ArticlePDF Available

Allosteric communication between protomers of dopamine Class A GPCR dimers modulates activation

Authors:

Abstract and Figures

A major obstacle to understanding the functional importance of dimerization between class A G protein-coupled receptors (GPCRs) has been the methodological limitation in achieving control of the identity of the components comprising the signaling unit. We have developed a functional complementation assay that enables such control, and we demonstrate it here for the human dopamine D2 receptor. The minimal signaling unit, two receptors and a single G protein, is maximally activated by agonist binding to a single protomer, which suggests an asymmetrical activated dimer. Inverse agonist binding to the second protomer enhances signaling, whereas agonist binding to the second protomer blunts signaling. Ligand-independent constitutive activation of the second protomer also inhibits signaling. Thus, GPCR dimer function can be modulated by the activity state of the second protomer, which for a heterodimer may be altered in pathological states. Our new methodology also makes possible the characterization of signaling from a defined heterodimer unit.
Content may be subject to copyright.
Allosteric communication between protomers of dopamine
Class A GPCR dimers modulates activation
Yang Han1,2,3, Irina S. Moreira4, Eneko Urizar1,2,3, Harel Weinstein4,5, and Jonathan A.
Javitch1,2,3,6
1Center for Molecular Recognition, 630 West 168th Street, New York, NY 10032, USA
2Department of Psychiatry, Columbia University College of Physicians and Surgeons, 630 West
168th Street, New York, NY 10032, USA
3Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032
USA
4Department of Physiology and Biophysics, Weill Medical College of Cornell University, 1300
York Ave, New York, NY 10065, USA.
5The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational
Biomedicine, Weill Medical College of Cornell University, 1300 York Ave, New York, NY 10065,
USA.
6Department of Pharmacology, Columbia University College of Physicians and Surgeons, 630
West 168th Street, New York, NY 10032
Abstract
A major obstacle to understanding the functional importance of dimerization between Class A G
protein-coupled receptors (GPCRs) has been the methodological limitation in achieving control of
the identity of the components comprising the signaling unit. We have developed a
functional
complementation assay
that enables such control and illustrate it for the human dopamine D2
receptor. The minimal signaling unit, two receptors and a single G protein, is maximally activated
by agonist binding to a single protomer, which suggests an asymmetrical activated dimer. Inverse
agonist binding to the second protomer enhances signaling, whereas agonist binding to the second
protomer blunts signaling. Ligand-independent constitutive activation of the second protomer also
inhibits signaling. Thus, GPCR dimer function can be modulated by the activity state of the
second protomer, which for a heterodimer may be altered in pathological states. Our novel
methodology also makes possible the characterization of signaling from a defined heterodimer
unit.
The most intriguing questions about the functional mechanisms of G protein-coupled
receptors (GPCRs) center on the role of dimerization, its physiological significance, and its
pharmacological consequences1. Many results from the literature are tantalizing because
they describe effects that have been attributed to activating one receptor in the presence of
another2 and the ability to modulate activity of one receptor by ligands targeting the second
receptor3,4. Compelling as these examples are, it has thus far been difficult to construct a
mechanism that would coherently explain all these phenomena. For most GPCRs, a major
obstacle has been methodological, especially the inability to control the identity of the
components of the G protein signaling unit that must include the interacting receptors and G
proteins. Here we present a mechanism for rhodopsin-like Class A GPCRs that we were able
Correspondence should be addressed to J.A.J (jaj2@columbia.edu).
NIH Public Access
Author Manuscript
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
Published in final edited form as:
Nat Chem Biol
. 2009 September ; 5(9): 688–695. doi:10.1038/nchembio.199.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
to identify using a novel approach that enabled us to control the identity of the participants
in the signaling complex.
In Class C GPCRs such control has been possible because of the unique cell biology of the
GABAB receptor. The R2 subunit does not signal by itself in response to GABA (1), but is
essential for surface expression of the R1 subunit and therefore for signaling of the
heterodimeric complex5. Therefore, the only species on the surface that can signal must
contain R1 and R2, which allows the study of defined heterodimers. These receptors have
been shown to function through a “transactivation” mechanism in which a GABA-binding
R1 signals through interactions of R2 with G protein5. A clever adaptation of the
endoplasmic reticulum (ER) retention signal from the GABAB receptor has enabled
controlled cell surface expression and study of signaling by defined metabotropic glutamate
receptor (mGluR) “hetero”-dimers6, which have been inferred to signal through
trans
-
activation as well as through
cis
-activation, in which the agonist-bound receptor interacting
directly with G protein6. Such an approach to engineered ER retention signals has not yet
been successful in Class A receptors, but Class A glycoprotein hormone receptors with large
extracellular binding domains also appear to be capable of both trans- and cis-activation7.
To date the native functional signaling unit in other Class A rhodopsin-like receptors
remains unclear. Indeed, both rhodopsin8 and the β2-adrenergic receptor (B2AR)9 have
been shown to signal efficiently to G proteins when reconstituted into lipid nanodiscs
containing only a single receptor. Thus, after solubilization and reconstitution, these GPCRs
can
function alone. Nevertheless, such studies cannot determine whether these receptors do
function alone
in vivo
, and this question must be addressed directly through an exploration
of signaling in their native organization. A number of studies have shown that coexpression
of two different Class A GPCRs can lead to signaling properties that differ from their
properties when expressed alone10,11. However, it is not possible from such studies to
differentiate downstream integration of signaling from an actual heteromeric signaling unit
in which the two protomers interact directly to modulate signaling.
Evidence for association of conformational change at a homodimer interface with activation
state12 supports state-dependent communication between protomers and a potential role for
inter-protomer modulation of signaling. However, in contrast to findings for the Class C
GABAB and mGlu receptors and the Class A receptors with large N-terminal binding sites,
TSH, FSH, and LH receptors, results for the rhodopsin-like leukotriene B4 receptor BLT1
support the existence of
cis
- but not
trans
-activation, with no functional role identified for
the second protomer, despite evidence that it changes conformation in response to agonist
binding to its dimer partner13,14.
Receptor-G protein fusion constructs, in which the C-terminus of a GPCR is fused to the N-
terminus of Gα, have been used to explore receptor signaling1518. Coexpression of such
GPCR-G protein fusions with a second GPCR has been used to study heterodimer signaling;
in such a scenario the unfused GPCR can activate the G protein fused to a coexpressed
GPCR1517. However, the participants in the signaling unit are not identifiable in this
experimental protocol because coexpression of GPCRs leads to a combination of different
signaling units consisting of both homodimers and heterodimers. Indeed, a tethered G
protein fused to a single membrane-spanning segment can be activated efficiently by a
coexpressed GPCR16,19, suggesting that a GPCR-G protein fusion construct might provide
G protein for activation by another receptor, or another dimer of receptors, without directly
participating in the relevant dimeric signaling unit. The long cytoplasmic tails and flexible
linkers through which G proteins have been fused to GPCRs are likely to allow promiscuous
interactions that exacerbate this problem. Indeed, the tether attaching the B2AR to fused Gs
can be dramatically shortened with preserved function20, but it remains unknown whether
Han et al. Page 2
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
the G protein in this case is activated by the receptor to which it is covalently attached, or by
another receptor.
Here, we have developed a
functional complementation assay
that allows us to control the
components of the human dopamine D2 receptor (D2R) signaling unit and thus to explore
the dimeric functional unit and the individual contributions from each GPCR protomer to G
protein signaling. Our system reports directly on receptor-G protein interactions, which
allows us to rule out downstream crosstalk as the mechanism of modulation of G protein
function upon coexpression of different partner receptors. This novel methodology allowed
us to propose a mechanistic explanation for the reciprocal modulation of protomer functions
in a dimeric signaling complex. The minimal signaling unit, consisting of two GPCRs and a
single heterotrimeric G protein, appears to be maximally activated by agonist binding to a
single protomer, which suggests an asymmetrical activated dimer. Indeed, agonist binding to
the second protomer blunts signaling, whereas inverse agonist binding to the second
protomer enhances signaling. Such allosteric modulation of one protomer by the state of the
other also has important ramifications for pharmacological manipulation of GPCR
heterodimers. That a non-binding constitutively active receptor blunts signaling of a
coexpressed wild type (WT) receptor highlights the importance of the conformational state
of the second protomer. Therefore, GPCR heterodimer function will be modulated not only
by ligand binding to the second protomer, but also by its ligand-independent constitutive
activity; both types of modulation may be altered in pathological states.
Results
Engineering a luminescence readout for D2R activation
To isolate signaling of the D2R, a prototypical Go/Gi coupled receptor, from endogenous G
proteins and to control each of the components of the signaling complex, we engineered Flp-
In T-REx-293 cells to stably express aequorin (AEQ cells) (see Methods). Aequorin
produces luminescence in a calcium-dependent manner in the presence of the substrate
coelenterazine21 (2), and it has been used to create a sensitive luminescence readout for
GPCR-mediated PLC activation22. In these cells, endogenous muscarinic or purinergic
receptors signaled robustly via endogenous Gq, resulting in strong agonist-induced (ACH
(3) and ATP (4), respectively) luminescence signals (Supplementary Fig. 1a online). In
contrast, when D2R was stably expressed in AEQ cells, treatment with the aqonist
quinpirole (5) did not lead to luminescence, consistent with a lack of D2R coupling to Gq
(Fig. 1a, Supplementary Fig. 2 online).
To couple D2R activation to a luminescence readout in these cells, we expressed a chimeric,
pertussis toxin (PTX)-resistant Gq (Gqi5) that could signal from Gi-coupled receptors23
(see Methods). D2R signaled robustly when stably coexpressed with free Gqi5 or when
fused at its C-terminus to Gqi5 through an 8 amino acid linker (D2-linker-Gqi5) (Fig. 1b, c).
The increase in luminescence was unaffected by PTX (Supplementary Fig. 1b online),
whereas a mutation in Gqi5 (Gqi5G208A) that prevents GTP-induced Gα activation24
prevented the luminescence response to D2R activation (Fig. 1c). No quinpirole response
was seen when free Gqi5 was expressed without D2R (data not shown), consistent both with
the absence of endogenous D2R in these cells and with the lack of other targets for
quinpirole-mediated signaling.
Curiously, expression of free Gqi5 fully rescued function of D2-linker-Gqi5G208A (Fig. 1d),
indicating that the linker afforded sufficient flexibility for the nonfunctional G protein to
swing away and permit a free functional Gqi5 to interact and to restore agonist-mediated
signaling. Therefore, we could not use the D2-linker-Gqi5 construct to monitor functional
coupling of two defined protomers, since the flexibility of the linker might allow this
Han et al. Page 3
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
construct to provide the Gα to another protomer (or to another dimer of protomers) without
the actual participation of the fused receptor in the signaling unit.
To address this problem, we developed another D2R/Gqi5 construct in which the linker was
removed and Gqi5 was fused more directly to the short cytoplasmic tail of the D2R (D2-
Gqi5). This construct expressed at the plasma membrane (Supplementary Fig. 3a, b online),
but agonist treatment failed to produce luminescence (Fig. 1e). We hypothesized that the
lack of signaling resulted from the inability of the short-tethered Gα to be positioned
appropriately for a productive interaction either with the cytoplasmic loops of the receptor to
which it was fused, or with a second protomer. Indeed, in contrast to the D2-linker-Gqi5,
D2-Gqi5 signaling was not rescued by free Gqi5 (Fig. 1e), most likely because the tethered
Gα sterically blocks free Gqi5 from making a productive interaction with the cytoplasmic
loops of the fused receptor.
Remarkably, however, coexpression in the AEQ cells of D2R (termed “Protomer A”) and
D2-Gqi5 (“Protomer B”), each of which is incapable of signaling in our assay when
expressed alone, led to robust agonist-mediated receptor activation (Fig. 1f), indicating that
when activated the fused Gqi5 is fully capable of interacting with PLC. That this effect was
mediated solely by the fused Gqi5 and not by endogenous Gi/o was demonstrated by the
lack of effect of PTX treatment on activation (Supplementary Fig. 1c online). This
reconstitution of a signaling unit from two “nonfunctioning” Class A GPCR protomers
provided us with the unique opportunity to manipulate each protomer independently and to
determine its role in signaling while eliminating the contribution of homodimers. These
experiments do not rule out a higher order receptor complex, but in the simplest scenario the
minimal signaling unit is composed of protomer A, protomer B and the G protein fused to
protomer B (Fig. 1f), and for simplicity we will subsequently refer to this receptor complex
as a “dimer”. The extremely close proximity between these protomers and the inability of
protomer B to signal to its own fused G protein or to a neighboring fused G protein indicates
that only one G protein serves this signaling unit of two GPCRs. Our inferences regarding
the signaling unit are entirely consistent with the results from our parallel computational
modeling studies (see below). These modeling studies make the essential point that the
relatively large size of the G protein heterotrimer matches the cytoplasmic surfaces of at
least two neighboring GPCR protomers.
Revealing asymmetry of signaling
In order to manipulate experimentally the function of each protomer in the dimeric unit, we
constructed a panel of D2R mutants predicted to be binding- and activationdeficient based
on findings in the literature for related Class A GPCRs. These include D1143.32A, which
does not bind agonists or antagonists25, as well as R1323.50A26 and V1363.54D/M1403.58E
in IL227, deletion of 213–219 in IL328, and D802.50A (previously characterized in D2R)29
and N3937.49A30 in the membrane-spanning segments (Fig. 2a), all of which were expected
to disrupt agonist-mediated G protein activation. We also expressed a D2R mutant
V912.61F/F1103.29L/V1113.28M/Y4087.35V (termed D2/D4) (Fig. 2a), which, unlike WT
D2R, is potently inhibited by the selective D4 antagonist L745,87031 (6) (Supplementary
Fig. 4 online). Each of these constructs expressed at the plasma membrane (Supplementary
Fig. 3 online). In contrast to the robust activation of WT, we observed a reduction in potency
and a large decrease in maximal activation by quinpirole in D2/D4 when expressed with free
Gqi5 (Fig. 2b). As anticipated, none of the mutants deficient in binding or signaling led to
agonist-mediated luminescence when placed into an unfused D2R construct coexpressed
with free Gqi5 (Fig. 2c), or when the mutations were placed in the D2-linker-Gqi5 construct
and expressed alone (Supplementary Fig. 5 online).
Han et al. Page 4
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
When D2/D4 was expressed as protomer A with WT D2R-Gqi5 as protomer B, we observed
a reduction in potency and a large decrease in maximal activation by quinpirole (Fig. 3a),
similar to its signaling properties when expressed with free Gqi5 (Fig. 2b). (These and all
subsequent activation data were normalized for surface expression of protomer B; see
Methods, Supplementary Fig. 3, Supplementary Fig. 6 online). Expression of any of the
nonbinding or nonsignaling receptor mutants as protomer A completely prevented activation
(Fig. 3b), despite the presence of WT D2R-Gqi5 in protomer B.
Thus, protomer A, which
must interact with the Gα provided by protomer B, appears to play a dominant role in the
activation process
. Note that the absence of trans-activation was not a result of our
functional complementation system or lack of sufficient mobility of fused G protein; we also
failed to see evidence for trans-activation even when nonbinding and noncoupling receptors
(without G protein fusions) were co-expressed with free Gqi5 (data not shown).
In contrast, we observed robust agonist-mediated activation with WT D2R as protomer A
and D2/D4-Gqi5 (Fig. 3c) or D114A-Gqi5 (Fig. 3d) as protomer B. These data suggest that
agonist binding to protomer A is sufficient for normal activation (see below) and imply an
asymmetric organization of the signaling complex comprising two GPCR protomers with G
protein. When R1323.50A-Gqi5 or V1363.54D/M1403.58E-Gqi5 was expressed as protomer B
with WT D2R as protomer A, no activation was observed (Fig. 3e). In contrast, although the
IL3 deletion construct abolished activation when placed in protomer A (Fig. 3b), it
supported signaling when coexpressed as protomer B (fused to Gqi5) along with WT D2 as
protomer A (Fig. 3d).
These data support a mechanism in which two GPCRs activate a
single G protein through interactions that involve IL2 from both protomers whereas IL3
from only one protomer is essential for signaling
. Note that the failure of R1323.50A-Gqi5 or
V1363.54D/M1403.58E-Gqi5 to function with WT is not due to an inability of these
protomers to interact, since we observed efficient bioluminescence resonance energy
transfer as well as bimolecular luminescence and fluorescence complementation32 between
these mutants and WT D2R (Supplementary Fig.7 online).
To explore the nature of the conformational changes in the dimeric receptor unit we also
studied inactivating mutations within the membrane-spanning segments. The transduction-
uncoupling mutants D802.50A and N3937.49A revealed additional differences in the roles of
protomers A and B. When either of these mutations was placed in protomer A, signaling was
abolished, consistent with the dominant role of this protomer (Fig. 3b). In contrast, when
placed in protomer B, D802.50A-Gqi5 signaled when coexpressed with WT D2R as
protomer A (Fig. 3d), whereas N3937.49A-Gqi5 did not (Fig. 3e).
These results suggest that
the nature of the conformational changes in protomer B during activation differs from those
in protomer A
.
The activation state of Protomer B modulates signaling
Agonist binding to only protomer A and not protomer B produced full activation, as
coexpressed D2R and D1143.32A-Gqi5 were robustly activated by quinpirole (Fig. 3d).
Moreover, it appeared that binding of a second agonist to protomer B might inhibit
signaling, as coexpression of both WT D2R and D2R-Gqi5 led to lower maximal activation
than did D2R coexpressed with D1143.32A-Gqi5 (Fig. 3d). We tested this hypothesis using
the D2/D4 chimeric receptor. As predicted, when D2/D4 was expressed as protomer A with
D2R-Gqi5 as protomer B, quinpirole’s ability to bind and activate was blocked by the D4-
selective antagonist L745,870, reflecting the primacy of protomer A (Fig. 4a). In contrast,
coexpressing D2/D4-Gqi5 as protomer B with D2R as protomer A led to robust receptor
activation that was greater than that seen with WT D2R and D2R-Gqi5 (Fig. 3c) and was
further enhanced in the presence of L745,870 (Fig. 4b), which blocks quinpirole binding to
protomer B but not protomer A.
Han et al. Page 5
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
It is the active conformation of the second protomer that inhibits signaling, and not agonist
binding
per se
. This is evidenced by the finding that activating protomer B by constitutively
activating mutations (Supplementary Fig. 8 online) in a nonbinding receptor (D1143.32A/
E3396.30A/T3436.34R-Gqi5)26,33 substantially reduced the signaling efficacy of a WT
protomer A (Fig. 4c).
Thus, activation of the second protomer, either by ligand binding or by
its inherent constitutive activity, inhibits signaling by its partner
.
Computational modeling of the D2R-G protein interface
Because our experimental findings require a structural context in which the new
mechanisms for GPCR–G protein interactions emerging from this study can be understood,
we carried out independent computational studies that combined molecular modeling with
experimental data available in the literature about the modes of interaction of the component
GPCRs and G protein, but without direct reference to the new findings. We used
information available from the current crystal structures of GPCRs and specific data about
inactive and activated states for bovine rhodopsin, because, unlike the D2R, such a structural
template is accompanied by much experimental data about details of the sites and mode of
interaction with G protein to guide protein-protein docking. Thus, we took advantage of
experimental data from cross-linking, alanine scanning mutagenesis, and other structural and
functional studies of the GPCR–G protein interfaces pointing to several amino acid residues
likely to be involved in complex formation between rhodopsin and the G protein α- and βγ-
subunits (Supplementary Methods online). The data derived from the literature were used
not only as constraints to guide docking of transducin (Gt) to a variety of dimer models of
rhodopsin (Supplementary Fig. 9 online), but also to screen for the oligomerization solution
that satisfied best these constraints, as detailed in Methods.
Both TM4/TM5 and TM1 have been implicated in D2R oligomerization12,32,34. In order to
discriminate between a functional dimer with an interface involving TM4 and TM5 (named
TM4,5 dimer) from one with a TM1 interface (TM1 dimer), we docked the molecular
structure of transducin to a rhodopsin nonamer (Fig. 5a; Supplementary Fig. 9 online)
subject to specific constraints for the interaction between Gt and the central rhodopsin
(Supplementary Table 1 online). As described in Methods, transducin was free to rotate in
any direction around this central rhodopsin to select any one of the dimeric forms in the
array. The Gt could select a second monomer from the oligomeric structure in which the
GPCR interface corresponds to either a TM4, 5 interface, or a TM1 interface dimer. The
probability for Gt selecting either dimer interface was compared in a scan for optimal
interaction carried out on the oligomeric structure shown in (Fig. 5a) and (Supplementary
Fig. 9 online). The complexes resulting from this scan were considered acceptable (and
counted) only if the underlying structural models satisfied at least 50% of the
experimentally-based constraints (set 1 in Supplementary Table 2 online). A substantial
fraction of TM4, 5 dimers (21.1%) satisfied this cutoff, but no complex with a TM1 dimer
met the filtering criteria.
A possible mode of oligomer reorganization associated with function had been suggested
based on crosslinking studies in D2R34 and rhodopsin35. To evaluate the functional impact
of such a reorganization, Gt was docked to the TM4, 5 and TM4 dimer alternatives
(Supplementary Fig. 9 online). The Cα-Cα distances for specific interactions between
rhodopsin and transducin in the optimal representative structures of the 1000 structures
obtained for each alternative in this dimer docking procedure (see Methods for details) are
summarized in Supplementary Table 2 online. For these optimal structures the sets of Cα-
Cα distances are very similar, but the frequency of appearance of optimally positioned
complexes is much higher for Model 2 (TM4 dimer; 76.1%) than for Model 1 (TM4, 5
dimer; 23.8%). This is evident from the average values of the distances (Supplementary
Table 1), which are mostly larger in Model 1 than in Model 2 constructs. Thus, Model 2 is
Han et al. Page 6
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
considered the better representation of the GPCR dimer complex with the G protein in the
context of the oligomeric arrangement. This is consistent with the proposed transition from a
TM4, 5 interface to a TM4 interface upon activation, suggested by crosslinking results for
the D2R34, and indicates the relation between optimal G protein binding to the dimer and an
activated state.
Notably, in the optimal G protein–dimer complex the cytoplasmic ends of TM3 and IL2
from both protomers interact with the docked G protein. This is shown in (Fig. 5) for Model
2, but holds as well for Model 1. In contrast, only IL3 from protomer A, but not from
protomer B, contacts the docked Gα, consistent with our experimental results showing that
an inactivating IL3 mutation is tolerated in protomer B but not in protomer A.
Discussion
We find that agonist binding to a single protomer maximally activates a signaling unit
comprising two Class A GPCRs and a single G protein. Whereas activation of the second
protomer inhibits the functional response, inverse agonist binding to the second protomer
enhances signaling (Fig. 6). Our results are consistent with studies in the Class C mGluR
using allosteric modulators that act within the transmembrane region to show that the
inactive state of a protomer caused by inverse agonist binding results in more efficient
activation of the adjacent protomer36,37. These findings are more difficult to reconcile with
other findings in the mGluR showing that although one agonist can activate the dimeric
signaling unit, two agonists are required for full activation38, suggesting differences in the
mechanisms of these receptors, which have very different agonist binding sites. Our findings
are, however, fully consistent with the proposed function of GABAB receptors, in which
only R1 binds GABA39 with efficient signaling by the complex. A similar scenario also
seems likely for rhodopsin’s ability to respond to single photons, which requires robust
activation by a single protomer in a dimeric unit. Indeed in this case, the strong inverse
agonist 11-
cis
-retinal (7) in the binding pocket of second protomer would in fact optimize
signaling, just as we observe in configuration 4 in (Fig. 6). Our findings also suggest that
optimal signaling in a heteromeric GPCR would result from co-stimulation with an agonist
to one protomer and an inverse agonist to the other.
Our data and models suggest that the way in which the two protomers contribute to the
activated complex with the G protein is not symmetrical, and that activation requires
different conformational changes in each protomer. Existing evidence for ligand-induced
conformational changes in a second non-binding protomer13,40 is consistent with the
proposal of conformational changes in both protomers. We previously demonstrated an
activation-related conformational change at the TM4 dimer interface34 that also would be
consistent with movement of either one or both TM4s. Our present finding that transduction-
deficient mutants in different TMs differentially affect the ability of protomer B to rescue
function is consonant with the importance of conformational changes in this protomer.
Interestingly, the apparent negative cooperativity of ligand binding observed in a number of
class A GPCRs41 may well relate to this proposed asymmetry of the signaling unit. For
example, in cells expressing chemokine receptor heterodimers, a selective ligand for one
protomer leads to dissociation of ligand bound to the other protomer42, consistent with
transmission of an altered conformation across the dimer interface, and with a decreased
propensity for simultaneous agonist binding to both protomers.
In summary, although a single B2AR or rhodopsin molecule can efficiently activate G
protein when reconstituted into a nanodisc, a second protomer is present
in vivo
and
profoundly modulates G protein activation of the first protomer, as we have shown with our
functional complementation studies. Importantly, we show that this allosteric modulation of
Han et al. Page 7
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
signaling results from a direct interaction of the receptor dimer with the G protein, rather
than from a downstream effect. This is likely to explain many of the surprising observations
concerning the mutual modulation of heteromeric receptor oligomers by ligand binding to
one protomer or the other. Moreover, we demonstrate that the constitutive activity of a
protomer will modulate the activity of the dimeric signaling unit in which it participates.
Thus, inverse agonists at one protomer in a heterodimer are likely to be allosteric
potentiators of the signaling of its heterodimer partner, whereas agonists of one protomer
will be allosteric inhibitors of the second protomer, offering a mechanistic explanation for
the often befuddling observations regarding pharmacological effects of ligands acting on
heterodimers. Moreover, our model suggests that modulators might be found that are
specific for heterodimers and not homodimers, but heretofore it has not been possible to
screen for such compounds without the interference of homodimer-mediated signaling.
Indeed, it is possible that findings of functional selectivity, that is, different agonists for a
given receptor having different effects on different downstream effectors, might reflect
differential pharmacological effects on different heteromeric species43. The novel
methodology we present here makes it possible to identify signaling from a defined
heterodimer, and thus to identify modulators of heterodimer function. The modulatory
mechanism we characterized and the approach that made this possible offer a new
understanding of GPCR signaling in units composed of at least two GPCRs; applied to
specific systems the approach will make it possible to understand the effects of drugs that
target each protomer of such a signaling unit, either identical or different.
METHODS
Materials
The D2R agonist quinpirole hydrochloride and the D4R antagonist L745,870 (3-(4-[4-
Chlorophenyl]piperazin-1-yl)-methyl-1H-pyrrolo[ 2,3-b]pyridine trihydrochloride ) were
from Sigma-Aldrich.
DNA constructs
Expression plasmids expressing Signal Peptide Flag-tagged D2short WT44 and mutant
receptors were created using standard molecular biology procedures, as described in
Supplementary Methods online. Receptor constructs were fused directly through their C-
terminus, or through an 8 amino acid linker, to a PTX-resistant Gqi5 (Supplementary Fig. 2
online).
Cell culture and transfection
Flp-In T-REx 293 cells (Invitrogen) were cultured and transfected, and stable lines were
selected as described in (Supplementary Methods online).
Cell Surface Expression assay
An aliquot of the cell solution used for the aequorin assay (see below) was used to determine
receptor cell surface expression as described in Costagliola
et al
.45 with anti-Flag M2
(Sigma) or anti-Myc (Mt. Sinai hybridoma facility) as primary and R-phycoerythrin goat
anti-mouse IgG (Invitrogen) as secondary antibodies using a Guava Easycyte (Guava
technologies).
Aequorin assay
A functional assay based on luminescence of mitochondrial aequorin following intracellular
Ca++ release was performed21,46. Cells were seeded in a 15 cm dish and grown in
antibiotic-free medium for 48 hr until mid-log phase. 1 µg/ml tetracycline was added to the
Han et al. Page 8
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
medium for 3–24 hours prior to harvest to induce the expression of the transfected D2R in
pcDNA5/FRT/TO. Cells were dissociated and pelleted at 0.6 g for 3 min. After washing
once with DMEM medium (Invitrogen) supplemented with 0.1% BSA), cells were
resuspended in this medium at a final concentration of 5 × 106 cells/ml in the presence of 5
µM coelenterazine
h
. After 4 hr rotating at room temperature in the dark, the cell solution
was diluted 10-fold, followed by 1 hr incubation under the same conditions. Concentration-
response curves were obtained by injecting 50 µL of cell solution into wells of a 96-well
microplate containing 50 µL of a 2X concentration of the desired compound in medium.
Luminescence signals from the first 15 seconds after injection were read by a POLARstar
optima reader (BMG). Total response was defined as the signal resulting from injecting 50
µL cell solution into 50 µL assay medium containing 0.1% triton, which raises the Ca++
concentration directly by membrane permeabilization.
To normalize for different levels of surface expression levels of the Flag-D2R-Gqi5 mutant
constructs, the Emax at each expression level (Supplementary Fig. 6c online) was plotted as
a function of different levels of expression of Flag-tagged wt D2R-Gqi5, the expression of
which was controlled by varying the time after tetracycline induction (Supplementary Fig.
6a online). The level of Myc-D2R remained essentially unchanged (Supplementary Fig. 6b
online). The standard curve was fit to a 1 site rectangular hyperbola using nonlinear
regression in Graph Pad PRISM 4.0 (Supplementary Fig. 6d online). The luminescence
response of the various Flag-D2R-Gqi5 constructs was normalized using this standard curve
to account for the effects of different expression levels, with activation of 1 defined as that
observed after 12 hour of tetracycline induction of WT D2R-Gqi5. The Flag detection was
approximately 5-fold more sensitive than that of Myc; thus, the excess of Myc-tagged
protomer A, which cannot signal on its own, ensures that normalization based on surface
expression of the Flag-tagged Gqi5-fused protomer B accurately reflects the productive
signaling entities, each of which must contain a protomer A and a protomer B.
Model Construction
In the absence of experimentally determined structures of D2R and Gqi5 the templates for
the oligomeric model constructs were complexes between crystallographically determined
structures of rhodopsin and heterotrimeric G proteins, as described in Supplementary
Methods online. To enable the simultaneous probing of G protein interaction with different
dimer arrangements, we constructed a rhodopsin oligomer composed of nine monomers. The
activated form of the rhodopsin monomers used here was constructed by inclusion of all
constraints reported for rhodopsin, as described previously47. Three dimeric interfaces were
analyzed: Model 1, in which the dimers have a TM4,5 interface, Model 2, with a symmetric
TM4 interface34, and Model 3, in which the dimers have a TM1 interface32.
G-Protein-Rhodopsin Docking
The docking was carried out with the HADDOCK (High Ambiguity Driven protein-protein
DOCKing) software48,49. The docking process was driven by ambiguous interaction
restraints (AIRs)48 to both monomers, as described in Supplementary Methods online. The
constraints, established from literature-derived experimental data for the binding complex,
are given in Supplementary Table 1 online. Notably, the docking protocol of Gt to such
models using this set of constraints was verified by the full agreement with the complex
obtained for the recent structure of opsin50 representing a putative activated form of the
protein (Supplementary methods online). To select a second protomer for the complex,
another docking run was made with restraints only to the central rhodopsin, allowing
transducin to explore freely different orientations with respect to the rhodopsin oligomer. By
application of the docking protocol consisting of randomization of orientations and rigid
body energy minimization, 1000 different conformations were generated. These structures
Han et al. Page 9
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
were ranked according to their average interaction energies (sum of Eelec, Evdw, EAIR), and
screened using the eighteen constraints listed in (Supplementary Table 2 online), which
represent the information extracted from the experimental data and translated into Cα-Cα
intermolecular constraints (in Supplementary Table 2 online, set 1 refers to the interactions
of the C-terminus of Gα with protomer A, set 2 to the interactions between the N-terminus
of Gα and protomer A, and set 3 to the interactions between the N-terminus of Gα and
protomer B). Only Cα-Cα distances <20Å were interpreted as direct rhodopsin-Gt
interactions. A cutoff of 50% fulfillment of the of the interaction criteria was used for
accepting valid constructs. The relative probabilities of such valid G protein complexes with
the various model dimers (TM4; TM4,5; TM1) were calculated from the corresponding
percentages of acceptable complexes found in the resulting set of 1000 structures retrieved
from the docking procedure. The construct fulfilling the largest number of experimentally
derived constraints and with the N-terminal helix of Gα parallel to the cytoplasmic face of
the rhodopsin dimer, was chosen as the “optimal representative structure” for each model.
Supplementary Material
Refer to Web version on PubMed Central for supplementary material.
Acknowledgments
We thank Céline Gáles for discussion and comments on the manuscript. Plasmids encoding apoaequorin were a gift
from V. Dupriez (Euroscreen, Belgium). This work was supported in part by NIH grants DA022413 and
MH054137 (J.A.J.), DA012923 (HW), the Lieber Center for Schizophrenia Research and Treatment, and an EMBO
Long term fellowship (E.U.). Computational resources of the David A Cofrin Center for Biomedical Information in
the Institute for Computational Biomedicine are gratefully acknowledged.
Reference List
1. Pin JP, et al. International Union of Basic and Clinical Pharmacology. LXVII. Recommendations
for the Recognition and Nomenclature of G Protein-Coupled Receptor Heteromultimers. Pharmacol
Rev. 2007; 59:5–13. [PubMed: 17329545]
2. Sartania N, Appelbe S, Pediani JD, Milligan G. Agonist occupancy of a single monomeric element
is sufficient to cause internalization of the dimeric beta2-adrenoceptor. Cell Signal. 2007; 19:1928–
1938. [PubMed: 17561373]
3. Parenty G, Appelbe S, Milligan G. CXCR2 chemokine receptor antagonism enhances DOP opioid
receptor function via allosteric regulation of the CXCR2-DOP receptor heterodimer. Biochem J.
2008; 412:245–256. [PubMed: 18307412]
4. Vilardaga JP, et al. Conformational cross-talk between alpha2A-adrenergic and mu-opioid receptors
controls cell signaling. Nat Chem Biol. 2008; 4:126–131. [PubMed: 18193048]
5. Pin JP, Galvez T, Prezeau L. Evolution, structure, and activation mechanism of family 3/C G-
protein-coupled receptors. Pharmacology & Therapeutics. 2003; 98:325–354. [PubMed: 12782243]
6. Brock C, et al. Activation of a Dimeric Metabotropic Glutamate Receptor by Intersubunit
Rearrangement. J. Biol. Chem. 2007; 282:33000–33008. [PubMed: 17855348]
7. Ji IH, Lee C, Song YS, Conn PM, Ji TH. Cis-and trans-activation of hormone receptors: the LH
receptor. Mol Endocrinol. 2002; 16:1299–1308. [PubMed: 12040016]
8. Bayburt TH, Leitz AJ, Xie G, Oprian DD, Sligar SG. Transducin Activation by Nanoscale Lipid
Bilayers Containing One and Two Rhodopsins. J. Biol. Chem. 2007; 282:14875–14881. [PubMed:
17395586]
9. Whorton MR, et al. A monomeric G protein-coupled receptor isolated in a highdensity lipoprotein
particle efficiently activates its G protein. PNAS. 2007; 104:7682–7687. [PubMed: 17452637]
10. George SR, et al. Oligomerization of mu-and delta-opioid receptors-Generation of novel functional
properties. J. Biol. Chem. 2000; 275:26128–26135. [PubMed: 10842167]
Han et al. Page 10
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
11. Lee SP, et al. Dopamine D1 and D2 Receptor Co-activation Generates a Novel Phospholipase C-
mediated Calcium Signal. J. Biol. Chem. 2004; 279:35671–35678. [PubMed: 15159403]
12. Guo W, Shi L, Javitch JA. The Fourth Transmembrane Segment Forms the Interface of the
Dopamine D2 Receptor Homodimer. J. Biol. Chem. 2003; 278:4385–4388. [PubMed: 12496294]
13. Mesnier D, Baneres JL. Cooperative Conformational Changes in a G-protein-coupled Receptor
Dimer, the Leukotriene B4 Receptor BLT1. J. Biol. Chem. 2004; 279:49664–49670. [PubMed:
15358776]
14. Damian M, Mary S, Martin A, Pin JP, Baneres JL. G protein activation by the leukotriene B4
receptor dimer: Evidence for an absence of trans-activation. J. Biol. Chem. 2008; 283:21084–
21092. [PubMed: 18490452]
15. Carrillo JJ, Pediani J, Milligan G. Dimers of Class A G Protein-coupled Receptors Function via
Agonist-mediated Trans-activation of Associated G Proteins. J. Biol. Chem. 2003; 278:42578–
42587. [PubMed: 12920117]
16. Molinari P, et al. Promiscuous Coupling at Receptor-Galpha Fusion Proteins. THE RECEPTOR
OF ONE COVALENT COMPLEX INTERACTS WITH THE alpha - SUBUNIT OF ANOTHER.
J. Biol. Chem. 2003; 278:15778–15788. [PubMed: 12598520]
17. Pascal G, Milligan G. Functional Complementation and the Analysis of Opioid Receptor
Homodimerization. Mol Pharmacol. 2005; 68:905–915. [PubMed: 15967873]
18. Seifert R, Wenzel-Seifert K, Kobilka BK. GPCR-G[alpha] fusion proteins: molecular analysis of
receptor-G-protein coupling. Trends in Pharmacological Sciences. 1999; 20:383–389. [PubMed:
10462762]
19. Lee TW, Seifert R, Guan X, Kobilka BK. Restricting the Mobility of Gs alpha: Impact on Receptor
and Effector Coupling. Biochemistry. 1999; 38:13801–13809. [PubMed: 10529225]
20. Wenzel-Seifert K, Lee TW, Seifert R, Kobilka BK. Restricting mobility of Gsalpha relative to the
beta2-adrenoceptor enhances adenylate cyclase activity by reducing Gsalpha GTPase activity.
Biochem. J. 1998; 334:519–524. [PubMed: 9729456]
21. Rizzuto R, Simpson AWM, Brini M, Pozzan T. Rapid changes of mitochondrial Ca2+ revealed by
specifically targeted recombinant aequorin. Nature. 1992; 358:325–327. [PubMed: 1322496]
22. Blanpain C, et al. Extracellular Cysteines of CCR5 Are Required for Chemokine Binding, but
Dispensable for HIV-1 Coreceptor Activity. J. Biol. Chem. 1999; 274:18902–18908. [PubMed:
10383387]
23. Conklin BR, Farfel Z, Lustig KD, Julius D, Bourne HR. Substitution of three amino acids switches
receptor specificity of Gq[alpha] to that of Gi[alpha]. Nature. 1993; 363:274–276. [PubMed:
8387644]
24. Miller RT, Masters SB, Sullivan KA, Beiderman B, Bourne HR. A mutation that prevents GTP-
dependent activation of the alpha chain of Gs. Nature. 1988; 334:712–715. [PubMed: 3137475]
25. Xu W, et al. Functional role of the spatial proximity of Asp114(2.50) in TMH 2 and Asn332(7.49)
in TMH 7 of the mu opioid receptor. FEBS Lett. 1999; 447:318–324. [PubMed: 10214970]
26. Ballesteros JA, et al. Activation of the beta(2)-adrenergic receptor involves disruption of an ionic
lock between the cytoplasmic ends of transmembrane segments 3 and 6. J. Biol. Chem. 2001;
276:29171–29177. [PubMed: 11375997]
27. Moro O, Lameh J, Hogger P, Sadee W. Hydrophobic amino acid in the i2 loop plays a key role in
receptor-G protein coupling. J. Biol. Chem. 1993; 268:22273–22276. [PubMed: 8226735]
28. Strader CD, et al. Mutations that uncouple the beta-adrenergic receptor from Gs and increase
agonist affinity [published erratum appears in J Biol Chem 1988 Feb 25;263(6):3050]. J. Biol.
Chem. 1987; 262:16439–16443. [PubMed: 2890637]
29. Neve KA, et al. Modeling and mutational analysis of a putative sodium-binding pocket on the
dopamine D-2 receptor. Mol Pharmacol. 2001; 60:373–381. [PubMed: 11455025]
30. Urizar E, et al. An Activation Switch in the Rhodopsin Family of G Protein-coupled Receptors:
The thyrotropin receptor. J. Biol. Chem. 2005; 280:17135–17141. [PubMed: 15722344]
31. Simpson MM, et al. Dopamine D4/D2 Receptor Selectivity Is Determined by A Divergent
Aromatic Microdomain Contained within the Second, Third, and Seventh Membrane-Spanning
Segments. Mol Pharmacol. 1999; 56:1116–1126. [PubMed: 10570038]
Han et al. Page 11
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
32. Guo W, et al. Dopamine D2 receptors form higher order oligomers at physiological expression
levels. EMBO J. 2008
33. Wilson J, Lin H, Fu D, Javitch JA, Strange PG. Mechanisms of inverse agonism of antipsychotic
drugs at the D2 dopamine receptor: use of a mutant D2 dopamine receptor that adopts the activated
conformation. J Neurochem. 2001; 77:493–504. [PubMed: 11299312]
34. Guo W, Shi L, Filizola M, Weinstein H, Javitch JA. From The Cover: Crosstalk in G protein-
coupled receptors: Changes at the transmembrane homodimer interface determine activation.
PNAS. 2005; 102:17495–17500. [PubMed: 16301531]
35. Kota P, Reeves PJ, RajBhandary UL, Khorana HG. Opsin is present as dimers in COS1 cells:
Identification of amino acids at the dimeric interface. PNAS. 2006; 103:3054–3059. [PubMed:
16492774]
36. Goudet C, et al. Asymmetric Functioning of Dimeric Metabotropic Glutamate Receptors Disclosed
by Positive Allosteric Modulators. J. Biol. Chem. 2005; 280:24380–24385. [PubMed: 15863499]
37. Hlavackova V, et al. Evidence for a single heptahelical domain being turned on upon activation of
a dimeric GPCR. Embo Journal. 2005; 24:499–509. [PubMed: 15660124]
38. Kniazeff J, et al. Closed state of both binding domains of homodimeric mGlu receptors is required
for full activity. Nature Structural & Molecular Biology. 2004; 11:706–713.
39. Kaupmann K, et al. GABAB-receptor subtypes assemble into functional heteromeric complexes.
Nature. 1998; 396:683–687. [PubMed: 9872317]
40. Damian M, Martin A, Mesnier D, Pin JP, Baneres JL. Asymmetric conformational changes in a
GPCR dimer controlled by G-proteins. EMBO J. 2006; 25:5693–5702. [PubMed: 17139258]
41. Springael JY, Urizar E, Costagliola S, Vassart G, Parmentier M. Allosteric properties of G protein-
coupled receptor oligomers. Pharmacol Ther. 2007; 115:410–418. [PubMed: 17655934]
42. Springael JY, et al. Allosteric modulation of binding properties between units of chemokine
receptor homo-and hetero-oligomers. Mol Pharmacol. 2006; 69:1652–1661. [PubMed: 16467191]
43. Urban JD, et al. Functional selectivity and classical concepts of quantitative pharmacology. J
Pharmacol Exp Ther. 2007; 320:1–13. [PubMed: 16803859]
44. Javitch JA, et al. The fourth transmembrane segment of the dopamine D2 receptor: Accessibility in
the binding-site crevice and position in the transmembrane bundle. Biochemistry. 2000; 39:12190–
12199. [PubMed: 11015197]
45. Costagliola S, et al. Structure-function relationships of two loss-of-function mutations of the
thyrotropin receptor gene. Thyroid. 1999; 9:995–1000. [PubMed: 10560953]
46. Brini M, et al. Transfected Aequorin in the Measurement of Cytosolic Ca2+ Concentration
([Ca2+]c). J. Biol. Chem. 1995; 270:9896–9903. [PubMed: 7730373]
47. Niv MY, Skrabanek L, Filizola M, Weinstein H. Modeling activated states of GPCRs: the
rhodopsin template. J. Comput. Aided Mol. Des. 2006; 20:437–448. [PubMed: 17103019]
48. van Dijk AD, et al. Data-driven : HADDOCK's adventures in CAPRI. Proteins. 2005; 60:232–238.
[PubMed: 15981252]
49. van Dijk M, van Dijk AD, Hsu V, Boelens R, Bonvin AM. Information-driven protein-DNA
docking using HADDOCK: it is a matter of flexibility. Nucleic Acids Res. 2006; 34:3317–3325.
[PubMed: 16820531]
50. Scheerer P, et al. Crystal structure of opsin in its G-protein-interacting conformation. Nature. 2008;
455:497–502. [PubMed: 18818650]
Han et al. Page 12
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 1. Functional complementation of two “non-functional receptors”
We used an aequorin assay that couples Gq (or Gqi5) activation to a luminescence readout.
(a) The agonist quinpirole did not lead to D2R-induced Gq activation. (b) D2R when
coexpressed with free Gqi5 or (c) D2R fused with Gqi5 via a linker (D2-linker-Gqi5) led to
quinpirole-induced luminescence. (c) A nonfunctional Gα deficient fusion construct, D2-
linker-Gqi5G208A failed to produce luminescence. (d) Free Gqi5 rescued the function of D2-
linker-Gqi5G208A. (e) Free Gqi5 failed to rescue the function of non-linker D2R-Gqi5,
which unlike D2R-linker-Gqi5, did not signal when expressed alone. (f) Coexpressing D2R
with D2R-Gqi5 (12 hour tetracycline induction) restored signaling, despite the inability of
either construct to signal in this assay when expressed alone. Activation data represent
Han et al. Page 13
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
luminescence relative to that seen with 0.1% triton treatment. The mean±SEM of at least 3
experiments, each conducted in triplicate, are shown. The symbols used in (Fig. 1–Fig. 4 and
Fig. 6) are explained in detail in (Supplementary Fig. 2 online).
Han et al. Page 14
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 2. Characterization of D2R mutants
(a) Schematic representation showing the positions of the mutations in the D2 receptor, with
coloring corresponding to the symbols and lines in (b) and (c). (b) D2/D4, a D2 mutant with
4 amino acids substituted from the D4 receptor (V912.61F/F1103.29L/V1113.28M/Y4087.35V)
making it 1000-times more sensitive to a D4-selective inhibitor (Supplementary Fig. 4
online), is activated by quinpirole, albeit with a lower potency and efficacy when compared
with WT D2R. (c) All the other mutants, which are described in the text, were non-
functional. Activation data were normalized as in Fig. 1. The mean±SEM of at least 3
experiments, each conducted in triplicate, are shown.
Han et al. Page 15
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 3. Asymmetric contributions of the protomers to signaling
(a,b) When all mutants (as protomer A) were coexpressed with WT D2R-Gqi5 (as protomer
B), only (a) WT and D2/D4 were able to signal. (b) None of the other mutants were able to
restore signaling when coexpressed with WT D2R-Gqi5. (c,d,e) The results differed when
WT D2R (as protomer A) was coexpressed with the various mutant-Gqi5 constructs (as
protomer B). (c) D2/D4-Gqi5 ( ), (d) D1143.32A-Gqi5 ( ), deletion 213–219-Gqi5 ( ),
and D802.50A-Gqi5 ( ) restored the ability of unfused WT D2R to signal. (e) Coexpressing
R1323.50A -Gqi5 ( ), V1363.54D/M1403.58E-Gqi5 ( ), or N3937.49A-Gqi5 ( ) with WT
D2R failed to rescue signaling (d). Note that D114A-Gqi5 ( ) and D2/D4-Gqi5 ( )
Han et al. Page 16
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
showed a higher maximal activation than WT. Activation data represent relative
luminescence when compared to WT D2R coexpressed with WT D2R-Gqi5 after
normalizing for surface expression of the Gqi5 fusion construct (see Methods). The mean
±SEM of at least 3 experiments, each conducted in triplicate, are shown.
Han et al. Page 17
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 4. The second protomer allosterically modulates signaling
Shown are effects on signaling with the D2/D4 construct expressed either as protomer A (a),
or as protomer B (D2/D4-Gqi5) (b). (a) The D4-selective antagonist L745,870 (1 µM) totally
blocked signaling of the D2/D4 construct expressed as protomer A with WT-Gqi5. (b) In
contrast, L745,870 increased maximal activation for WT D2R coexpressed with D2/D4-
Gqi5 to156.7±7.3% (n=9) (p<0.01*** by Student’s t-test) of that observed for D2R
coexpressed with WT D2R-Gqi5 (Fig. 3a). (c) Coexpression of a constitutively active
mutant (Supplementary Fig. 8 online) that was unable to bind ligand (D1143.32A/CAM-
Gqi5), to enhance the fraction of protomer B in an active conformation, led to 49.6±8.4%
(n=9) (p<0.01*** by Student’s t-test) of maximal activity () when compared to WT D2R
coexpressed with D114A-Gqi5 (◊). Activation data were normalized to surface expression as
described in Fig. 3. The mean±SEM of at least 3 experiments, each conducted in triplicate,
are shown.
Han et al. Page 18
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 5. Computational model of the complex between the rhodopsin dimer and heterotrimeric
Gt
(a) Structural representation of the nonameric oligomer array; the dashed box identifies the
TM4 dimer contained in Model 2. (b) Structural representation of the complex formed
between transducin and the nonameric oligomer array. The optimal representative structure
(defined in Methods) is shown for Model 2. (c) Closeup view of the interaction between
specific residues of Gα (rendered in red, CPK representation) and the IL3 (cyan) and IL2
loops of protomer A and B (magenta and blue, respectively). (d) Side view of the complex
showing Gtα (red), Gtβ (wheat), Gtγ (orange), protomer A (green), protomer B (light blue),
Han et al. Page 19
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
IL2 of protomer A (magenta), IL2 of protomer B (blue), and IL3 of protomer B (cyan).
Other views of the model complex are shown in (Supplementary Fig. 10 online).
Han et al. Page 20
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 6. Cartoon of different D2R dimer activation states, with activation data for these states,
from the perspective of agonist-mediated activation of protomer A
Bound agonist is represented by a black square. Activation is represented by a trapezoid
with a bold base. Extent of activation is indicated by increasingly bold trapezoid boundaries.
The inverse agonist bound state is represented by an inverted trapezoid. (1) Neither
protomer is activated. (2) Protomer A binds agonist and protomer B is constitutively active
(or in the case of a heterodimer, is occupied by protomer B’s agonist). (3) Protomer A binds
agonist, whereas protomer B cannot bind (or in a heterodimer, is not agonist-bound). Note
that although protomer B is not activated by ligand, it can isomerize to the active state,
which would result in configuration (2). (4) Protomer A binds agonist, whereas protomer B
is stabilized in the inactive state by inverse agonist. Experimentally determined maximal
activation representing these idealized conformations: (1) no ligand, (2) WT D2R
coexpressed with D114A/CAM-Gqi5, (3) WT D2R coexpressed with D114A-Gqi5, (5) WT
D2R coexpressed with D2/D4-Gqi5 in the presence of the selective D4 antagonist,
L745,870. Activation data are normalized to that of WT D2R coexpressed with WT D2R-
Gqi5, which is indicated by a dotted line to indicate the potential range of enhanced and
reduced signaling achievable by modulation of the “heterodimer” partner.
Han et al. Page 21
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 7.
Han et al. Page 22
Nat Chem Biol
. Author manuscript; available in PMC 2010 March 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
... These receptor complexes possess two or more orthosteric binding sites from which endogenous agonists initiate a functional response. These sites mutually affect each other via allosteric interactions [17][18][19]. However, apparent allosteric cooperativity does not always imply the dimerization of GPCRs [20]. ...
... One of the reasons why bell-shaped curves are rare in the literature is the way results are reported. For example, asymmetric activation of the D 2 -dopamine receptor was described [18]. The maximal activation was observed when the agonist quinpirole was bound to a single protomer. ...
... The introduced slope factor n affects both the observed maximal response E' MAX , Equation (18), and the ratio of EC 50 to K A , Equation (19). ...
Article
Introduction Accurate ranking of efficacies and potencies of agonists is essential in the discovery of new selective agonists. For the purpose of system-independent ranking of agonists, the operational model of agonism (OMA) has become a standard. Many receptors function as oligomers which makes functional responses more complex, requiring an extension of the original OMA. Areas covered Explicit equations of the operational model of agonism of receptor dimers (OMARD) were derived. The OMARD can be applied to any receptor possessing two orthosteric sites. The behaviour of OMARD was analysed to demonstrate its complexity and relation to experimental data. Properties of OMARD and OMA equations were compared to demonstrate their pros and cons. Expert opinion Extension of OMA by slope factors gives simple equations of functional response that are easy to fit experimental data but results may be inaccurate because of exponentiation of operational efficacy. Also, such equations cannot accommodate bell-shaped curves. Explicit equations of OMARD give accurate results but are complex and tedious to fit experimental data. All operational models use inter-dependent parameters that are a hurdle in the fitting. A good understanding of OMARD behaviour helps to overcome such obstacles.
... Previous studies of Han and coworkers on the D2 receptor homodimer demonstrated asymmetric allosteric communication between the protomers. The authors engineered a functional complementation assay within a minimal signaling unit, which consists of two GPCRs and a single heterotrimeric G protein 41 (Figure 1). A cell line was established stably expressing aequorin, a protein that produces luminescence in a calcium-dependent manner and can therefore be used to generate a luminescence readout for GPCR-induced phospholipase C (PLC) activation. ...
... It was hypothesized that the short-tethered Gαqi5 is lacking the ability to be adequately arranged for an efficient interaction with the protomer to which it was fused or an adjacent protomer and blocks the appropriate coupling of free Gαqi5 to the receptor. Indeed, coexpression of wild-type D2R (protomer A) and D2R-Gαqi5 (protomer B) restored signaling, providing a minimal signaling unit for dimerspecific signaling 41 . This reconstituted signaling entity offered the opportunity to determine the role of each receptor in the signaling process by independently manipulating the protomers. ...
... However, activation of the second protomer blunts signaling while inverse agonist binding to the second protomer even enhances activation, indicating an allosteric modulation within the dimer. Importantly, this allosteric modulation was shown to be the consequence from direct interaction of the dimer with the G protein and not a result from downstream effects 41 . This novel methodology gave crucial insight into the complex mechanisms within the dopamine D2R homodimer during activation and offers the possibility to further identify the signaling properties of all kinds of homo-and heterodimers. ...
Thesis
Class A GPCRs have traditionally been considered to exist and signal as monomeric entities but growing evidence supports the hypothesis that GPCRs can assemble as transient homo- or heterodimers. These protein-protein interactions can dramatically modify the pharmacological properties of the individual protomers and targeting GPCR dimers and investigating their signaling behavior may provide useful information on their physiological relevance. A prominent example of GPCRs that were shown to exist as transient homomeric and heteromeric complexes are dopamine D2 receptors, which are critically involved in the pathogenesis of multiple neurological and psychotic disorders including schizophrenia, Parkinson’s disease, depression and substance abuse. While the detection of those receptor complexes has been facilitated by advanced biochemical and biophysical methods, information about the functional consequences of dimerization is often lacking. The aim of this thesis was to specifically address D2R/NTS1R, D2R/D2R and D2R/D4R dimers and to investigate their functional characteristics. Previously developed bivalent ligands served as tools for a detailed analysis. Starting with a BRET based evaluation whether these bivalent ligands can influence dimerization, the functional properties of these different dimeric complexes regarding G protein activation were elucidated using different signaling assays. The dopaminergic system is highly interconnected with the neurotensinergic system91 and the existence of D2R-NTS1R heterodimers that may be implicated in the pathophysiology of neurological diseases is well established. The development of various heterobivalent ligands comprising a dopamine pharmacophore linked to a neurotensin pharmacophore by PEG-based spacers of variable length (ranging from 44 to 88 atoms) in our group represented the starting point for the investigation of the functional properties of the D2R-NTS1R heterodimer regarding G protein activation. These bivalent ligands showed extremely high affinity for the heterodimer compared to their monovalent analogs and are therefore promising tools for selectively targeting the D2R-NTS1R dimer. In this thesis, I established a cAMP BRET assay based on the biosensor CAMYEL for cells expressing either D2SR, NTS1R or both receptors. I characterized bivalent ligands regarding their ability to modulate intracellular cAMP levels in these cells. In D2SR-expressing cells, all bivalent ligands bearing an aminoindane-type agonist as dopamine pharmacophore were able to inhibit forskolin-induced cAMP accumulation as effective as the reference quinpirole, with CS142 (44-atom linker) being the most potent. In contrast, ligands with a phenylpiperazine-based scaffold or the antagonistic eticlopride moiety showed very low or no intrinsic activity. When the NTS1R was expressed alone, all bivalent ligands bearing the active neurotensin fragment NT(8-13) increased cAMP levels to the same extent as NT(8-13) alone , although with reduced potency. As expected from their behavior in monoexpressing cells, by addressing the heterodimer in D2SR-NTS1R coexpressing cells, bivalent ligands with a phenylpiperazine moiety or an eticlopride structure increased cAMP levels in the same manner as seen in NTS1R expressing cells. Interestingly, bivalent ligands comprising the agonistic aminoindane pharmacophore showed a similar activation behavior in coexpressing cells as bivalent ligands with a D2SR antagonist structure. Although acting as potent full D2SR agonists, they increased cAMP levels to the same extent as the reference NT(8-13) (24% above basal cAMP level), while the reference quinpirole reduced cAMP levels 57% below basal level. This indicates that these bivalent ligands were not able to induce an inhibitory effect on the NTS1R-mediated cAMP accumulation. No dependence of the linker length between the pharmacophores was determined and control experiments in the absence of forskolin or under conditions leading to a higher propensity of heterodimerization excluded any correlations between these conditions and the activation properties of the bivalent ligands. Interestingly, the lack of D2R activation is not a consequence of bivalent binding but simultaneous activation of both receptors, since co-stimulation with quinpirole and 5 nM NT(8-13) also resulted in a complete loss of D2R activation. A reversed inhibition of NTS1R signaling by quinpirole was not obtained, further indicating a NTS1R-dominated signaling within the heterodimer. According to recent evidence, the D2 receptor homodimer is most likely involved in the pathophysiology of schizophrenia37. Dimerization of class A GPCRs is a transient process and the participating receptors are in an equilibrium between monomeric and dimeric state. Since it was already shown that dopaminergic agonists can influence the dynamic equilibrium between D2R monomers and dimers, the investigation of homobivalent ligands comprising two dopamine pharmacophores linked by PEG spacers of variable length (48- and 92-atom linker) regarding their ability to influence receptor interactions should provide useful information to further understand the pharmacological role of D2R homodimers. To distinguish between unspecific and specific receptor-receptor interactions, BRET saturation experiments are a highly sensitive and validated method. By coexpression of D2R tagged with Renilla luciferase and mVenus, respectively, it was possible to detect specific receptor-receptor interactions for both the D2SR and the D2LR homodimer, as reflected in hyperbolic saturation curves that are indicative of these specific receptor interactions. This experimental setup was further used to determine whether the bivalent ligands are able to affect the propensity of the D2R to form homodimers and stabilize or destabilize D2R homodimers. Independent of the pharmacophore, incubation with short-linker bivalent ligands (48-atoms) and monovalent control compounds had no effect on BRETmax and BRET50 values, suggesting that these ligands had no particular influence on the homodimerization properties of D2SR or D2LR. In contrast, incubation with the long-linker bivalent ligands (92-atoms) with an aminoindane (MG245) and a phenylpiperazine (MG244) head group significantly reduced BRET50 for both D2SR and D2LR homodimers, while BRETmax values remained unchanged. This is indicative for an affinity change between the protomers withing the dimer, suggesting that MG245 and MG244 are able to engage both orthosteric binding sites simultaneously and foster homodimerization (Figure 42). However, it is not possible to quantify the increase in dimerization provoked by the bivalent ligands, since changes in the relative orientation of both protomers might account for the overall BRET changes. Although the linker length of MG247 comprising an eticlopride headgroup is equivalent to MG244 and MG245, no influence on BRETmax and BRET50 was observed. This may either be explained by a lack of bivalent binding or a concentration dependent phenomenon, since previous studies suggested that high concentrations of bivalent ligands prefer a monovalent binding mode over bivalent binding and the applied concentration of MG247 might be too high to enhance dimerization. Experiments with a ligand-binding deficient D2R mutant (D2-D114A) as BRET donor further confirmed the dimerization enhancing effect of MG244 and MG245, as the enhancing effect was absent in this set-up in which bivalent binding was not possible. Moreover, BRET competition experiments with increasing concentrations of D2wt validated the specificity of the ligand effects on homodimerization. ΔBRET after stimulation with MG244 or MG245 was gradually diminished, due to competition of D2wt with the BRET acceptor for interaction with the BRET donor. A novel IP-One assay approach was established to study dimerspecific signaling at the D2R homodimer. A chimeric Gαqi5 was C-terminally fused to the D2R and coexpressed with wild-type D2R, both unable to signal in this assay when expressed alone. However, in this assay set-up, these two ‘non-functioning’ receptors complement to a functional signaling unit after forming a dimeric receptor complex. Bivalent ligands with an aminoindane pharmacophore showed almost full D2S receptor activation, while only a very weak signal was detected for the phenylpiperazine-derived bivalent ligands. Interestingly, MG245 was found to exhibit an enhanced potency in this dimerspecific signaling assay compared to its analog MG242, even though similar potencies were observed in IP-One assays for unfused D2SR coexpressed with free Gαqi5, representing typical monomer receptor activation. The increased potency might be a consequence of bivalent binding and is in accordance with the results from the BRET saturation experiments. In addition, it was evaluated if these homobivalent ligands were also able to influence D2R/NTS1R heterodimerization. BRET saturation experiments with NTS1-Rluc and D2S-mVenus revealed that the heterobivalent D2R-NTS1R ligand CS411 was able to foster dimerization between D2SR and NTS1R, as reflected by a decreased BRET50 value. In contrast, the D2R-homobivalent ligands MG244 and MG245 with a 92-atom spacer, but not with a 48-atom spacer, markedly decreased interactions between both receptors. Those findings were also confirmed with BRET competition experiments, suggesting that there is a dynamic equilibrium between D2R homodimerization and D2SR/NTS1R heterodimerization that could be modulated with bivalent ligands. Thus, the possibility of modifying the dynamic equilibrium between monomers, homodimers and heterodimers with these bivalent ligands may pave the way toward novel pharmacological concepts and innovative drugs. To elucidate ligand influences on D2R-D4R dimer formation, BRET saturation experiments were performed in analogy to D2R homodimerization and specific receptor-receptor interactions between D2R and D4R were confirmed by hyperbolic saturation curves. Incubation with the previously described homobivalent ligands resulted in an enhanced dimerization between the receptors, as reflected by decreased BRET50 and increased BRETmax values. Unlike the D2R homodimer, this effect was also seen for the short-linker bivalent ligands (MG241 and MG242), although not as pronounced as for MG244 and MG245 with a long PEG-spacer. G protein activation of the heterodimer was investigated with cAMP BRET assays. Both receptors couple to Gi proteins, making it difficult to distinguish between the activation properties of the individual receptors within the dimer. Therefore, subtype-selective agonists and antagonists from commercial sources and the in-house library were identified as pharmacological instruments for functional experiments at the heterodimer. Thereby, the in-house compound ST120 was found to be a D4R selective agonist, while antagonizing the D2R. Yet, due to natural limitations of the cAMP BRET assay, coexpression experiments with D2S and D4 receptors were difficult to interpret and did not provide any new information about functional interactions between the protomers. Consequently, the dimerspecific assay approach described for the D2R homodimer was adapted to the D2R/D4R heterodimer. Coexpression of D2S-Gαqi5 and D4wt indeed led to the formation of a functional signaling unit, as reflected by increased IP1 accumulation after quinpirole stimulation. The unfused protomer was identified as the dominant part of the signaling unit, since coexpression of the signaling incompetent mutant D2SD80A-Gαqi5 and D4wt resulted in a similar activation signal. Importantly, the selective D4 receptor agonist ST120 exhibited increased efficacy by activating the dimer. Co-stimulation with quinpirole and 25 nM eticlopride, which selectively blocks the D2R, indicated a correlation between the inactive or antagonist-bound state of the D2R and the enhanced efficacy of the D4R. Coexpression of a binding-deficient D2S receptor mutant D2S-D114A-Gαqi5 with D4wt did not result in an increased maximal response for ST120. This further indicates that the activation of the D4R within the heterodimer may be positively modulated by the inactive state of the D2S receptor. Using BRET-based dimerization and ligand activation assays combined with the adaption of the IP-One assay to a dimerspecific signaling readout and diagnostic receptor mutants, I was able to further elucidate the dimerspecific signaling of D2R upon interaction with NTS1R, D4R or D2R itself. The results highlight the potential of tailor-made bivalent ligands for future drug development.
... Other possible interfaces were determined for the ac-ac-B, ac-in, and in-ar dimers. For D 2 R, only in-in, ac-ac, and ac-in D 2 R homodimers have been experimentally detected; however, other combinations are also possible [17,94,95]. ...
Article
Full-text available
G protein-coupled receptors (GPCRs) are known to dimerize, but the molecular and structural basis of GPCR dimers is not well understood. In this study, we developed a computational framework to generate models of symmetric and asymmetric GPCR dimers using different monomer activation states and identified their most likely interfaces with molecular details. We chose the dopamine receptor D2 (D2R) homodimer as a case study because of its biological relevance and the availability of structural information. Our results showed that transmembrane domains 4 and 5 (TM4 and TM5) are mostly found at the dimer interface of the D2R dimer and that these interfaces have a subset of key residues that are mostly nonpolar from TM4 and TM5, which was in line with experimental studies. In addition, TM2 and TM3 appear to be relevant for D2R dimers. In some cases, the inactive configuration is unaffected by the partnered protomer, whereas in others, the active protomer adopts the properties of an inactive receptor. Additionally, the β-arrestin configuration displayed the properties of an active receptor in the absence of an agonist, suggesting that a switch to another meta-state during dimerization occurred. Our findings are consistent with the experimental data, and this method can be adapted to study heterodimers and potentially extended to include additional proteins such as G proteins or β-arrestins. In summary, this approach provides insight into the impact of the conformational status of partnered protomers on the overall quaternary GPCR macromolecular structure and dynamics.
... Our quan?ta?ve understanding of the func?on of many membrane proteins would greatly improve if we were able to accurately determine the molecular density in the plasma membrane of living cells. For example, this would enable measurements of binding interac?ons between membrane proteins and intracellular proteins [8,9] directly in the living cell. Since such processes depend on the dis?nct density of both interac?ng partners it is essen?al that we can accurately determine the (which was not certified by peer review) is the author/funder. ...
Preprint
Biological systems are regulated by molecular interactions which are tuned by the concentrations of each of the molecules involved. Cells exploit this feature by regulating protein expression, to adapt their responses to overstimulation. Correlating events in single cells to the concentrations of proteins involved can therefore provide important mechanistic insight into cell behavior. Unfortunately, quantification of molecular densities by fluorescence imaging becomes non-trivial due to the diffraction limited resolution of the imaged volume. We show here an alternative approach to overcome this limitation in optical quantification of protein concentrations which is based on calibrating protein volume and surface densities in a model membrane system. We exploit the ability of fluorescently labeled annexin V to bind membranes in presence of calcium. By encapsulating known concentrations of annexin V, we can directly infer the membrane density of annexin V after addition of Ca2+ and correlate the density with the measured fluorescence signal. Our method, named Calmet, enables quantitative determination of the concentration of cytosolic and membrane associated proteins. The applicability of Calmet is demonstrated by quantification of a transmembrane protein receptor (beta 1 adrenergic receptor) labeled by SNAP tagged fluorophores and expressed in HEK293 cells. Calmet is a generic method suitable for the determination of a broad range of concentrations and densities and can be used on regular fluorescence images captured by confocal laser scanning microscopy.
... The allosteric binding site is an alternative binding site distinct from the orthosteric site, and the binding of allosteric modulators potentiates or inhibits activation of the receptor by its natural ligand [107]. Allosteric ligands and their binding to allosteric binding sites influence the ability to fine-tune the response to an orthosteric ligand in a time-and spatially dependent manner and may confer signaling bias and probe dependence, further contributing to the possibility for remarkably precise pharmacological modulation [108][109][110][111]. Our analysis showed only a few mutations in allosteric binding sites. ...
Article
Full-text available
G protein-coupled receptors (GPCRs) mediate several signaling pathways through a general mechanism that involves their activation, upholding a chain of events that lead to the release of molecules responsible for cytoplasmic action and further regulation. These physiological functions can be severely altered by mutations in GPCR genes. GPCRs subfamily A17 (dopamine, serotonin, adrenergic and trace amine receptors) are directly related with neurodegenerative diseases, and as such it is crucial to explore known mutations on these systems and their impact in structure and function. A comprehensive and detailed computational framework - MUG (Mutations Understanding GPCRs) - was constructed, illustrating key reported mutations and their effect on receptors of the subfamily A17 of GPCRs. We explored the type of mutations occurring overall and in the different families of subfamily A17, as well their localization within the receptor and potential effects on receptor functionality. The mutated residues were further analyzed considering their pathogenicity. The results reveal a high diversity of mutations in the GPCR subfamily A17 structures, drawing attention to the considerable number of mutations in conserved residues and domains. Mutated residues were typically hydrophobic residues enriched at the ligand binding pocket and known activating microdomains, which may lead to disruption of receptor function. MUG as an interactive web application is available for the management and visualization of this dataset. We expect that this interactive database helps the exploration of GPCR mutations, their influence, and their familywise and receptor-specific effects, constituting the first step in elucidating their structures and molecules at the atomic level.
... Structural and biochemical studies suggested that different GPCRs can self-associate through distinct transmembrane helical (TMH) interfaces. Computational modeling approaches based on molecular dynamics simulations have also identified different possible modes and lifetimes of GPCR associations 14,15 but the functional relevance of these oligomeric forms remains poorly understood [5][6][7][16][17][18][19][20][21][22] . For example, chemokine receptor CXCR4 signaling is linked to the formation of nanoclusters at the cell membrane 23 . ...
Article
Full-text available
Communication across membranes controls critical cellular processes and is achieved by receptors translating extracellular signals into selective cytoplasmic responses. While receptor tertiary structures can be readily characterized, receptor associations into quaternary structures are challenging to study and their implications in signal transduction remain poorly understood. Here, we report a computational approach for predicting receptor self-associations, and designing receptor oligomers with various quaternary structures and signaling properties. Using this approach, we designed chemokine receptor CXCR4 dimers with reprogrammed binding interactions, conformations, and abilities to activate distinct intracellular signaling proteins. In agreement with our predictions, the designed CXCR4s dimerized through distinct conformations and displayed different quaternary structural changes upon activation. Consistent with the active state models, all engineered CXCR4 oligomers activated the G protein Gi, but only specific dimer structures also recruited β-arrestins. Overall, we demonstrate that quaternary structures represent an important unforeseen mechanism of receptor biased signaling and reveal the existence of a bias switch at the dimer interface of several G protein-coupled receptors including CXCR4, mu-Opioid and type-2 Vasopressin receptors that selectively control the activation of G proteins vs β-arrestin-mediated pathways. The approach should prove useful for predicting and designing receptor associations to uncover and reprogram selective cellular signaling functions.
... The receptors for which the effect of homo-dimerization or homo-oligomerization was examined on G protein coupling do not provide a clear view of what could happen in most GPCRs. Indeed, when using purified receptors, GPCR dimerization was previously reported to decrease G protein coupling efficacy 37,38 , supporting the view that in a GPCR dimer, only one protomer is active [86][87][88] . In class C dimeric GPCRs, only one protomer at a time is considered to activate G proteins 87,89-92 and oligomerization further decreases G protein coupling efficacy 93,94 . ...
Article
Full-text available
G protein-coupled receptors (GPCRs) are important drug targets thatmediate various signaling pathways by activating G proteins and engaging β-arrestin proteins. Despite its importance for the development of therapeutics with fewer side effects, the underlying mechanism that controls the balance between these signaling modes of GPCRs remains largely unclear. Here, we show that assembly into dimers and oligomers can largely influence the signaling mode of the platelet-activating factor receptor (PAFR). Single-particle analysis results show that PAFR can form oligomers at low densities through two possible dimer interfaces. Stabilization of PAFR oligomers through crosslinking increases G protein activity, and decreases β-arrestin recruitment and agonist-induced internalization significantly. Reciprocally, β-arrestin prevents PAFR oligomerization. Our results highlight a mechanism involved in the control of receptor signaling, and thereby provide important insights into the relationship between GPCR oligomerization and downstream signaling.
... Yet, a large number of studies have explored the relevance of GPCR dimers at functional level, especially for heterodimers. For example, in case of the dopamine D 2 receptor (D 2 R), activation of both protomers within a D 2 R dimer shows less efficacy in signaling than that of the situation where only one receptor is active (Han et al., 2009). This addresses a negative cooperativity between the dimeric protomers. ...
Thesis
Full-text available
G protein-coupled receptors (GPCRs) constitute the largest class of membrane proteins, and are the master components that translate extracellular stimulus into intracellular signaling, which in turn modulates key physiological and pathophysiological processes. Research within the last three decades suggests that many GPCRs can form complexes with each other via mechanisms that are yet unexplored. Despite a number of functional evidence in favor of GPCR dimers and oligomers, the existence of such complexes remains controversial, as different methods suggest diverse quaternary organizations for individual receptors. Among various methods, high resolution fluorescence microscopy and imagebased fluorescence spectroscopy are state-of-the-art tools to quantify membrane protein oligomerization with high precision. This thesis work describes the use of single molecule fluorescence microscopy and implementation of two confocal microscopy based fluorescence fluctuation spectroscopy based methods for characterizing the quaternary organization of two class A GPCRs that are important clinical targets: the C-X-C type chemokine receptor 4 (CXCR4) and 7 (CXCR7), or recently named as the atypical chemokine receptor 3 (ACKR3). The first part of the results describe that CXCR4 protomers are mainly organized as monomeric entities that can form transient dimers at very low expression levels allowing single molecule resolution. The second part describes the establishment and use of spatial and temporal brightness methods that are based on fluorescence fluctuation spectroscopy. Results from this part suggests that ACKR3 forms clusters and surface localized monomers, while CXCR4 forms increasing amount of dimers as a function of receptor density in cells. Moreover, CXCR4 dimerization can be modulated by its ligands as well as receptor conformations in distinct manners. Further results suggest that antagonists of CXCR4 display distinct binding modes, and the binding mode influences the oligomerization and the basal activity of the receptor: While the ligands that bind to a “minor” subpocket suppress both dimerization and constitutive activity, ligands that bind to a distinct, “major” subpocket only act as neutral antagonists on the receptor, and do not modulate neither the quaternary organization nor the basal signaling of CXCR4. Together, these results link CXCR4 dimerization to its density and to its activity, which may represent a new strategy to target CXCR4.
Article
Full-text available
Bioluminescence and fluorescence resonance energy transfer (BRET and FRET) together with the proximity ligation method revealed the existence of G-protein-coupled receptors, Ionotropic and Receptor tyrosine kinase heterocomplexes, e.g., A2AR–D2R, GABAA–D5R, and FGFR1–5-HT1AR heterocomplexes. Molecular integration takes place through allosteric receptor–receptor interactions in heteroreceptor complexes of synaptic and extra-synaptic regions. It involves the modulation of receptor protomer recognition, signaling and trafficking, as well as the modulation of behavioral responses. Allosteric receptor–receptor interactions in hetero-complexes give rise to concepts like meta-modulation and protein modulation. The introduction of receptor–receptor interactions was the origin of the concept of meta-modulation provided by Katz and Edwards in 1999, which stood for the fine-tuning or modulation of nerve cell transmission. In 2000–2010, Ribeiro and Sebastiao, based on a series of papers, provided strong support for their view that adenosine can meta-modulate (fine-tune) synaptic transmission through adenosine receptors. However, another term should also be considered: protein modulation, which is the key feature of allosteric receptor–receptor interactions leading to learning and consolidation by novel adapter proteins to memory. Finally, it must be underlined that allosteric receptor–receptor interactions and their involvement both in brain disease and its treatment are of high interest. Their pathophysiological relevance has been obtained, especially for major depressive disorder, cocaine use disorder, and Parkinson’s disease.
Chapter
Intracellular cell signaling is a well understood process. However, extracellular signals such as hormones, adipokines, cytokines and neurotransmitters are just as important but have been largely ignored in other works. They are causative agents for diseases including hypertension, diabetes, heart disease, and arthritis so offer new, and often more approachable, targets for drug design. Aimed at medical professionals and pharmaceutical specialists, this book integrates extracellular and intracellular signalling processes and offers a fresh perspective on new drug targets. Written by colleagues at the same institution, but with contributions from leading international authorities, it is the result of close cooperation between the authors of different chapters. Readers are introduced to a new approach to disease causation by adipokines and toxic lipids. Heart disease, migraines, stroke, Alzheimer's disease, diabetes, cancer, and arthritis are approached from the perspective of prevention and treatment by alteration of extracellular signalling. Evidence is presented that the avoidance of toxic lifestyles can reduce the incidence of such illnesses and new therapeutic targets involving adipokines, ceramide and endocannabinoids are discussed.
Article
Full-text available
Signal transduction of the heptahelical G protein-coupled receptors (GPCRs) involves multiple receptor domains, but a universal consensus domain for coupling has not yet been defined. Alanine mutagenesis scanning was performed on the intracellular loops and the COOH tail of the human muscarinic cholinergic receptor (Hm1) to identify coupling domains. Stimulation of phosphatidylinositol (PI) turnover was determined after transfection of the alanine mutants into U293 human embryonic kidney cells. Alanine substitutions in four regions (loops i1, i2, and NH2 and COOH junctions of i3) impaired coupling efficiency by approximately 50% or more, but the strongest reduction (> 80%) resulted from alanine replacement of a single amino acid, leucine 131. This residue is located in the middle of the second intracellular loop (i2), within the highly conserved GPCR motif (DRYXXV(I)XXPL). The position equivalent to Leu-131 in Hm1 contains a bulky hydrophobic amino acid (L, I, V, M, or F) in nearly all cloned GPCRs. Substitution of Leu-131 with polar amino acids (aspartate and asparagine) also resulted in strongly defective coupling, whereas phenylalanine (found in the equivalent position in the beta 2 adrenoceptor) can replace leucine without losing PI coupling ability of Hm1. Alanine substitution of the corresponding amino acid in the Hm3 receptor (L174A) also inhibited agonist-stimulated PI turnover, while replacing Phe-139 with alanine in the beta 2 adrenoceptor suppressed stimulation of adenylyl cyclase. We propose that a bulky hydrophobic amino acid in the middle of the i2 loop serves as a general site relevant to G protein coupling, whereas coupling selectivity is governed by other receptor domains.
Article
Full-text available
The deletion of residues 239-272 from the hamster beta-adrenergic receptor resulted in a loss of the ability of the receptor, expressed in mouse L cells, to stimulate adenylate cyclase (Dixon, R. A. F., Sigal, I. S., Rands, E., Register, R. B., Candelore, M. R., Blake, A. D., and Strader, C. D. (1987) Nature 326, 73-77). This mutant receptor (D(239-272)beta AR) bound the agonist isoproterenol with a single class of binding sites, in contrast to the wild-type beta-adrenergic receptor, which exhibited two classes of agonist affinity sites. We now report that the affinity of D(239-272)beta AR for isoproterenol is relatively insensitive to detergent solubilization or to treatment with either GTP or NaF, indicating the absence of a receptor-Gs interaction. Whereas deletions within the region of amino acids 229-258 did not reduce the ability of the receptor to couple to Gs or to stimulate adenylate cyclase, the deletion of either of the regions 222-229 or 258-270 resulted in receptors which were unable to couple to Gs. The affinities of D(222-229)beta AR, D(239-272)beta AR, and D(258-270)beta AR toward isoproterenol were greater than that observed for the low affinity, uncoupled form of the wild-type receptor. These results suggest a role for the regions of the beta-adrenergic receptor encompassing amino acids 222-229 and 258-270, which are predicted to form amphiphilic helices, in the agonist-promoted activation of Gs.
Article
Full-text available
The concept of intrinsic efficacy has been enshrined in pharmacology for half of a century, yet recent data have revealed that many ligands can differentially activate signaling pathways mediated via a single G protein-coupled receptor in a manner that challenges the traditional definition of intrinsic efficacy. Some terms for this phenomenon include functional selectivity, agonist-directed trafficking, and biased agonism. At the extreme, functionally selective ligands may be both agonists and antagonists at different functions mediated by the same receptor. Data illustrating this phenomenon are presented from serotonin, opioid, dopamine, vasopressin, and adrenergic receptor systems. A variety of mechanisms may influence this apparently ubiquitous phenomenon. It may be initiated by differences in ligand-induced intermediate conformational states, as shown for the beta(2)-adrenergic receptor. Subsequent mechanisms that may play a role include diversity of G proteins, scaffolding and signaling partners, and receptor oligomers. Clearly, expanded research is needed to elucidate the proximal (e.g., how functionally selective ligands cause conformational changes that initiate differential signaling), intermediate (mechanisms that translate conformation changes into differential signaling), and distal mechanisms (differential effects on target tissue or organism). Besides the heuristically interesting nature of functional selectivity, there is a clear impact on drug discovery, because this mechanism raises the possibility of selecting or designing novel ligands that differentially activate only a subset of functions of a single receptor, thereby optimizing therapeutic action. It also may be timely to revise classic concepts in quantitative pharmacology and relevant pharmacological conventions to incorporate these new concepts.
Article
Full-text available
Targeted recombinant aequorins represent to date the most specific means of monitoring [Ca²⁺] in subcellular organelles (Rizzuto, R., Simpson, A. W. M., Brini, M., and Pozzan, T. (1992) Nature 358, 325-328; Brini, M., [Medline] Murgia, M., Pasti, L., Picard, D., Pozzan, T., and Rizzuto, R. (1993) EMBO J. 12, 4813-4819; Kendall, J. M., Dormer, R. L., and Campbell, A. K. (1992) Biochem. Biophys. Res. Commun. 189, 1008-1016). Up until now, however, only limited attention has been paid to the use of recombinant photoproteins for measuring, in mammalian cells, the [Ca²⁺] in the cytoplasm, a compartment for which effective Ca²⁺probes are already available. Here we describe this approach in detail, highlighting the advantages, under various experimental conditions, of using recombinant cytosolic aequorin (cytAEQ) instead of classical fluorescent indicators. We demonstrate that cytAEQ is expressed recombinantly at high levels in transiently transfected cell lines and primary cultures as well as in stably transfected clones, and we describe a simple algorithm for converting aequorin luminescence data into [Ca²⁺] values. We show that although fluorescent indicators at the usual intracellular concentrations (50-100 μM) are associated with a significant buffering of the [Ca²⁺]ctransients, this problem is negligible with recombinantly expressed aequorin. The large dynamic range of the photoprotein also allows an accurate estimate of the large [Ca²⁺]cincreases that are observed in some cell types such as neurons. Finally, cytAEQ appears to be an invaluable tool for measuring [Ca²⁺]cin cotransfection experiments. In particular, we show that when cotransfected with an α1-adrenergic receptor (coupled to inositol 1,4,5-trisphosphate generation), cytAEQ faithfully monitors the subpopulation of cells expressing the receptor, whereas the signal of fura-2, at the population level, is dominated largely by that of the untransfected cells.
Article
Full-text available
Morphine, a powerful analgesic, and norepinephrine, the principal neurotransmitter of sympathetic nerves, exert major inhibitory effects on both peripheral and brain neurons by activating distinct cell-surface G protein–coupled receptors—the -opioid receptor (MOR) and 2A-adrenergic receptor (2A-AR), respectively. These receptors, either singly or as a heterodimer, activate common signal transduction pathways mediated through the inhibitory G proteins (Gi and Go). Using fluorescence resonance energy transfer microscopy, we show that in the heterodimer, the MOR and 2A-AR communicate with each other through a cross-conformational switch that permits direct inhibition of one receptor by the other with subsecond kinetics. We discovered that morphine binding to the MOR triggers a conformational change in the norepinephrine-occupied 2A-AR that inhibits its signaling to Gi and the downstream MAP kinase cascade. These data highlight a new mechanism in signal transduction whereby a G protein–coupled receptor heterodimer mediates conformational changes that propagate from one receptor to the other and cause the second receptor's rapid inactivation.
Article
Full-text available
Nanodiscs are nanometer scale planar membranes of controlled size that are rendered soluble in aqueous solution via an encircling amphipathic membrane scaffold protein "belt" (Bayburt, T. H., Grinkova, Y. V., and Sligar, S. G. (2002) Nano. Lett. 2, 853-856). Integral membrane proteins can be self-assembled into the Nanodisc bilayer with defined stoichiometry, which allows an unprecedented opportunity to investigate the nature of the oligomerization state of a G-protein-coupled receptor and its coupling to heterotrimeric G-proteins. We generated Nanodiscs having one and two rhodopsins present in the 10-nm-diameter lipid bilayer domain. Efficient transducin activation and isolation of a high affinity transducin-metarhodopsin II complex was demonstrated for a monodisperse and monomeric receptor. A population of Nanodiscs containing two rhodopsins was generated using an increased ratio of receptor to membrane scaffold protein in the self-assembly mixture. The two-rhodopsin population was isolated and purified by density gradient centrifugation. Interestingly, in this case, only one of the two receptors present in the Nanodisc was able to form a stable metarhodopsin II-G-protein complex. Thus there is clear evidence that a monomeric rhodopsin is capable of full coupling to transducin. Importantly, presumably due to steric interactions, it appears that only a single receptor in the Nanodiscs containing two rhodopsins can interact with G-protein. These results have important implications for the stoichiometry of receptor-G-protein coupling and cross talk in signaling pathways.
Article
Full-text available
Opsin, the ligand-free form of the G-protein-coupled receptor rhodopsin, at low pH adopts a conformationally distinct, active G-protein-binding state known as Ops*. A synthetic peptide derived from the main binding site of the heterotrimeric G protein-the carboxy terminus of the alpha-subunit (GalphaCT)-stabilizes Ops*. Here we present the 3.2 A crystal structure of the bovine Ops*-GalphaCT peptide complex. GalphaCT binds to a site in opsin that is opened by an outward tilt of transmembrane helix (TM) 6, a pairing of TM5 and TM6, and a restructured TM7-helix 8 kink. Contacts along the inner surface of TM5 and TM6 induce an alpha-helical conformation in GalphaCT with a C-terminal reverse turn. Main-chain carbonyl groups in the reverse turn constitute the centre of a hydrogen-bonded network, which links the two receptor regions containing the conserved E(D)RY and NPxxY(x)(5,6)F motifs. On the basis of the Ops*-GalphaCT structure and known conformational changes in Galpha, we discuss signal transfer from the receptor to the G protein nucleotide-binding site.
Article
The α-subunit of the stimulatory G protein, Gs, has been shown to dissociate from the plasma membrane into the cytosol following activation by G protein-coupled receptors (GPCR) in some experimental systems. This dissociation may involve depalmitoylation of an amino-terminal cysteine residue. However, the functional significance of this dissociation is not known. To investigate the functional consequence of Gsα dissociation, we constructed a membrane-tethered Gsα (tetGsα), expressed it in Sf9 insect cells, and examined its ability to couple with the β2 adrenoceptor and to activate adenylyl cyclase. Compared to wild-type Gsα, tetGsα coupled much more efficiently to the β2 adrenoceptor and the D1 dopamine receptor as determined by agonist-stimulated GTPγS binding and GTPase activity. The high coupling efficiency was abolished when Gsα was proteolytically cleaved from the membrane tether. The membrane tether did not prevent the coupling of tetGSα to adenylyl cyclase. These results demonstrate that regulating the mobility of Gsα relative to the plasma membrane, through fatty acylation or perhaps interactions with cytoskeletal proteins, could have a significant impact on receptor−G protein coupling. Furthermore, by enabling the use of more direct measures of receptor−G protein coupling (GTPase activity, GTPγS binding), tetGSα can facilitate the study for receptor−G protein interactions.
Article
The efficiency of interactions between G-protein-coupled receptors (GPCRs) and heterotrimeric guanine nucleotide-binding proteins (G proteins) is greatly influenced by the absolute and relative densities of these proteins in the plasma membrane. The study of these interactions has been facilitated by the use of GPCR–Gα fusion proteins, which are formed by the fusion of GPCR to Gα. These fusion proteins ensure a defined 1:1 stoichiometry of GPCR to Gα and force the physical proximity of the signalling partners. Thus, fusion of GPCR to Gα enhances coupling efficiency can be used to study aspects of receptor–G-protein coupling that could not otherwise be examined by co-expressing GPCRs and G proteins as separate proteins. The results of studies that have made use of GPCR–Gα fusion proteins will be discussed in this article, along with the strengths and limitations of this approach.
Article
Fusion proteins between heptahelical receptors (GPCR) and G protein α-subunits show enhanced signaling efficiency in transfected cells. This is believed to be the result of molecular proximity, because the interaction between linked modules of one protein chain, if not constrained by structure, should be strongly favored compared with the same in which partners react as free species. To test this assumption we made a series of fusion proteins (type 1 and 4 opioid receptors with Go and β2adrenergic and dopamine 1 receptors with GsL) and some mutated analogs carrying different tags and defective GPCR or Gα subunits. Using cotransfection experiments with readout protocols able to distinguish activation at fused and non-fused α-subunits, we found that both the GPCR and the Gα limb of one fusion protein can freely interact with non-fused proteins and the tethered partners of a neighboring fusion complex. Moreover, a bulky polyanionic inhibitor can suppress with identical potency receptor-Gα interaction, either when occurring between latched domains of a fused system or separate elements of distinct molecules, indicating that the binding surfaces are equally accessible in both cases. These data demonstrate that there is no entropy drive from the linked condition of fusion proteins and suggest that their signaling may result from the GPCR of one complex interacting with the α-subunit of another. Moreover, the enhanced coupling efficiency commonly observed for fusion proteins is not due to the receptor tether, but to the transmembrane helix that anchors Gα to the membrane.