998; DeWire,

ARTICLE IN PRESS

6. b-Arrestin recruitment

(A)

L
L
G protein

Formation and activation


of R/G protein complex

Binding to the sensor

Intracellular production
of second messengers
(cAMP, Ca2+)

Sensor

G protein

Sensor

RET decrease

Basal RET

(B)

L
L
G protein

Formation and activation


of R/G protein complex

G protein

cAMP or IP1

Basal RET

RET
decrease

pERK1/2

Intracellular production
of second messengers

RET

FIGURE 4 G protein-coupled receptor (GPCR)-induced modulation of intracellular second


messengers assessed by resonance energy transfer (RET)-based assays.
(A) In the case of a cytoplasmic sensor for intracellular second messengers (cAMP, Ca2,
etc.), both donor and acceptor are fused the molecular sensor at different positions (mostly
N- and C-terminus), and changes in the intramolecular RET signals (here shown as a
decrease) are measured as a consequence of changes in the cytoplasmic concentrations of
the second messenger upon ligand binding and GPCReG protein activation. (B) For the
assessment of intracellular concentrations of the second messengers (cAMP, IP1, pERK1/2,
etc.), specific antibodies recognizing the different forms of the second messengers and
conjugated to donor and acceptor are used. The changes in the cytoplasmic concentrations
of the second messenger upon GPCR activation is measured either par competition (for
cAMP and IP1) or increase (pERK1/2) of the RET signals between the two donor- and
acceptor-conjugated antibodies.

Ahn, Lefkowitz, & Shenoy, 2007; Lefkowitz & Shenoy, 2005; Luttrell & Lefkowitz,
2002; Reiter & Lefkowitz, 2006). Therefore, the study of the physical association
between GPCRs and b-arrestins constitutes an important field of research and investigation both in vitro and in vivo. One of the important features of the physical

13

ARTICLE IN PRESS

14

Resonance Energy Transfer

interaction between GPCRs and b-arrestins is of course its temporal and spatial
dynamic, since the recruitment of the cytosolic pools of b-arrestins to the plasma
membrane receptors is thought to be initiated by receptor activation and mostly
mediated through receptor phosphorylation by specific GRKs at the receptor
C-terminus (Gurevich & Gurevich, 2004; Reiter & Lefkowitz, 2006; Shenoy &
Lefkowitz, 2011). The first fluorescence-based assays to investigate GPCRebarrestin association were based on confocal microscopy using GFP-fused b-arrestins
allowing the assessment of their membrane translocation upon GPCR activation
(Barak, Ferguson, Zhang, & Caron, 1997; Evans et al., 2001; Ferguson & Caron,
2004; Heding et al., 2000; Holliday, Holst, Rodionova, Schwartz, & Cox, 2007;
Laporte, Oakley, Holt, Barak, & Caron, 2000; Lin & Trejo, 2013; Shenoy &
Lefkowitz, 2003). However, the major advances in BRET and FRET technologies
to study GPCRs also covered b-arrestins as key components in GPCR signaling
and regulation (Lohse et al., 2012). Thus, various RET-based assays have been
developed to monitor GPCR-promoted b-arrestin recruitment in real-time and live
cells using either BRET (Heding, 2004; Kaczor et al., 2014; Kamal et al., 2009;
Kocan, Dalrymple, Seeber, Feldman, & Pfleger, 2010; Kocan & Pfleger, 2009;
Pfleger, Dalrymple, Dromey, & Eidne, 2007; Vrecl, Jorgensen, Pogacnik, & Heding,
2004), HTRF (Ayoub et al., 2010), or FRET (Krasel, Bunemann, Lorenz, & Lohse,
2005; Violin, Ren, & Lefkowitz, 2006; Wehbi et al., 2013) technologies. The most
used approaches consist of the fusion of both the receptor and b-arrestin to energy
donor and acceptor, and the increase of intermolecular RET signals reflecting
b-arrestin recruitment is measured in real-time and live cells upon receptor activation (Figure 5(A)). These different assays were used in different contexts to assess
agonist-induced b-arrestin recruitment mediated by many GPCRs including neuropeptide Y (Berglund, Schober, Statnick, McDonald, & Gehlert, 2003), PAR1/2
(Ayoub et al., 2009; Ayoub et al., 2007; Ayoub & Pin, 2013; Ayoub et al., 2010),
follicle-stimulating hormone (FSHR) and luteinizing hormone (LHR) (Ayoub,
Landomiel, et al., 2015), vasopressin type 2 (V2R) (Armstrong et al., 2013; Bertrand
et al., 2002; Kocan et al., 2009; Tenenbaum et al., 2009), b2AR (Krasel et al., 2005;
Vrecl et al., 2004), neurokinin type 1 (NK1R) (Vrecl et al., 2004), chemokine
(CCR5, CXCR4, and CXCR2) (See, Seeber, Kocan, Eidne, & Pfleger, 2011),
CCR2 (Ayoub, Zhang, et al., 2015), angiotensin II type 1 (AT1R) (Ayoub, Zhang,
et al., 2015; Porrello et al., 2011), orexin (OX1/2R) (Dalrymple, Jaeger, Eidne, &
Pfleger, 2011; Jaeger, Seeber, Eidne, & Pfleger, 2014), bradykinin (See et al.,
2011), melatonin (MT1) (Levoye, Dam, Ayoub, Guillaume, Couturier, et al.,
2006), glucagon-like-1 (GLP-1R) (Jorgensen, Martini, Schwartz, & Elling, 2005),
and PTHR (Wehbi et al., 2013) receptors. Alternatively, another RET-based
approach to assess b-arrestin recruitment has been used which measures intramolecular BRET changes (increase or decrease) within a b-arrestin double brilliance,
where both energy donor and acceptor are genetically fused to b-arrestin (Charest,
Terrillon, & Bouvier, 2005). In this configuration, the b-arrestin sensor is coexpressed with the unmodified GPCR of interest, and agonist-induced b-arrestin
recruitment is then assessed as changes in the conformation of b-arrestin following

ARTICLE IN PRESS

7. Receptor internalization

(A)

L
L
Formation
of R/-arrestin complex

-arrestin

D
RET

-arrestin

(B)

L
L
Formation
of R/-arrestin complex

-arrestin

-arrestin

RET decrease
Conformational changes
within R/-arrestin complex

Basal RET
FIGURE 5 G protein-coupled receptor (GPCR)earrestin interaction assessed by resonance
energy transfer (RET)-based assays.
(A) The increase in the intermolecular RET signals between the donor-fused receptor (ReD)
and acceptor-fused b-arrestins (b-arrestin-A) is measured upon ligand binding and GPCR
activation. (B) In the case of a cytoplasmic b-arrestin sensor (double brilliance), both donor
and acceptor are fused the molecular sensor at different positions (N- and C-terminus), and
the changes in the intramolecular RET signals (here shown as a decrease) are measured
reflecting the plasma membrane translocation of b-arrestin sensor upon GPCR activation.

its association with the activated receptor (Figure 5(B)). This assay has been reported for the association of b-arrestin with various GPCRs such as V2R, d-opioid,
CCR5, and AT1R (Charest et al., 2005; Shukla et al., 2008).

7. RECEPTOR INTERNALIZATION
Following their activation, GPCRs are known to undergo desensitization followed
by the internalization process in clathrin-coated pits as a consequence of receptor

15

ARTICLE IN PRESS

16

Resonance Energy Transfer

phosphorylation by specific GRKs and their association with b-arrestins (Carman &
Benovic, 1998; DeWire et al., 2007; Lefkowitz & Shenoy, 2005; Luttrell &
Lefkowitz, 2002; Reiter & Lefkowitz, 2006). Like for b-arrestin recruitment the classical approaches to assess such GPCR trafficking are based on confocal microcopy
and cell surface ELISA using fluorescently fused or labeled receptors. More recently,
RET-based assays have been proposed as alternative approaches to monitor
GPCR internalization in real-time and live cells and miniaturized formats (96- and
384-well plates) compatible with HTS using astute adaptation of both BRET and
TR-FRET techniques. BRET-based assays consist of the assessment of the agonistpromoted changes in the basal BRET signal specifically measured at the intracellular
face of the plasma membrane between the C-terminally energy donor-fused receptor
(GPCR-Luc) and an energy acceptor-fused membrane marker (GFP-KRas protein)
(Figure 6(A)). In this configuration, the vicinity between the two partners is significantly decreased upon agonist activation due to the reduction in receptors at the
plasma membrane, which is then measured as a decrease in the basal BRET signal
between GPCR-Luc and GFP-KRas as described in Figure 6(A). This assay has
been successfully applied to investigate the internalization of many GPCRs
including b2AR (Lan, Kuravi, & Lambert, 2011), OX2R (Jaeger, Seeber, et al.,
2014), bile acid receptor (TGR5R) (Jensen et al., 2013), and FSHR and LHR
(Ayoub, Landomiel, et al., 2015). The second RET-based assay for GPCR internalization uses TR-FRET technology where the receptor of interest is N-terminally
fused to a relatively small fluorescent protein and labeled with specific dyes compatible with the extracellular TR-FRET process with free-energy acceptor molecules
added to the cells (Figure 6(B)). This assay has been elegantly validated on
OX1R and cannabinoid CB1 (Ward, Pediani, & Milligan, 2011), vasopressin V1a
(Faklaris et al., 2015) as well as GLP-1R (Roed et al., 2015, 2014) receptors.

8. RECEPTOR OLIGOMERIZATION
Since the discovery of GPCRs, their oligomerization constitutes undoubtedly one of
the major concepts that has profoundly impacted our view on this family of membrane proteins. Indeed, early literature in 1980s and 1990s proposed the concept
of receptorereceptor interaction as a mechanism of integration of GPCR signaling
in the central nervous system (Agnati, Ferre, Lluis, Franco, & Fuxe, 2003; Fuxe &
Agnati, 1985; Fuxe, Marcellino, Guidolin, Woods, & Agnati, 2008). From the
2000s, a particular interest has been given to the study of the oligomerization of
GPCRs and its impact on their functioning. Consequently, over the years interesting
findings have been obtained regarding the role of such organization in the maturation, activation, G protein coupling, downstream signaling, and regulation of GPCRs
(Angers, Salahpour, & Bouvier, 2002; Bai, 2004; Bouvier, 2001; Bulenger, Marullo,
& Bouvier, 2005; Devi, 2001; Ferre et al., 2009; Gomes et al., 2016; Jordan &
Devi, 1999; Milligan, 2004b; Prinster, Hague, & Hall, 2005; Szidonya, Cserzo, &
Hunyady, 2008; Terrillon & Bouvier, 2004b; Vischer, Castro, & Pin, 2015).

ARTICLE IN PRESS

8. Receptor oligomerization

(A)

L
Protein X

Ligand - induced
receptor internalization

Protein X

A
Endosome

Basal RET

RET decrease

(B)
Basal RET

A
D

Free RET acceptor

A
A

Ligand - induced
receptor internalization

RET decrease

Endosome

FIGURE 6 G protein-coupled receptor (GPCR) internalization assessed by resonance energy


transfer (RET)-based assays.
(A) The decrease in the intermolecular basal RET signals between the donor-fused receptor
(ReD) and a plasma membrane protein marker (Protein X) fused to the acceptor (Protein X-A) is
measured as a consequence of GPCR internalization upon its activation. (B) The decrease in the
intermolecular basal RET signals between the donor-fused receptor (ReD) and free acceptor
(A) fluorescent molecules is measured as readout of GPCR internalization upon activation.

The major advances in the field concern the heterodimerization between the different
subfamilies of GPCRs including the orphan receptors and its impact on the functioning of GPCRs, as well as its implication in diverse physiological and pathophysiological models (Devi, 2001; Ferre et al., 2009; Gomes et al., 2016; Gomes, Gupta,
& Devi, 2013; Jordan & Devi, 1999; Levoye, Dam, Ayoub, Guillaume, & Jockers,
2006; Prinster et al., 2005; Vischer et al., 2015). Therefore, heterodimerization would
constitute one of the mechanisms of diversifying and finely regulating GPCR
signaling as multiple signaling pathways are possible through a limited number of
receptors expressed within a given organism. Importantly, the reality of oligomers
in native tissues has been elegantly demonstrated for many GPCRs in different
models (Albizu et al., 2010; Baba et al., 2013; Kaupmann et al., 1998; Rivero-Muller
et al., 2010; Vischer et al., 2015; Waldhoer et al., 2005).

17

ARTICLE IN PRESS

18

Resonance Energy Transfer

From the methodological point of view, in addition to the classical pharmacological and biochemical techniques, RET-based approaches have contributed considerably to the emergence of GPCR oligomerization and its investigation directly in live
cells. Indeed, different BRET and FRET assays have been developed and used over
the years, including their simple and combined application using fluorescent and
luminescent proteins, fluorescently conjugated antibodies as well as their combination with fluorescent ligands, and complementation-based approaches (Angers,
Salahpour, & Bouvier, 2001; Angers et al., 2000; Ayoub & Pfleger, 2010; Carriba
et al., 2008; Cottet, Albizu, et al., 2011; Cottet et al., 2012; Doumazane et al.,
2011; Faklaris et al., 2015; Gandia et al., 2008; Gomes et al., 2016; Hamdan,
Percherancier, Breton, & Bouvier, 2006; James, Oliveira, Carmo, Iaboni, & Davis,
2006; Johnstone & Pfleger, 2012; Kaczor & Selent, 2011; Kent, McAlpine, Sabetnia,
& Presland, 2007; Overton & Blumer, 2002; Pfleger, Seeber, & Eidne, 2006; Pin
et al., 2008; Robitaille, Heroux, Baragli, & Hebert, 2009). The most used configurations of RET-based assay consist of the fusion of the two protomers of the
GPCR dimer (homo- or heterodimer) with the energy donor and acceptor at either
their C-terminus (Figure 7(A)) or N-terminus (Figure 7(B)) and the measurement
of BRET and FRET signals directly in intact cells. Genetic fusion of the fluorescent
and luminescent proteins to the receptors of interest is required in most of the cases,
although sometimes alternative covalent labeling is used (TR-FRET/Tag-Lite). In
addition, fluorescently conjugated antibodies compatible with TR-FRET and recognizing the small tags (Flag, HA, myc) were also used to investigate GPCR oligomerization (Maurel et al., 2008, 2004). These approaches were applied in either the
absence or presence of GPCR activation and the appropriate controls (Ayoub &
Pfleger, 2010), and in most cases receptor activation had no effect on the dimer-specific RET signals, supporting the notion of constitutive and agonist-independent
GPCR oligomerization (Angers et al., 2002; Bai, 2004; Bouvier, 2001; Bulenger
et al., 2005; Devi, 2001; Gomes et al., 2016; Gurevich & Gurevich, 2008; Jordan
& Devi, 1999; Milligan, 2004b; Prinster et al., 2005; Rovira, Pin, & Giraldo,
2010; Szidonya et al., 2008; Terrillon & Bouvier, 2004b). Thus, the ligand-induced
BRET changes (increase) observed with some GPCR homo- and heterodimers have
been interpreted to reflect conformational changes within the constitutive dimers
rather than changes in its dimerization state (Figure 7(C)) as shown for MT1R/
MT2R (Ayoub et al., 2002, 2004) and CXCR4 (Percherancier et al., 2005). These
conclusions are somehow consistent with the extracellular TR-FRET changes
(decrease) using a sensor for glutamate binding on the well-established constitutive
mGluR2 dimers (Doumazane et al., 2013). However, the FRET decrease measured
on the somatostatin receptor (SSTR2) homodimer (Grant, Collier, & Kumar, 2004)
and BRET increase within the heterodimer formed by m-opioid and cholecystokinin
(CCK2) receptors (Zheng et al., 2009) were associated with the dissociation and association of the complexes, respectively. However, recent studies have challenged
some of the applications of the BRET technique and the different controls used to
reveal the oligomerization of GPCRs (Bouvier, Heveker, Jockers, Marullo, &
Milligan, 2007; James et al., 2006; Lan et al., 2015; Szalai et al., 2014).

ARTICLE IN PRESS

8. Receptor oligomerization

(A)
R2

R1

Dimerization
R1/R2

RET

RET

(B)
R1

(C)

Dimerization
R1/R2

R1/R2 dimer

R1/R2 dimer

R2

R1

R2

Basal RET

L1

L2

Binding of L1 and L2
Conformational changes
within R1/R2 dimer

L2

L1

RET increase

FIGURE 7 G protein-coupled receptor (GPCR) oligomerization assessed by resonance energy


transfer (RET)-based assays.
To investigate the homo- and heterodimerization of GPCRs, the two protomers (R1 and R2)
are fused with the donor (R1-D) and acceptor (R2-A) at either C-terminus (A) or N-terminus
(B) of R1 and R2, and the changes in the intermolecular RET signals are measured as
consequence of their physical interaction/proximity. (C) The effect of receptor activation on
GPCR dimerization is also examined by assessing the changes in the basal intermolecular
RET signals (here shown as an increase) measured within the constitutive R1/R2 dimers
reflecting the conformational changes induced by ligand binding.

Consequently, the homodimerization of some GPCRs such as b2AR has been questioned (James et al., 2006; Lan et al., 2015). Furthermore, the development of recent
fluorescence and luminescence approaches based on protein fragment complementation led to their combination with the classical BRET and FRET technologies. The
complementation of two protein moieties of luciferase and/or variants of GFP genetically fused to the receptors of interest allows specific BRET or FRET signals to be
measured as a consequence of receptor homo- or heterodimerization (Ciruela,
Vilardaga, & Fernandez-Duenas, 2010; Hebert, Gales, & Rebois, 2006; Molinari,

19

ARTICLE IN PRESS

20

Resonance Energy Transfer

Casella, & Costa, 2008; Rebois et al., 2008; Robitaille et al., 2009). The RET signals
can be measured through the complementation of the energy donor and/or acceptor
within the different protomers forming the oligomers using BRET, FRET, or both
combined (Faklaris et al., 2015). This has been used to provide evidence for the
following GPCR interactions: CB1R/dopamine D2R/adenosine A2AR (Carriba
et al., 2008), AT1R/AT2R (Porrello et al., 2011), mGluR5/dopamine D2R/adenosine
A2aR (Cabello et al., 2009), and different homo- and heteromers of mGluRs
(Doumazane et al., 2011). In addition, the heterodimerization between two or
more GPCRs has also been investigated by RET-based approaches in a liganddependent manner as described in Figure 8. The first configuration measured RET
signals between the complemented dimer protomers and their related interacting
protein (i.e., G proteins, b-arrestins) upon receptor activation and this has been reported on AT1R/a2cAR (Bellot et al., 2015), CCR2/CXCR4 (Armando et al.,
2014), dopamine D1R/D2R (Urizar et al., 2011), a1AR/m-opioid (Vilardaga et al.,
2008), and adenosine A2aR/dopamine D2R (Bonaventura et al., 2015) heterodimers
as well as the adenosine A2aR homodimer (Gandia et al., 2008) (Figure 8(A)). The
second assay is based on the GPCR-heteromer identification technology (GPCRHIT) recently developed to detect GPCR heterodimers in a ligand-dependent format
(Ayoub & Pfleger, 2010; Johnstone & Pfleger, 2012, 2015; See et al., 2011). This
assay can be applied using an intracellular interacting molecule or a fluorescently
labeled ligand (Jaeger, Armstrong, Hill, & Pfleger, 2014). GPCR-HIT using an intracellular interacting molecule was successfully applied to reveal CCR2/CCR5 (See
et al., 2011), CCR2/CXCR4 (See et al., 2011; Armando et al., 2014), AT1R/
AT2R (Porrello et al., 2011), a1AR/CXCR2 (Mustafa et al., 2012), GLP1R/GIPR
(Schelshorn et al., 2012), CXCR3/CXCR4 (Watts et al., 2013), and AT1R/CCR2
(Ayoub, Zhang, et al., 2015) heteromers (Figure 8(B)). Importantly, these two
RET configurations not only revealed new GPCR heterodimers but also shed
more light into their mechanisms of activation and/or regulation in terms of G protein coupling and b-arrestin recruitment (Armando et al., 2014; Ayoub, Zhang, et al.,
2015; Bellot et al., 2015; Mustafa et al., 2012; Terrillon & Bouvier, 2004a; Urizar
et al., 2011).
The recent advances in TR-FRET and Tag-Lite technologies using fluorescent
ligands allowed labeling the extracellular domains of GPCRs without disturbing their
function as well as the detection of homo- and heterodimers at the cell surface
(Albizu et al., 2010; Cottet, Albizu, et al., 2011; Cottet et al., 2013, 2012;
Doumazane et al., 2013; Faklaris et al., 2015; Hounsou et al., 2015). The TR-FRET
approach with the fluorescent ligands is mostly based on two different ligand-binding
assays as described in Figure 9(A) and (B). The combination of fluorescent agonists/
antagonists selective for the two GPCRs of interest is used to measure RET signal as
consequence of ligand binding on the heterodimers. This approach has been used in a
seminal work by Albizu et al. (2010) revealing for the first time the existence of
GPCR dimers in native tissues as shown for vasopressin V1a and oxytocin receptors
expressed in the mammary glands. In addition, the utilization of fluorescent ligands to
track the dimers brought interesting conclusions regarding the binding properties of

ARTICLE IN PRESS

8. Receptor oligomerization

(A)
R2

R1

Dimerization
R1/R2

L2
A2

A1

L2-induced
R1/R2/protein X complex

Protein X

R2

Protein X

RET

(B)
R1

L2

Dimerization
R1/R2

L2

L2
L2-induced
R1/R2/protein X complex

D
Protein X

Protein X

A
RET

FIGURE 8 G protein-coupled receptor (GPCR) oligomerization assessed by ligand-dependent


resonance energy transfer (RET)-based assays.
(A) In the complementation assay, the two protomers (R1 and R2) are fused with the two
moieties (R1-A1 and R2-A2) of the acceptor at their C-terminus, and an interacting protein
(i.e., G proteins, b-arrestins, etc.) is fused with the energy donor (Protein X-D). The
increase in the intermolecular RET signals between the complemented R1/R2 dimer and
protein X is measured as a consequence of R1-A1/R2-A2 interaction and the recruitment
of Protein X-D to the activated dimer upon the binding of R2-selective ligand (L2). (B)
In GPCR-heteromer identification technology configuration, only one protomer is fused with
the donor at its C-terminus (R1-D), and the interacting protein X is fused with the acceptor
(Protein X-A). The increase in the intermolecular RET signals between R1-D and protein
X-A is measured upon the activation of unmodified R2 by its selective ligand (L2)
demonstrating physical interaction between R1 and R2 and the recruitment of Protein X to
the activated R1/R2 dimer.

dimers as well as ligandedimer stoichiometry when agonists and antagonists were


used (Albizu et al., 2010). Alternatively, the ligand-binding form of the GPCRHIT assay can be utilized (Jaeger, Armstrong, et al., 2014), by the binding of donor
or acceptor fluorescent ligands on one GPCR protomer combined with the labeling
(covalent or not) of the energy donor or acceptor fused to the N-terminus of the other
protomer within the GPCR dimer as described in Figure 9(C). This is exemplified by
the study of the D1/D3 heteromer (Hounsou et al., 2015). The combination of
different Tag-Lite labeling based on SNAP-tag, CLIP-tag, and HALO-tag with the
fluorescent ligands with various spectral properties implies more possibilities for
multiplexing TR-FRET assays and simultaneous detection of the heterodimerization

21

ARTICLE IN PRESS

22

Resonance Energy Transfer

(A)
D

L1

L2

R2

R1

RET

Dimerization
R1/R2

Binding of L1 and L2

RET

(B)
R1

R2

L2
Dimerization
R1/R2

Binding of L2

(C)
D

TR-FRET

L1

R2

R1

L2
Dimerization
R1/R2

Binding of L1 and L2

Protein X

L1/L2-induced
R1/R2/protein X complex

Protein X

D A
BRET

FIGURE 9 Detection of G protein-coupled receptor oligomerization at the cell surface using


resonance energy transfer (RET)-based assays with fluorescent ligands.
(A) In the case of RET between two fluorescent ligands, the two protomers (R1 and R2) are
unmodified and labeled with their selective fluorescently conjugated ligands (D-L1 for R1 and
A-L2 for R2). The increase in the intermolecular RET signals measured between the two
bound ligands is assessed as a consequence of R1/R2 interaction at the cell surface.
(B) Alternatively, RET can be measured between one protomer N-terminally fused to the
donor (D-R1) and protomer R2-selective ligand conjugated with the acceptor (A-L2). The
increase in the intermolecular RET signals measured between D-R1 and the A-L2 bound on
R2 is assessed to reveal R1/R2 interaction at the cell surface. (C) The RET configuration in
(A) can be combined with that in Figure 8(B) where the cell surface specific time-resolved
fluorescence resonance energy transfer (TR-FRET) signals measured on R1/R2 dimers upon
the binding of their selective fluorescent ligands (D-L1 and A-L2) are correlated with the
intracellular bioluminescence resonance energy transfer (BRET) signals measured between
the R1-D0 protomer and protein-A0 as a consequence of the recruitment of the latter to the
R1/R2 dimer.

ARTICLE IN PRESS

References

between various GPCRs coexpressed in the same cell (Doumazane et al., 2011;
Faklaris et al., 2015). Interestingly, the utilization of fluorescent ligands to measure
the extracellular TR-FRET signals on GPCR heterodimers opens a possibility for
its combination with BRET measurements at the cytoplasmic side. Consequently,
conformational changes or association of the heterodimers with their signaling or
regulatory proteins (i.e., G proteins, b-arrestins) can be simultaneously measured
upon the binding of the fluorescent ligands (Figure 9(C)). Such multiplexed
TR-FRET/BRET assays would allow the correlation of the heterodimerization
between two GPCRs with its functional consequences in terms of activation, desensitization, and internalization of the heterodimers.

9. SUMMARY AND OUTLOOK


In summary, RET-based technologies have substantially contributed to the recent advances in GPCRs. Their applications have been evolved from the detection of receptorereceptor interactions (oligomerization) to dissecting, in real-time and live cells,
the molecular mechanisms of activation/deactivation, functional coupling, intracellular signaling, and trafficking of complexes involving GPCRs. Since the development of RET-based approaches, the major advances in their application to GPCRs
concern the discovery of various functional and physical interactions between
GPCRs with specific pharmacological and/or signaling properties as well as implication in physiology and pathophysiology. More importantly, the new generation of
RET-based assays using TR-FRET with fluorescent ligands has been shown to be
powerful for the detection of GPCR oligomers in their native contexts. Also, the
development of fluorescent ligands compatible with BRET and TR-FRET allows
the assessment of ligand-binding properties opening a new era for the development
of robust ligand-binding assays compatible with HTS programs. Furthermore, the
RET-based technologies enable henceforth the assessment of the pharmacology of
GPCRs as well as their function in quantitative and qualitative ways by considering
the recent concepts such as heterodimerization and biased signaling.

ACKNOWLEDGMENTS
I would like to thank LE STUDIUM Loire Valley Institute for Advanced Studies, AgreenSkills Plus, and ARD 2020 Biomedicament program for their support. Special thanks to
A/Prof. Kevin Pfleger for his comments on and review of this chapter.

REFERENCES
Agnati, L. F., Ferre, S., Lluis, C., Franco, R., & Fuxe, K. (2003). Molecular mechanisms and
therapeutical implications of intramembrane receptor/receptor interactions among

23

ARTICLE IN PRESS

24

Resonance Energy Transfer

heptahelical receptors with examples from the striatopallidal GABA neurons. Pharmacological Reviews, 55, 509e550.
Albizu, L., Cottet, M., Kralikova, M., Stoev, S., Seyer, R., Brabet, I., et al. (2010). Timeresolved FRET between GPCR ligands reveals oligomers in native tissues. Nature Chemical Biology, 6, 587e594.
Angers, S., Salahpour, A., & Bouvier, M. (2001). Biochemical and biophysical demonstration
of GPCR oligomerization in mammalian cells. Life Sciences, 68, 2243e2250.
Angers, S., Salahpour, A., & Bouvier, M. (2002). Dimerization: an emerging concept for G
protein-coupled receptor ontogeny and function. Annual Review of Pharmacology and
Toxicology, 42, 409e435.
Angers, S., Salahpour, A., Joly, E., Hilairet, S., Chelsky, D., Dennis, M., et al. (2000). Detection of beta 2-adrenergic receptor dimerization in living cells using bioluminescence
resonance energy transfer (BRET). Proceedings of the National Academy of Sciences of
the United States of America, 97, 3684e3689.
Armando, S., Quoyer, J., Lukashova, V., Maiga, A., Percherancier, Y., Heveker, N., et al.
(2014). The chemokine CXC4 and CC2 receptors form homo- and heterooligomers
that can engage their signaling G-protein effectors and betaarrestin. FASEB Journal:
Official Publication of the Federation of American Societies for Experimental Biology,
28, 4509e4523.
Armstrong, S. P., Seeber, R. M., Ayoub, M. A., Feldman, B. J., & Pfleger, K. D. (2013). Characterization of three vasopressin receptor 2 variants: an apparent polymorphism (V266A)
and two loss-of-function mutations (R181C and M311V). PLoS One, 8, e65885.
Audet, N., Gales, C., Archer-Lahlou, E., Vallieres, M., Schiller, P. W., Bouvier, M., et al.
(2008). Bioluminescence resonance energy transfer assays reveal ligand-specific conformational changes within preformed signaling complexes containing delta-opioid
receptors and heterotrimeric G proteins. The Journal of Biological Chemistry, 283,
15078e15088.
Ayoub, M. A., Al-Senaidy, A., & Pin, J. P. (2012). Receptor-G protein interaction studied by
bioluminescence resonance energy transfer: lessons from protease-activated receptor 1.
Frontiers in Endocrinology, 3, 82.
Ayoub, M. A., Angelicheva, D., Vile, D., Chandler, D., Morar, B., Cavanaugh, J. A., et al.
(2012). Deleterious GRM1 mutations in schizophrenia. PLoS One, 7, e32849.
Ayoub, M. A., Couturier, C., Lucas-Meunier, E., Angers, S., Fossier, P., Bouvier, M., et al.
(2002). Monitoring of ligand-independent dimerization and ligand-induced conformational changes of melatonin receptors in living cells by bioluminescence resonance energy
transfer. The Journal of Biological Chemistry, 277, 21522e21528.
Ayoub, M. A., Damian, M., Gespach, C., Ferrandis, E., Lavergne, O., De Wever, O., et al.
(2009). Inhibition of heterotrimeric G-protein signaling by a small molecule acting on
Galpha subunit. The Journal of Biological Chemistry, 284, 29136e29145.
Ayoub, M. A., Landomiel, F., Gallay, N., Jegot, G., Poupon, A., Crepieux, P., et al. (2015).
Assessing gonadotropin receptor function by resonance energy transfer-based assays.
Frontiers in Endocrinology, 6, 130.
Ayoub, M. A., Levoye, A., Delagrange, P., & Jockers, R. (2004). Preferential formation of
MT1/MT2 melatonin receptor heterodimers with distinct ligand interaction properties
compared with MT2 homodimers. Molecular Pharmacology, 66, 312e321.
Ayoub, M. A., Maurel, D., Binet, V., Fink, M., Prezeau, L., Ansanay, H., et al. (2007). Realtime analysis of agonist-induced activation of protease-activated receptor 1/Galphai1

ARTICLE IN PRESS

References

protein complex measured by bioluminescence resonance energy transfer in living cells.


Molecular Pharmacology, 71, 1329e1340.
Ayoub, M. A., & Pfleger, K. D. (2010). Recent advances in bioluminescence resonance energy
transfer technologies to study GPCR heteromerization. Current Opinion in Pharmacology, 10, 44e52.
Ayoub, M. A., & Pin, J. P. (2013). Interaction of protease-activated receptor 2 with G Proteins
and beta-arrestin 1 studied by bioluminescence resonance energy transfer. Frontiers in
Endocrinology (Lausanne), 4, 196.
Ayoub, M. A., Trebaux, J., Vallaghe, J., Charrier-Savournin, F., Al-Hosaini, K., Gonzalez
Moya, A., et al. (2014). Homogeneous time-resolved fluorescence-based assay to monitor
extracellular signal-regulated kinase signaling in a high-throughput format. Frontiers in
Endocrinology, 5, 94.
Ayoub, M. A., Trinquet, E., Pfleger, K. D., & Pin, J. P. (2010). Differential association modes
of the thrombin receptor PAR1 with Galphai1, Galpha12, and beta-arrestin 1. FASEB
Journal: Official Publication of the Federation of American Societies for Experimental
Biology, 24, 3522e3535.
Ayoub, M. A., Zhang, Y., Kelly, R. S., See, H. B., Johnstone, E. K., McCall, E. A., et al. (2015).
Functional interaction between angiotensin II receptor type 1 and chemokine (C-C motif)
receptor 2 with implications for chronic kidney disease. PLoS One, 10, e0119803.
Azpiazu, I., & Gautam, N. (2004). A fluorescence resonance energy transfer-based sensor
indicates that receptor access to a G protein is unrestricted in a living mammalian cell.
The Journal of Biological Chemistry, 279, 27709e27718.
Baba, K., Benleulmi-Chaachoua, A., Journe, A. S., Kamal, M., Guillaume, J. L., Dussaud, S.,
et al. (2013). Heteromeric MT1/MT2 melatonin receptors modulate photoreceptor
function. Science Signaling, 6, ra89.
Bai, M. (2004). Dimerization of G-protein-coupled receptors: roles in signal transduction.
Cellular Signalling, 16, 175e186.
Barak, L. S., Ferguson, S. S., Zhang, J., & Caron, M. G. (1997). A beta-arrestin/green fluorescent protein biosensor for detecting G protein-coupled receptor activation. The Journal of
Biological Chemistry, 272, 27497e27500.
Barak, L. S., Salahpour, A., Zhang, X., Masri, B., Sotnikova, T. D., Ramsey, A. J., et al.
(2008). Pharmacological characterization of membrane-expressed human trace amineassociated receptor 1 (TAAR1) by a bioluminescence resonance energy transfer cAMP
biosensor. Molecular Pharmacology, 74, 585e594.
Bellot, M., Galandrin, S., Boularan, C., Matthies, H. J., Despas, F., Denis, C., et al. (2015).
Dual agonist occupancy of AT1-R-a2C-AR heterodimers results in atypical Gs-PKA
signaling. Nature Chemical Biology, 11, 271e279.
Berglund, M. M., Schober, D. A., Statnick, M. A., McDonald, P. H., & Gehlert, D. R. (2003).
The use of bioluminescence resonance energy transfer 2 to study neuropeptide Y receptor
agonist-induced beta-arrestin 2 interaction. Journal of Pharmacology and Experimental
Therapeutics, 306, 147e156.
Bertrand, L., Parent, S., Caron, M., Legault, M., Joly, E., Angers, S., et al. (2002). The
BRET2/arrestin assay in stable recombinant cells: a platform to screen for compounds
that interact with G protein-coupled receptors (GPCRS). Journal of Receptor and Signal
Transduction Research, 22, 533e541.
Bockaert, J., Homburger, V., & Rouot, B. (1987). GTP binding proteins: a key role in cellular
communication. Biochimie, 69, 329e338.

25

ARTICLE IN PRESS

26

Resonance Energy Transfer

Bockaert, J., & Philippe Pin, J. (1999). Molecular tinkering of G protein-coupled receptors: an
evolutionary success. The EMBO Journal, 18, 1723e1729.
Bonaventura, J., Navarro, G., Casado-Anguera, V., Azdad, K., Rea, W., Moreno, E., et al.
(2015). Allosteric interactions between agonists and antagonists within the adenosine
A2A receptor-dopamine D2 receptor heterotetramer. Proceedings of the National Academy
of Sciences of the United States of America, 112, E3609eE3618.
Bourne, H. R. (1997). How receptors talk to trimeric G proteins. Current Opinion in Cell
Biology, 9, 134e142.
Bouvier, M. (2001). Oligomerization of G-protein-coupled transmitter receptors. Nature
Reviews Neuroscience, 2, 274e286.
Bouvier, M., Heveker, N., Jockers, R., Marullo, S., & Milligan, G. (2007). BRET analysis of
GPCR oligomerization: newer does not mean better. Nature Methods, 4, 3e4.
Brady, A. E., & Limbird, L. E. (2002). G protein-coupled receptor interacting proteins:
emerging roles in localization and signal transduction. Cellular Signalling, 14,
297e309.
Breton, B., Lagace, M., & Bouvier, M. (2010). Combining resonance energy transfer methods
reveals a complex between the alpha2A-adrenergic receptor, Galphai1beta1gamma2, and
GRK2. FASEB Journal: Official Publication of the Federation of American Societies for
Experimental Biology, 24, 4733e4743.
Bulenger, S., Marullo, S., & Bouvier, M. (2005). Emerging role of homo- and heterodimerization in G-protein-coupled receptor biosynthesis and maturation. Trends in Pharmacological Science, 26, 131e137.
Cabello, N., Ganda, J., Bertarelli, D. C. G., Watanabe, M., Llus, C., Franco, R., et al. (2009).
Metabotropic glutamate type 5, dopamine D2 and adenosine A2a receptors form higherorder oligomers in living cells. Journal of Neurochemistry, 109, 1497e1507.
Carman, C. V., & Benovic, J. L. (1998). G-protein-coupled receptors: turn-ons and turn-offs.
Current Opinion in Neurobiology, 8, 335e344.
Carriba, P., Navarro, G., Ciruela, F., Ferre, S., Casado, V., Agnati, L., et al. (2008). Detection
of heteromerization of more than two proteins by sequential BRET-FRET. Nature
Methods, 5, 727e733.
Castro, M., Nikolaev, V. O., Palm, D., Lohse, M. J., & Vilardaga, J. P. (2005). Turn-on switch
in parathyroid hormone receptor by a two-step parathyroid hormone binding mechanism.
Proceedings of the National Academy of Sciences of the United States of America, 102,
16084e16089.
Charest, P. G., Terrillon, S., & Bouvier, M. (2005). Monitoring agonist-promoted conformational changes of beta-arrestin in living cells by intramolecular BRET. EMBO Reports, 6,
334e340.
Ciruela, F., Vilardaga, J. P., & Fernandez-Duenas, V. (2010). Lighting up multiprotein complexes: lessons from GPCR oligomerization. Trends in Biotechnology, 28, 407e415.
Comps-Agrar, L., Kniazeff, J., Brock, C., Trinquet, E., & Pin, J. P. (2012). Stability of
GABAB receptor oligomers revealed by dual TR-FRET and drug-induced cell surface
targeting. FASEB Journal: Official Publication of the Federation of American Societies
for Experimental Biology, 26, 3430e3439.
Conn, P. M. (2013). Receptor-receptor interactions. In P. M. Conn (Ed.), Methods in cell
biology (Vol. 117, p. 538). Elsevier Science.
Cottet, M., Albizu, L., Comps-Agrar, L., Trinquet, E., Pin, J. P., Mouillac, B., et al. (2011).
Time resolved FRET strategy with fluorescent ligands to analyze receptor interactions

ARTICLE IN PRESS

References

in native tissues: application to GPCR oligomerization. Methods in Molecular Biology,


746, 373e387.
Cottet, M., Faklaris, O., Falco, A., Trinquet, E., Pin, J. P., Mouillac, B., et al. (2013). Fluorescent ligands to investigate GPCR binding properties and oligomerization. Biochemical
Society Transactions, 41, 148e153.
Cottet, M., Faklaris, O., Maurel, D., Scholler, P., Doumazane, E., Trinquet, E., et al. (2012).
BRET and Time-resolved FRET strategy to study GPCR oligomerization: from cell lines
toward native tissues. Frontiers in Endocrinology (Lausanne), 3, 92.
Cottet, M., Faklaris, O., Zwier, J. M., Trinquet, E., Pin, J. P., & Durroux, T. (2011). Original
fluorescent ligand-based assays open new perspectives in G protein-coupled receptor drug
screening. Pharmaceuticals, 4, 202e214.
Dacres, H., Wang, J., Leitch, V., Horne, I., Anderson, A. R., & Trowell, S. C. (2011). Greatly
enhanced detection of a volatile ligand at femtomolar levels using bioluminescence resonance energy transfer (BRET). Biosensors and Bioelectronics, 29, 119e124.
Dalrymple, M. B., Jaeger, W. C., Eidne, K. A., & Pfleger, K. D. (2011). Temporal profiling of
orexin receptor-arrestin-ubiquitin complexes reveals differences between receptor
subtypes. The Journal of Biological Chemistry, 286, 16726e16733.
Damian, M., Mary, S., Maingot, M., MKadmi, C., Gagne, D., Leyris, J. P., et al. (2015).
Ghrelin receptor conformational dynamics regulate the transition from a preassembled
to an active receptor:Gq complex. Proceedings of the National Academy of Sciences of
the United States of America, 112, 1601e1606.
Damian, M., Mary, S., Martin, A., Pin, J. P., & Baneres, J. L. (2008). G protein activation by
the leukotriene B4 receptor dimer. Evidence for an absence of trans-activation. The Journal of Biological Chemistry, 283, 21084e21092.
De, A., Loening, A. M., & Gambhir, S. S. (2007). An improved bioluminescence resonance
energy transfer strategy for imaging intracellular events in single cells and living subjects.
Cancer Research, 67, 7175e7183.
Degorce, F., Card, A., Soh, S., Trinquet, E., Knapik, G. P., & Xie, B. (2009). HTRF: a technology tailored for drug discovery e a review of theoretical aspects and recent applications.
Current Chemical Genomics, 3, 22e32.
Devi, L. A. (2001). Heterodimerization of G-protein-coupled receptors: pharmacology,
signaling and trafficking. Trends in Pharmacological Sciences, 22, 532e537.
DeWire, S. M., Ahn, S., Lefkowitz, R. J., & Shenoy, S. K. (2007). Beta-arrestins and cell
signaling. Annual Review of Physiology, 69, 483e510.
Digby, G. J., Lober, R. M., Sethi, P. R., & Lambert, N. A. (2006). Some G protein heterotrimers physically dissociate in living cells. Proceedings of the National Academy of
Sciences of the United States of America, 9, 9.
DiPilato, L. M., Cheng, X., & Zhang, J. (2004). Fluorescent indicators of cAMP and Epac
activation reveal differential dynamics of cAMP signaling within discrete subcellular
compartments. Proceedings of the National Academy of Sciences of the United States
of America, 101, 16513e16518.
Doumazane, E., Scholler, P., Fabre, L., Zwier, J. M., Trinquet, E., Pin, J. P., et al. (2013). Illuminating the activation mechanisms and allosteric properties of metabotropic glutamate
receptors. Proceedings of the National Academy of Sciences of the United States of
America, 110, E1416eE1425.
Doumazane, E., Scholler, P., Zwier, J. M., Trinquet, E., Rondard, P., & Pin, J. P. (2011). A new
approach to analyze cell surface protein complexes reveals specific heterodimeric

27

ARTICLE IN PRESS

28

Resonance Energy Transfer

metabotropic glutamate receptors. FASEB Journal: Official Publication of the Federation


of American Societies for Experimental Biology, 25, 66e77.
Dragulescu-Andrasi, A., Chan, C. T., De, A., Massoud, T. F., & Gambhir, S. S. (2011). Bioluminescence resonance energy transfer (BRET) imaging of proteineprotein interactions
within deep tissues of living subjects. Proceedings of the National Academy of Sciences
of the United States of America, 108, 12060e12065.
El Moustaine, D., Granier, S., Doumazane, E., Scholler, P., Rahmeh, R., Bron, P., et al. (2012).
Distinct roles of metabotropic glutamate receptor dimerization in agonist activation and
G-protein coupling. Proceedings of the National Academy of Sciences of the United States
of America, 109, 16342e16347.
Evans, N. A., Groarke, D. A., Warrack, J., Greenwood, C. J., Dodgson, K., Milligan, G., et al.
(2001). Visualizing differences in ligand-induced beta-arrestin-GFP interactions and trafficking between three recently characterized G protein-coupled receptors. Journal of
Neurochemistry, 77, 476e485.
Faklaris, O., Cottet, M., Falco, A., Villier, B., Laget, M., Zwier, J. M., et al. (2015). Multicolor
time-resolved Forster resonance energy transfer microscopy reveals the impact of GPCR
oligomerization on internalization processes. FASEB Journal: Official Publication of the
Federation of American Societies for Experimental Biology, 29, 2235e2246.
Ferguson, S. S., & Caron, M. G. (2004). Green fluorescent protein-tagged beta-arrestin translocation as a measure of G protein-coupled receptor activation. Methods in Molecular
Biology, 237, 121e126.
Ferre, S., Baler, R., Bouvier, M., Caron, M. G., Devi, L. A., Durroux, T., et al. (2009). Building a new conceptual framework for receptor heteromers. Nature Chemical Biology, 5,
131e134.
Forster, T. (1948). Intermolecular energy migration and fluorescence. Annals of Physics, 437,
55e75.
Fredholm, B. B., Hokfelt, T., & Milligan, G. (2007). G-protein-coupled receptors: an update.
Acta Physiologica (Oxford, England), 190, 3e7.
Fuxe, K., & Agnati, L. F. (1985). Receptor-receptor interactions in the central nervous system.
A new integrative mechanism in synapses. Medicinal Research Reviews, 5, 441e482.
Fuxe, K., Marcellino, D., Guidolin, D., Woods, A. S., & Agnati, L. F. (2008). Heterodimers
and receptor mosaics of different types of G-protein-coupled receptors. Physiology
(Bethesda, Md.), 23, 322e332.
Gabriel, D., Vernier, M., Pfeifer, M. J., Dasen, B., Tenaillon, L., & Bouhelal, R. (2003). High
throughput screening technologies for direct cyclic AMP measurement. Assay and Drug
Development Technologies, 1, 291e303.
Gales, C., Rebois, R. V., Hogue, M., Trieu, P., Breit, A., Hebert, T. E., et al. (2005). Realtime monitoring of receptor and G-protein interactions in living cells. Nature Methods,
2, 177e184.
Gales, C., Van Durm, J. J., Schaak, S., Pontier, S., Percherancier, Y., Audet, M., et al. (2006).
Probing the activation-promoted structural rearrangements in preassembled receptor-G
protein complexes. Nature Structural & Molecular Biology, 13, 778e786.
Gandia, J., Galino, J., Amaral, O. B., Soriano, A., Lluis, C., Franco, R., et al. (2008). Detection
of higher-order G protein-coupled receptor oligomers by a combined BRET-BiFC
technique. FEBS Letters, 582, 2979e2984.
Garland, S. L. (2013). Are GPCRs still a source of new targets? Journal of Biomolecular
Screening, 18, 947e966.

ARTICLE IN PRESS

References

Gether, U. (2000). Uncovering molecular mechanisms involved in activation of G proteincoupled receptors. Endocrine Reviews, 21, 90e113.
Gether, U., Lin, S., & Kobilka, B. K. (1995). Fluorescent labeling of purified beta 2 adrenergic
receptor. Evidence for ligand-specific conformational changes. The Journal of Biological
Chemistry, 270, 28268e28275.
Ghanouni, P., Gryczynski, Z., Steenhuis, J. J., Lee, T. W., Farrens, D. L., Lakowicz, J. R., et al.
(2001). Functionally different agonists induce distinct conformations in the G protein
coupling domain of the beta 2 adrenergic receptor. The Journal of Biological Chemistry,
276, 24433e24436.
Ghanouni, P., Steenhuis, J. J., Farrens, D. L., & Kobilka, B. K. (2001). Agonist-induced
conformational changes in the G-protein-coupling domain of the beta 2 adrenergic
receptor. Proceedings of the National Academy of Sciences of the United States of
America, 98, 5997e6002.
Gibson, S. K., & Gilman, A. G. (2006). Gialpha and Gbeta subunits both define selectivity
of G protein activation by alpha2-adrenergic receptors. Proceedings of the National
Academy of Sciences of the United States of America, 103, 212e217.
Gilman, A. G. (1987). G proteins: transducers of receptor-generated signals. Annual Review of
Biochemistry, 56, 615e649.
Gomes, I., Ayoub, M. A., Fujita, W., Jaeger, W. C., Pfleger, K. D. G., & Lakshmi, A. D.
(2016). G protein-coupled receptor heteromers. Annual Review of Pharmacology and
Toxicology, 56 (in press).
Gomes, I., Gupta, A., & Devi, L. A. (2013). G-protein-coupled heteromers: regulation in
disease. Methods in Enzymology, 521, 219e238.
Gorokhovatsky, A. Y., Marchenkov, V. V., Rudenko, N. V., Ivashina, T. V., Ksenzenko, V. N.,
Burkhardt, N., et al. (2004). Fusion of Aequorea victoria GFP and aequorin provides their
Ca(2)-induced interaction that results in red shift of GFP absorption and efficient bioluminescence energy transfer. Biochemical and Biophysical Research Communications,
320, 703e711.
Granier, S., Kim, S., Fung, J. J., Bokoch, M. P., & Parnot, C. (2009). FRET-based measurement of GPCR conformational changes. Methods in Molecular Biology, 552, 253e268.
Granier, S., Terrillon, S., Pascal, R., Demene, H., Bouvier, M., Guillon, G., et al. (2004). A
cyclic peptide mimicking the third intracellular loop of the V2 vasopressin receptor inhibits signaling through its interaction with receptor dimer and G protein. The Journal
of Biological Chemistry, 279, 50904e50914.
Grant, M., Collier, B., & Kumar, U. (2004). Agonist-dependent dissociation of human
somatostatin receptor 2 dimers: a role in receptor trafficking. The Journal of Biological
Chemistry, 279, 36179e36183.
Gurevich, V. V., & Gurevich, E. V. (2004). The molecular acrobatics of arrestin. Trends in
Pharmacological Sciences, 25, 105e111.
Gurevich, V. V., & Gurevich, E. V. (2008). How and why do GPCRs dimerize? Trends in Pharmacological Sciences, 29, 234e240.
Hamdan, F. F., Percherancier, Y., Breton, B., & Bouvier, M. (2006). Monitoring proteinprotein interactions in living cells by bioluminescence resonance energy transfer
(BRET). Current Protocols in Neuroscience (Chapter 5: Unit 5.23). http://dx.doi.org/
10.1002/0471142301.ns0523s34.
Hamm, H. E. (1998). The many faces of G protein signaling. The Journal of Biological
Chemistry, 273, 669e672.

29

ARTICLE IN PRESS

30

Resonance Energy Transfer

Harvey, C. D., Ehrhardt, A. G., Cellurale, C., Zhong, H., Yasuda, R., Davis, R. J., et al. (2008).
A genetically encoded fluorescent sensor of ERK activity. Proceedings of the National
Academy of Sciences of the United States of America, 105, 19264e19269.
Hasbi, A., Nguyen, T., Fan, T., Cheng, R., Rashid, A., Alijaniaram, M., et al. (2007). Trafficking of preassembled opioid mu-delta heterooligomer-Gz signaling complexes to the
plasma membrane: coregulation by agonists. Biochemistry, 46, 12997e13009.
Hebert, T. E., Gales, C., & Rebois, R. V. (2006). Detecting and imaging protein-protein interactions during G protein-mediated signal transduction in vivo and in situ by using fluorescence-based techniques. Cell Biochemistry and Biophysics, 45, 85e109.
Heding, A. (2004). Use of the BRET 7TM receptor/beta-arrestin assay in drug discovery and
screening. Expert Review of Molecular Diagnostics, 4, 403e411.
Heding, A., Vrecl, M., Hanyaloglu, A. C., Sellar, R., Taylor, P. L., & Eidne, K. A. (2000). The
rat gonadotropin-releasing hormone receptor internalizes via a beta-arrestin-independent,
but dynamin-dependent, pathway: addition of a carboxyl-terminal tail confers betaarrestin dependency. Endocrinology, 141, 299e306.
Hein, P., Frank, M., Hoffmann, C., Lohse, M. J., & Bunemann, M. (2005). Dynamics of
receptor/G protein coupling in living cells. The EMBO Journal, 24, 4106e4114.
Hermans, E. (2003). Biochemical and pharmacological control of the multiplicity of coupling
at G-protein-coupled receptors. Pharmacology and Therapeutics, 99, 25e44.
Hoffmann, C., Gaietta, G., Bunemann, M., Adams, S. R., Oberdorff-Maass, S., Behr, B., et al.
(2005). A FlAsH-based FRET approach to determine G protein-coupled receptor activation in living cells. Nature Methods, 2, 171e176.
Holliday, N. D., Holst, B., Rodionova, E. A., Schwartz, T. W., & Cox, H. M. (2007). Importance of constitutive activity and arrestin-independent mechansims for intracellular trafficking of the ghrelin receptor. Molecular Endocrinology, 21, 3100e3112.
Hounsou, C., Margathe, J. F., Oueslati, N., Belhocine, A., Dupuis, E., Thomas, C., et al.
(2015). Time-resolved FRET binding assay to investigate hetero-oligomer binding properties: proof of concept with dopamine D1/D3 heterodimer. ACS Chemical Biology, 10,
466e474.
Hu, L. A., Zhou, T., Hamman, B. D., & Liu, Q. (2008). A homogeneous G protein-coupled
receptor ligand binding assay based on time-resolved fluorescence resonance energy
transfer. Assay and Drug Development Technologies, 6, 543e550.
Irannejad, R., Tomshine, J. C., Tomshine, J. R., Chevalier, M., Mahoney, J. P., Steyaert, J.,
et al. (2013). Conformational biosensors reveal GPCR signalling from endosomes.
Nature, 495, 534e538.
Jaeger, W., Armstrong, S., Hill, S., & Pfleger, K. D. G. (2014). Biophysical detection of
diversity and bias in GPCR function. Frontiers in Endocrinology, 5, 26.
Jaeger, W. C., Seeber, R. M., Eidne, K. A., & Pfleger, K. D. G. (2014). Molecular determinants
of orexin receptor-arrestin-ubiquitin complex formation. British Journal of Pharmacology, 171, 364e374.
James, J. R., Oliveira, M. I., Carmo, A. M., Iaboni, A., & Davis, S. J. (2006). A rigorous experimental framework for detecting protein oligomerization using bioluminescence resonance energy transfer. Nature Methods, 3, 1001e1006.
Janetopoulos, C., & Devreotes, P. (2002). Monitoring receptor-mediated activation of heterotrimeric G-proteins by fluorescence resonance energy transfer. Methods, 27, 366e373.
Janetopoulos, C., Jin, T., & Devreotes, P. (2001). Receptor-mediated activation of heterotrimeric G-proteins in living cells. Science, 291, 2408e2411.

ARTICLE IN PRESS

References

Jensen, D. D., Godfrey, C. B., Niklas, C., Canals, M., Kocan, M., Poole, D. P., et al. (2013).
The bile acid receptor TGR5 does not interact with beta-arrestins or traffic to endosomes
but transmits sustained signals from plasma membrane rafts. The Journal of Biological
Chemistry, 288, 22942e22960.
Jia, Y., Quinn, C. M., Gagnon, A. I., & Talanian, R. (2006). Homogeneous time-resolved fluorescence and its applications for kinase assays in drug discovery. Analytical Biochemistry,
356, 273e281.
Jiang, L. I., Collins, J., Davis, R., Lin, K.-M., DeCamp, D., Roach, T., et al. (2007). Use of a
cAMP BRET sensor to characterize a novel regulation of cAMP by the sphingosine
1-phosphate/G13 pathway. The Journal of Biological Chemistry, 282, 10576e10584.
Johnstone, E. K., & Pfleger, K. D. (2012). Receptor-Heteromer Investigation Technology and
its application using BRET. Frontiers in Endocrinology (Lausanne), 3, 101.
Johnstone, K. M., & Pfleger, K. D. G. (2015). Bioluminescence resonance energy transfer
approaches to discover bias in GPCR signaling. In M. Filizola (Ed.), Methods in molecular biology (Vol. 1335, pp. 191e204). New York: Springer Science.
Jordan, B. A., & Devi, L. A. (1999). G-protein-coupled receptor heterodimerization modulates receptor function. Nature, 399, 697e700.
Jorgensen, R., Martini, L., Schwartz, T. W., & Elling, C. E. (2005). Characterization of
glucagon-like peptide-1 receptor beta-arrestin 2 interaction: a high-affinity receptor
phenotype. Molecular Endocrinology, 19, 812e823.
Journe, A.-S., Habib, S. A. M., Dodda, B. R., Morcos, M. N. F., Sadek, M. S., Tadros, S. A. A.,
et al. (2014). N1-linked melatonin dimers as bivalent ligands targeting dimeric melatonin
receptors. Medicinal Chemistry Communication, 5, 792e796.
Kaczor, A. A., Makarska-Bialokoz, M., Selent, J., de la Fuente, R. A., Marti-Solano, M., &
Castro, M. (2014). Application of BRET for studying G protein-coupled receptors.
Mini Reviews in Medicinal Chemistry, 14, 411e425.
Kaczor, A. A., & Selent, J. (2011). Oligomerization of G protein-coupled receptors: biochemical and biophysical methods. Current Medicinal Chemistry, 18, 4606e4634.
Kamal, M., Marquez, M., Vauthier, V., Leloire, A., Froguel, P., Jockers, R., et al. (2009).
Improved donor/acceptor BRET couples for monitoring beta-arrestin recruitment to G
protein-coupled receptors. Biotechnology Journal, 4, 1337e1344.
Katritch, V., Cherezov, V., & Stevens, R. C. (2013). Structure-function of the G protein-coupled
receptor superfamily. Annual Review of Pharmacology and Toxicology, 53, 531e556.
Kaupmann, K., Malitschek, B., Schuler, V., Heid, J., Froest, W., Beck, P., et al. (1998).
GABA(B)-receptor subtypes assemble into functional heteromeric complexes. 396,
683e687.
Kenakin, T. P. (2012). Biased signalling and allosteric machines: new vistas and challenges for
drug discovery. British Journal of Pharmacology, 165, 1659e1669.
Kent, T., McAlpine, C., Sabetnia, S., & Presland, J. (2007). G-protein-coupled receptor heterodimerization: assay technologies to clinical significance. Current Opinion in Drug Discovery and Development, 10, 580e589.
Klarenbeek, J., Goedhart, J., van Batenburg, A., Groenewald, D., & Jalink, K. (2015). Fourthgeneration epac-based FRET sensors for cAMP feature exceptional brightness, photostability and dynamic range: characterization of dedicated sensors for FLIM, for ratiometry
and with high affinity. PLoS One, 10, e0122513.
Klarenbeek, J., & Jalink, K. (2014). Detecting cAMP with an EPAC-based FRET sensor in
single living cells. Methods in Molecular Biology, 1071, 49e58.

31

ARTICLE IN PRESS

32

Resonance Energy Transfer

Kobilka, B., & Schertler, G. F. (2008). New G-protein-coupled receptor crystal structures: insights and limitations. Trends in Pharmacological Sciences, 29, 79e83.
Kocan, M., Dalrymple, M. B., Seeber, R. M., Feldman, B. J., & Pfleger, K. D. (2010). Enhanced
BRET technology for the monitoring of agonist-induced and agonist-independent interactions between GPCRs and beta-arrestins. Frontiers in Endocrinology (Lausanne), 1, 12.
Kocan, M., & Pfleger, K. D. (2009). Detection of GPCR/beta-arrestin interactions in live cells
using bioluminescence resonance energy transfer technology. Methods in Molecular
Biology, 552, 305e317.
Kocan, M., See, H. B., Sampaio, N. G., Eidne, K. A., Feldman, B. J., & Pfleger, K. D. (2009).
Agonist-independent interactions between beta-arrestins and mutant vasopressin type II
receptors associated with nephrogenic syndrome of inappropriate antidiuresis. Molecular
Endocrinology, 4, 559e571.
Koelle, M. R. (2006). Heterotrimeric G protein signaling: getting inside the cell. Cell, 126,
25e27.
Krasel, C., Bunemann, M., Lorenz, K., & Lohse, M. J. (2005). Beta-arrestin binding to the
beta2-adrenergic receptor requires both receptor phosphorylation and receptor
activation. The Journal of Biological Chemistry, 280, 9528e9535.
Kuravi, S., Lan, T. H., Barik, A., & Lambert, N. A. (2010). Third-party bioluminescence
resonance energy transfer indicates constitutive association of membrane proteins: application to class a g-protein-coupled receptors and g-proteins. Biophysical Journal, 98,
2391e2399.
Lachance, M., Ethier, N., Wolbring, G., Schnetkamp, P. P., & Hebert, T. E. (1999). Stable association of G proteins with beta 2AR is independent of the state of receptor activation.
Cellular Signalling, 11, 523e533.
Lagerstrom, M. C., & Schioth, H. B. (2008). Structural diversity of G protein-coupled receptors and significance for drug discovery. Nature Reviews Drug Discovery, 7, 339e357.
Lan, T. H., Kuravi, S., & Lambert, N. A. (2011). Internalization dissociates beta2-adrenergic
receptors. PLoS One, 6, e17361.
Lan, T. H., Liu, Q., Li, C., Wu, G., Steyaert, J., & Lambert, N. A. (2015). BRET evidence that
beta2 adrenergic receptors do not oligomerize in cells. Scientific Reports, 5, 10166.
Laporte, S. A., Oakley, R. H., Holt, J. A., Barak, L. S., & Caron, M. G. (2000). The interaction
of beta-arrestin with the AP-2 adaptor is required for the clustering of beta 2-adrenergic
receptor into clathrin-coated pits. The Journal of Biological Chemistry, 275, 23120e23126.
Leff, P. (1995). The two-state model of receptor activation. Trends in Pharmacological
Sciences, 16, 89e97.
Leff, P., & Scaramellini, C. (1998). Promiscuity, pre-coupling and instability. Trends in Pharmacological Sciences, 19, 13.
Lefkowitz, R. J., & Shenoy, S. K. (2005). Transduction of receptor signals by beta-arrestins.
Science, 308, 512e517.
Levoye, A., Balabanian, K., Baleux, F., Bachelerie, F., & Lagane, B. (2009). CXCR7 heterodimerizes with CXCR4 and regulates CXCL12-mediated G protein signaling. Blood, 113,
6085e6093.
Levoye, A., Dam, J., Ayoub, M. A., Guillaume, J. L., Couturier, C., Delagrange, P., et al.
(2006). The orphan GPR50 receptor specifically inhibits MT1 melatonin receptor function
through heterodimerization. The EMBO Journal, 25, 3012e3023.
Levoye, A., Dam, J., Ayoub, M. A., Guillaume, J. L., & Jockers, R. (2006). Do orphan
G-protein-coupled receptors have ligand-independent functions? New insights from receptor heterodimers. EMBO Reports, 7, 1094e1098.

ARTICLE IN PRESS

References

Limbird, L. E. (1983). Beta-adrenergic stimulation of adenylate cyclase and alpha-adrenergic


inhibition of adenylate cyclase: GTP-binding proteins as macromolecular messengers.
Advances in Experimental Medicine and Biology, 161, 91e111.
Limbird, L. E. (2004). The receptor concept: a continuing evolution. Molecular Interventions,
4, 326e336.
Lin, H., & Trejo, J. (2013). Transactivation of the PAR1-PAR2 heterodimer by thrombin
elicits beta-arrestin-mediated endosomal signaling. The Journal of Biological Chemistry,
288, 11203e11215.
Lohse, M. J., Bunemann, M., Hoffmann, C., Vilardaga, J. P., & Nikolaev, V. O. (2007). Monitoring receptor signaling by intramolecular FRET. Current Opinion in Pharmacology,
3(3).
Lohse, M. J., Nuber, S., & Hoffmann, C. (2012). Fluorescence/Bioluminescence resonance
energy transfer techniques to study g-protein-coupled receptor activation and signaling.
Pharmacological Reviews, 64, 299e336.
Louie, A. Y., & Tromberg, B. J. (1998). Fluorescence resonance energy Transfer: FRET
studies of ligand binding of cell surface receptors. Journal of Fluorescence, 8, 13e20.
Lundstrom, K. (2006). Latest development in drug discovery on G protein-coupled receptors.
Current Protein & Peptide Science, 7, 465e470.
Luttrell, L. M., & Lefkowitz, R. J. (2002). The role of beta-arrestins in the termination and
transduction of G-protein-coupled receptor signals. Journal of Cell Science, 115, 455e465.
Martikkala, E., Lehmusto, M., Lilja, M., Rozwandowicz-Jansen, A., Lunden, J., Tomohiro, T.,
et al. (2009). Cell-based beta2-adrenergic receptor-ligand binding assay using synthesized
europium-labeled ligands and time-resolved fluorescence. Analytical Biochemistry, 392,
103e109.
Maurel, D., Comps-Agrar, L., Brock, C., Rives, M. L., Bourrier, E., Ayoub, M. A., et al. (2008).
Cell-surface protein-protein interaction analysis with time-resolved FRET and snap-tag
technologies: application to GPCR oligomerization. Nature Methods, 5, 561e567.
Maurel, D., Kniazeff, J., Mathis, G., Trinquet, E., Pin, J. P., & Ansanay, H. (2004). Cell surface
detection of membrane protein interaction with homogeneous time-resolved fluorescence
resonance energy transfer technology. Analytical Biochemistry, 329, 253e262.
Milligan, G. (2004a). Applications of bioluminescence- and fluorescence resonance energy
transfer to drug discovery at G protein-coupled receptors. European Journal of Pharmaceutical Sciences, 21, 397e405.
Milligan, G. (2004b). G protein-coupled receptor dimerization: function and ligand
pharmacology. Molecular Pharmacology, 66, 1e7.
Molinari, P., Casella, I., & Costa, T. (2008). Functional complementation of high-efficiency
resonance energy transfer: a new tool for the study of protein binding interactions in living
cells. Biochemical Journal, 409, 251e261.
Monnier, C., Tu, H., Bourrier, E., Vol, C., Lamarque, L., Trinquet, E., et al. (2011). Transactivation between 7TM domains: implication in heterodimeric GABAB receptor
activation. The EMBO Journal, 30, 32e42.
Moreira, I. S. (2014). Structural features of the G-protein/GPCR interactions. Biochimica et
Biophysica Acta, 1840, 16e33.
Mustafa, S., See, H. B., Seeber, R. M., Armstrong, S. P., White, C. W., Ventura, S., et al.
(2012). Identification and profiling of novel alpha1A-adrenoceptor-CXC chemokine receptor 2 heteromer. The Journal of Biological Chemistry, 287, 12952e12965.
Nakamura, T., Aoki, K., & Matsuda, M. (2005). Monitoring spatio-temporal regulation of Ras
and Rho GTPase with GFP-based FRET probes. Methods, 37, 146e153.

33

ARTICLE IN PRESS

34

Resonance Energy Transfer

Nakamura, T., Kurokawa, K., Kiyokawa, E., & Matsuda, M. (2006). Analysis of the spatiotemporal activation of rho GTPases using Raichu probes. Methods in Enzymology, 406, 315e332.
Naumann, E. A., Kampff, A. R., Prober, D. A., Schier, A. F., & Engert, F. (2010). Monitoring
neural activity with bioluminescence during natural behavior. Nature Neuroscience, 13,
513e520.
Neumann, L., Wohland, T., Whelan, R. J., Zare, R. N., & Kobilka, B. K. (2002). Functional
immobilization of a ligand-activated G-protein-coupled receptor. Chembiochem, 3,
993e998.
Nguyen, A. W., & Daugherty, P. S. (2005). Evolutionary optimization of fluorescent proteins
for intracellular FRET. Nature Biotechnology, 23, 355e360.
Nobles, M., Benians, A., & Tinker, A. (2005). Heterotrimeric G proteins precouple with
G protein-coupled receptors in living cells. Proceedings of the National Academy of
Sciences of the United States of America, 102, 18706e18711.
Norskov-Lauritsen, L., Thomsen, A. R., & Brauner-Osborne, H. (2014). G protein-coupled
receptor signaling analysis using homogenous time-resolved Forster resonance energy
transfer (HTRF(R)) technology. International Journal of Molecular Sciences, 15,
2554e2572.
Overton, M. C., & Blumer, K. J. (2002). Use of fluorescence resonance energy transfer to
analyze oligomerization of G-protein-coupled receptors expressed in yeast. Methods,
27, 324e332.
Percherancier, Y., Berchiche, Y. A., Slight, I., Volkmer-Engert, R., Tamamura, H., Fujii, N.,
et al. (2005). Bioluminescence resonance energy transfer reveals ligand-induced conformational changes in CXCR4 homo- and heterodimers. The Journal of Biological Chemistry, 280, 9895e9903.
Pfleger, K. D., Dalrymple, M. B., Dromey, J. R., & Eidne, K. A. (2007). Monitoring interactions between G-protein-coupled receptors and beta-arrestins. Biochemical Society Transactions, 35, 764e766.
Pfleger, K. D., & Eidne, K. A. (2006). Illuminating insights into protein-protein interactions
using bioluminescence resonance energy transfer (BRET). Nature Methods, 3, 165e174.
Pfleger, K. D., Seeber, R. M., & Eidne, K. A. (2006). Bioluminescence resonance energy
transfer (BRET) for the real-time detection of protein-protein interactions. Nature Protocols, 1, 337e345.
Philip, F., Sengupta, P., & Scarlata, S. (2007). Signaling through a G Protein-coupled receptor
and its corresponding G protein follows a stoichiometrically limited model. The Journal of
Biological Chemistry, 282, 19203e19216.
Pin, J. P., Ayoub, M. A., Maurel, D., Perroy, J., & Trinquet, E. (2008). Energy transfer technologies to monitor the dynamics and signaling properties of Geproteinecoupled receptors in
living cells. In E. Pebay-Peyroula (Ed.), Biophysical analysis of membrane proteinse
Investigating structure and function, 13, pp. 311e334). Weinheim: Wiley-VCH.
Porrello, E. R., Pfleger, K. D., Seeber, R. M., Qian, H., Oro, C., Abogadie, F., et al. (2011).
Heteromerization of angiotensin receptors changes trafficking and arrestin recruitment
profiles. Cellular Signalling, 23, 1767e1776.
Prinster, S. C., Hague, C., & Hall, R. A. (2005). Heterodimerization of G protein-coupled receptors: specificity and functional significance. Pharmacological Reviews, 57, 289e298.
Qin, K., Dong, C., Wu, G., & Lambert, N. A. (2011). Inactive-state preassembly of G(q)coupled receptors and G(q) heterotrimers. Nature Chemical Biology, 7, 740e747.
Rahmeh, R., Damian, M., Cottet, M., Orcel, H., Mendre, C., Durroux, T., et al. (2012). Structural insights into biased G protein-coupled receptor signaling revealed by fluorescence

ARTICLE IN PRESS

References

spectroscopy. Proceedings of the National Academy of Sciences of the United States of


America, 109, 6733e6738.
Rajagopal, S., Rajagopal, K., & Lefkowitz, R. J. (2010). Teaching old receptors new tricks:
biasing seven-transmembrane receptors. Nature Reviews Drug Discovery, 9, 373e386.
Rasmussen, S. G., DeVree, B. T., Zou, Y., Kruse, A. C., Chung, K. Y., Kobilka, T. S., et al.
(2011). Crystal structure of the beta2 adrenergic receptor-Gs protein complex. Nature,
477, 549e555.
Rebois, R. V., Robitaille, M., Petrin, D., Zylbergold, P., Trieu, P., & Hebert, T. E. (2008).
Combining protein complementation assays with resonance energy transfer to detect multipartner protein complexes in living cells. Methods, 45, 214e218.
Reiter, E., & Lefkowitz, R. J. (2006). GRKs and beta-arrestins: roles in receptor silencing,
trafficking and signaling. Trends in Endocrinology and Metabolism, 17, 159e165.
Rivero-Muller, A., Chou, Y.-Y., Ji, I., Lajic, S., Hanyaloglu, A. C., Jonas, K., et al. (2010).
Rescue of defective G proteinecoupled receptor function in vivo by intermolecular
cooperation. Proceedings of the National Academy of Sciences of the United States of
America, 107, 2319e2324.
Robinson, K. H., Yang, J. R., & Zhang, J. (2014). FRET and BRET-based biosensors in live
cell compound screens. Methods in Molecular Biology, 1071, 217e225.
Robitaille, M., Heroux, I., Baragli, A., & Hebert, T. E. (2009). Novel tools for use in bioluminescence resonance energy transfer (BRET) assays. Methods in Molecular Biology,
574, 215e234.
Roda, A., Guardigli, M., Pasini, P., & Mirasoli, M. (2003). Bioluminescence and chemiluminescence in drug screening. Analytical and Bioanalytical Chemistry, 377, 826e833.
Roed, S. N., Nohr, A. C., Wismann, P., Iversen, H., Brauner-Osborne, H., Knudsen, S. M.,
et al. (2015). Functional consequences of glucagon-like peptide-1 receptor cross-talk
and trafficking. The Journal of Biological Chemistry, 290, 1233e1243.
Roed, S. N., Wismann, P., Underwood, C. R., Kulahin, N., Iversen, H., Cappelen, K. A., et al.
(2014). Real-time trafficking and signaling of the glucagon-like peptide-1 receptor.
Molecular and Cellular Endocrinology, 382, 938e949.
Roka, F., Brydon, L., Waldhoer, M., Strosberg, A. D., Freissmuth, M., Jockers, R., et al.
(1999). Tight association of the human Mel(1a)-melatonin receptor and G(i): precoupling
and constitutive activity. Molecular Pharmacology, 56, 1014e1024.
Rovira, X., Pin, J. P., & Giraldo, J. (2010). The asymmetric/symmetric activation of GPCR
dimers as a possible mechanistic rationale for multiple signalling pathways. Trends in
Pharmacological Sciences, 31, 15e21.
Salahpour, A., Espinoza, S., Masri, B., Lam, V., Barak, L., & Gainetdinov Raul, R. (2012).
BRET biosensors to study GPCR biology, pharmacology and signal transduction. Frontiers in Endocrinology (Lausanne), 3.
Schelshorn, D., Joly, F., Mutel, S., Hampe, C., Breton, B., Mutel, V., et al. (2012).
Lateral allosterism in the glucagon receptor family: glucagon-like peptide 1 induces
g-protein-coupled receptor heteromer formation. Molecular Pharmacology, 81,
309e318.
Schlyer, S., & Horuk, R. (2006). I want a new drug: G-protein-coupled receptors in drug
development. Drug Discovery Today, 11, 481e493.
See, H. B., Seeber, R. M., Kocan, M., Eidne, K. A., & Pfleger, K. D. (2011). Application of
G protein-coupled receptor-heteromer identification technology to monitor beta-arrestin
recruitment to G protein-coupled receptor heteromers. Assay and Drug Development
Technologies, 9, 21e30.

35

ARTICLE IN PRESS

36

Resonance Energy Transfer

Seifert, R., & Wenzel-Seifert, K. (2002). Constitutive activity of G-protein-coupled receptors:


cause of disease and common property of wild-type receptors. Naunyn Schmiedebergs
Archieves of Pharmacology, 366, 381e416.
Selvin, P. R. (2000). The renaissance of fluorescence resonance energy transfer. Nature Structural Biology, 7, 730e734.
Senogles, S. E., Benovic, J. L., Amlaiky, N., Unson, C., Milligan, G., Vinitsky, R., et al.
(1987). The D2-dopamine receptor of anterior pituitary is functionally associated with
a pertussis toxin-sensitive guanine nucleotide binding protein. The Journal of Biological
Chemistry, 262, 4860e4867.
Sexton, M., Woodruff, G., Horne, E. A., Lin, Y. H., Muccioli, G. G., Bai, M., et al. (2011).
NIR-mbc94, a fluorescent ligand that binds to endogenous CB(2) receptors and is
amenable to high-throughput screening. Chemistry and Biology, 18, 563e568.
Shenoy, S. K., & Lefkowitz, R. J. (2003). Trafficking patterns of beta-arrestin and G proteincoupled receptors determined by the kinetics of beta-arrestin deubiquitination. The Journal of Biological Chemistry, 278, 14498e14506.
Shenoy, S. K., & Lefkowitz, R. J. (2011). Beta-arrestin-mediated receptor trafficking and
signal transduction. Trends in Pharmacological Sciences, 32, 521e533.
Shukla, A. K., Violin, J. D., Whalen, E. J., Gesty-Palmer, D., Shenoy, S. K., & Lefkowitz, R. J.
(2008). Distinct conformational changes in beta-arrestin report biased agonism at seventransmembrane receptors. Proceedings of the National Academy of Sciences of the United
States of America, 105, 9988e9993.
Simon, M. I., Strathmann, M. P., & Gautam, N. (1991). Diversity of G proteins in
transduction. Science, 252, 802e808.
Smith, S. K., & Limbird, L. E. (1981). Solubilization of human platelet alpha-adrenergic receptors: evidence that agonist occupancy of the receptor stabilizes receptoreeffector
interactions. Proceedings of the National Academy of Sciences of the United States of
America, 78, 4026e4030.
Stadel, J. M., Shorr, R. G., Limbird, L. E., & Lefkowitz, R. J. (1981). Evidence that a betaadrenergic receptor-associated guanine nucleotide regulatory protein conveys guanosine
50 -O-(3-thiotriphosphate)- dependent adenylate cyclase activity. The Journal of Biological
Chemistry, 256, 8718e8723.
Stoddart, L. A., Johnstone, E. K., Wheal, A. J., Goulding, J., Robers, M. B., Machleidt, T.,
et al. (2015). Application of BRET to monitor ligand binding to GPCRs. Nature Methods,
12, 661e663.
Stoddart, L. A., Vernall, A. J., Denman, J. L., Briddon, S. J., Kellam, B., & Hill, S. J. (2012).
Fragment screening at adenosine-A(3) receptors in living cells using a fluorescence-based
binding assay. Chemistry and Biology, 19, 1105e1115.
Stoddart, L. A., White, C. W., Nguyen, K., Hill, S. J., & Pfleger, K. D. (2015). Fluorescenceand bioluminescence-based approaches to study GPCR ligand binding. British Journal of
Pharmacology (in press).
Strange, P. G. (2008a). Agonist binding, agonist affinity and agonist efficacy at G proteincoupled receptors. British Journal of Pharmacology, 153, 1353e1363.
Strange, P. G. (2008b). Signaling mechanisms of GPCR ligands. Current Opinion in Drug
Discovery and Development, 11, 196e202.
Szalai, B., Hoffmann, P., Prokop, S., Erdelyi, L., Varnai, P., & Hunyady, L. (2014). Improved
methodical approach for quantitative BRET analysis of G Protein Coupled Receptor
dimerization. PLoS One, 9, e109503.

ARTICLE IN PRESS

References

Szidonya, L., Cserzo, M., & Hunyady, L. (2008). Dimerization and oligomerization of
G-protein-coupled receptors: debated structures with established and emerging functions.
Journal of Endocrinology, 196, 435e453.
Tahtaoui, C., Guillier, F., Klotz, P., Galzi, J. L., Hibert, M., & Ilien, B. (2005). On the use of
nonfluorescent dye labeled ligands in FRET-based receptor binding studies. Journal of
Medicinal Chemistry, 48, 7847e7859.
Tateyama, M., Abe, H., Nakata, H., Saito, O., & Kubo, Y. (2004). Ligand-induced rearrangement of the dimeric metabotropic glutamate receptor 1alpha. Nature Structural and
Molecular Biology, 11, 637e642.
Tenenbaum, J., Ayoub, M. A., Perkovska, S., Adra-Delenne, A. L., Mendre, C., Ranchin, B.,
et al. (2009). The constitutively active V2 receptor mutants conferring NSIAD are weakly
sensitive to agonist and antagonist regulation. PLoS One, 4, e8383.
Terrillon, S., & Bouvier, M. (2004a). Receptor activity-independent recruitment of betaarrestin2 reveals specific signalling modes. The EMBO Journal, 23, 3950e3961.
Terrillon, S., & Bouvier, M. (2004b). Roles of G-protein-coupled receptor dimerization.
EMBO Reports, 5, 30e34.
Tian, W. N., Duzic, E., Lanier, S. M., & Deth, R. C. (1994). Determinants of alpha 2-adrenergic
receptor activation of G proteins: evidence for a precoupled receptor/G protein state.
Molecular Pharmacology, 45, 524e531.
Trinquet, E., Bouhelal, R., & Dietz, M. (2011). Monitoring Gq-coupled receptor response
through inositol phosphate quantification with the IP-One assay. Expert Opinion on
Drug Discovery, 6, 981e994.
Trinquet, E., Fink, M., Bazin, H., Grillet, F., Maurin, F., Bourrier, E., et al. (2006). D-myoinositol 1-phosphate as a surrogate of D-myo-inositol 1,4,5-tris phosphate to monitor
G protein-coupled receptor activation. Analytical Biochemistry, 358, 126e135.
Urban, J. D., Clarke, W. P., von Zastrow, M., Nichols, D. E., Kobilka, B., Weinstein, H., et al.
(2007). Functional selectivity and classical concepts of quantitative pharmacology.
Journal of Pharmacology and Experimental Therapeutics, 320, 1e13.
Urizar, E., Yano, H., Kolster, R., Gales, C., Lambert, N., & Javitch, J. A. (2011). CODA-RET
reveals functional selectivity as a result of GPCR heteromerization. Nature Chemical
Biology, 7, 624e630.
Vernall, A. J., Hill, S. J., & Kellam, B. (2013). The evolving small-molecule fluorescentconjugate toolbox for class a GPCRs. British Journal of Pharmacology, 171, 1073e1084.
Vilardaga, J. P., Bunemann, M., Feinstein, T. N., Lambert, N., Nikolaev, V. O., Engelhardt, S.,
et al. (2009). GPCR and G proteins: drug efficacy and activation in live cells. Molecular
Endocrinology, 23, 590e599.
Vilardaga, J. P., Bunemann, M., Krasel, C., Castro, M., & Lohse, M. J. (2003). Measurement
of the millisecond activation switch of G protein-coupled receptors in living cells. Nature
Biotechnology, 21, 807e812.
Vilardaga, J. P., Nikolaev, V. O., Lorenz, K., Ferrandon, S., Zhuang, Z., & Lohse, M. J. (2008).
Conformational cross-talk between alpha2A-adrenergic and mu-opioid receptors controls
cell signaling. Nature Chemical Biology, 4, 126e131.
Violin, J. D., Ren, X. R., & Lefkowitz, R. J. (2006). G-protein-coupled receptor kinase
specificity for beta-arrestin recruitment to the beta2-adrenergic receptor revealed by
fluorescence resonance energy transfer. The Journal of Biological Chemistry, 281,
20577e20588.
Vischer, H. F., Castro, M., & Pin, J. P. (2015). G protein-coupled receptor multimers: a question
still open despite the use of novel approaches. Molecular Pharmacology, 88, 561e571.

37

ARTICLE IN PRESS

38

Resonance Energy Transfer

Vrecl, M., Jorgensen, R., Pogacnik, A., & Heding, A. (2004). Development of a BRET2
screening assay using beta-arrestin 2 mutants. Journal of Biomolecular Screening, 9,
322e333.
Waldhoer, M., Fong, J., Jones, R. M., Lunzer, M. M., Sharma, S. K., Kostenis, E., et al. (2005).
A heterodimer-selective agonist shows in vivo relevance of G protein-coupled receptor
dimers. Proceedings of the National Academy of Sciences of the United States of America,
102, 9050e9055.
Ward, R. J., Pediani, J. D., & Milligan, G. (2011). Ligand-induced internalization of the orexin
OX(1) and cannabinoid CB(1) receptors assessed via N-terminal SNAP and CLIPtagging. British Journal of Pharmacology, 162, 1439e1452.
Watts, A. O., van Lipzig, M. M., Jaeger, W. C., Seeber, R. M., van Zwam, M., Vinet, J., et al.
(2013). Identification and profiling of CXCR3-CXCR4 chemokine receptor heteromer
complexes. British Journal of Pharmacology, 168, 1662e1674.
Wehbi, V. L., Stevenson, H. P., Feinstein, T. N., Calero, G., Romero, G., & Vilardaga, J. P.
(2013). Noncanonical GPCR signaling arising from a PTH receptor-arrestin-Gbetagamma
complex. Proceedings of the National Academy of Sciences of the United States of
America, 110, 1530e1535.
Whitaker, M. (2010). Genetically encoded probes for measurement of intracellular calcium.
Methods in Cell Biology, 99, 153e182.
Williams, C., & Hill, S. J. (2009). GPCR signaling: understanding the pathway to successful
drug discovery. Methods in Molecular Biology, 552, 39e50.
Wise, A., Gearing, K., & Rees, S. (2002). Target validation of G-protein coupled receptors.
Drug Discovery Today, 7, 235e246.
Wootten, D., Christopoulos, A., & Sexton, P. M. (2013). Emerging paradigms in GPCR allostery: implications for drug discovery. Nature Reviews Drug Discovery, 12, 630e644.
Xu, C., Peter, M., Bouquier, N., Ollendorff, V., Villamil, I., Liu, J., et al. (2013). REV, a
BRET-based sensor of ERK activity. Frontiers in Endocrinology (Lausanne), 4, 95.
Xu, X., Soutto, M., Xie, Q., Servick, S., Subramanian, C., von Arnim, A. G., et al. (2007).
Imaging protein interactions with bioluminescence resonance energy transfer (BRET)
in plant and mammalian cells and tissues. Proceedings of the National Academy of
Sciences of the United States of America, 104, 10264e10269.
Yi, T. M., Kitano, H., & Simon, M. I. (2003). A quantitative characterization of the yeast heterotrimeric G protein cycle. Proceedings of the National Academy of Sciences of the
United States of America, 100, 10764e10769.
Zhang, J., Campbell, R. E., Ting, A. Y., & Tsien, R. Y. (2002). Creating new fluorescent
probes for cell biology. Nature Reviews Molecular Cell Biology, 3, 906e918.
Zheng, Y., Akgun, E., Harikumar, K. G., Hopson, J., Powers, M. D., Lunzer, M. M., et al.
(2009). Induced association of mu opioid (MOP) and type 2 cholecystokinin (CCK2) receptors by novel bivalent ligands. Journal of Medicinal Chemistry, 52, 247e258.
Zurn, A., Zabel, U., Vilardaga, J. P., Schindelin, H., Lohse, M. J., & Hoffmann, C. (2009).
Fluorescence resonance energy transfer analysis of alpha 2a-adrenergic receptor activation reveals distinct agonist-specific conformational changes. Molecular Pharmacology,
75, 534e541.
Zwier, J. M., Roux, T., Cottet, M., Durroux, T., Douzon, S., Bdioui, S., et al. (2010). A fluorescent ligand-binding alternative using Tag-lite(R) technology. Journal of Biomolecular
Screening, 15, 1248e1259.

S-ar putea să vă placă și