19
267 From: Contemporary Clinical Neuroscience: The G Protein-Coupled Receptors Handbook Edited by: L. A. Devi © Humana Press Inc., Totowa, NJ 12 Functional Complementation and the Analysis of GPCR Dimerization Graeme Milligan, Juan J. Carrillo, and Geraldine Pascal 1. INTRODUCTION Cloning of complementary DNAs (cDNAs), which are predicted to encode G protein-coupled receptors (GPCRs), required a mechanism to ascertain if the single polypeptide encoded by such cDNAs was sufficient to generate the pharmacology and function anticipated for the receptor in question. Because this generally was the case—and despite evidence of greater com- plexity (reviewed in ref. 1)—it became axiomatic that GPCRs were single, seven-transmembrane-span polypeptides. However, a series of immunoblotting studies has suggested that a fraction of cellular GPCRs might exist as dimers or higher order species in both transfected cell lines and na- tive tissues (2–4). The known propensity of hydrophobic proteins to aggre- gate—particularly when samples were heated prior to separation by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE)—meant that it was possible to disregard these data. However, studies in which differ- entially epitope-tagged forms of a single GPCR could be co-immunoprecipi- tated following their co-expression in heterologous cell lines provided considerable evidence for the presence of dimeric or, indeed, higher oligo- meric complexes (2,5). This was generally not observed when the two forms of the GPCR were expressed in separate cell populations that were mixed prior to membrane solubilization and immunoprecipitation. These results in- dicate that co-immunoprecipitation did not result simply from aggregation of the hydrophobic transmembrane elements of these polypeptides following removal of lipid by treatment with detergents. Equivalent studies then began to examine the proclivity of different, co-expressed GPCRs to be co-immu- noprecipitated. These studies have produced a large body of evidence sup-

[Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

Embed Size (px)

Citation preview

Page 1: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

Functional Complementation 267

267

From: Contemporary Clinical Neuroscience: The G Protein-Coupled Receptors HandbookEdited by: L. A. Devi © Humana Press Inc., Totowa, NJ

12Functional Complementation and the

Analysis of GPCR Dimerization

Graeme Milligan, Juan J. Carrillo, and Geraldine Pascal

1. INTRODUCTION

Cloning of complementary DNAs (cDNAs), which are predicted to encodeG protein-coupled receptors (GPCRs), required a mechanism to ascertain ifthe single polypeptide encoded by such cDNAs was sufficient to generatethe pharmacology and function anticipated for the receptor in question.Because this generally was the case—and despite evidence of greater com-plexity (reviewed in ref. 1)—it became axiomatic that GPCRs were single,seven-transmembrane-span polypeptides. However, a series ofimmunoblotting studies has suggested that a fraction of cellular GPCRs mightexist as dimers or higher order species in both transfected cell lines and na-tive tissues (2–4). The known propensity of hydrophobic proteins to aggre-gate—particularly when samples were heated prior to separation by sodiumdodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE)—meantthat it was possible to disregard these data. However, studies in which differ-entially epitope-tagged forms of a single GPCR could be co-immunoprecipi-tated following their co-expression in heterologous cell lines providedconsiderable evidence for the presence of dimeric or, indeed, higher oligo-meric complexes (2,5). This was generally not observed when the two formsof the GPCR were expressed in separate cell populations that were mixedprior to membrane solubilization and immunoprecipitation. These results in-dicate that co-immunoprecipitation did not result simply from aggregation ofthe hydrophobic transmembrane elements of these polypeptides followingremoval of lipid by treatment with detergents. Equivalent studies then beganto examine the proclivity of different, co-expressed GPCRs to be co-immu-noprecipitated. These studies have produced a large body of evidence sup-

Page 2: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

268 Milligan, Carrillo, and Pascal

porting the capacity of GPCRs to exist as heterodimers/-oligomers (for re-view, see refs. 6 and 7). However, studies that have reported a very broadcapacity of particular GPCRs to allow co-immunoprecipitation of co-ex-pressed GPCRs (8) have questioned the relevance of GPCR co-immunopre-cipitation data when this is not accompanied by similar data produced byalternative strategies.

There is also a concern that many “co-immunoprecipitation” studies arepoorly performed and controlled. For example, many studies in this areacentrifuge samples to remove particulate material after detergent treatmentto generate a “soluble” preparation for the immunoprecipitation steps. Often,however, the centrifugation step is performed for only short periods, and thecentrifugal force is too low to achieve this end. This inevitably results insmall membrane fragments being present in the “soluble” fraction and thepossibility that “co-immunoprecipitation” represents nothing more than bothpolypeptides presenting within the same membrane fragment.

These types of issues have resulted in the widespread adoption and use oftechniques that can monitor protein–protein interactions in either membranepreparations or intact living cells. Various forms of resonance energy trans-fer techniques have been employed to explore GPCR dimerization/oligo-merization because of their relative simplicity and exquisite dependence ondistance between the energy donor and acceptor species (9–14). These issuesand the utility of the systems have recently been reviewed (15,16) and arediscussed further within this volume (17). An alternative strategy that canbe used instead of, or in parallel with, resonance energy transfer techniquesis functional complementation produced following co-expression of pairs ofdistinct GPCR or GPCR–G protein fusion mutants.

2. COMPLEMENTATION OF PAIRS OF MUTANT GPCRS

GPCRs can be assembled from co-expressed fragments (12,18). This hasled to the hypothesis that segments of GPCRs can be considered distinctdomains capable of independent folding. Extensions of this idea have alsoallowed the envisaging of models of GPCR dimers that include explicit re-quirement for domain swapping (19,20). Although direct experimental evi-dence in favor of such models is somewhat limited, such evidence wouldpotentially explain marked variations in ligand pharmacology that are some-times observed when two GPCRs capable of heterodimer formation are co-expressed (21) or when chimeric GPCRs are constructed (21,22). Reciprocalchimeric GPCRs between the α2C-adrenoceptor and the m3 muscarinic ace-tylcholine receptor that contained the N-terminus and transmembrane seg-ments I–V of one receptor and transmembrane segments VI–VII and the

Page 3: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

Functional Complementation 269

C-terminal tail of the second bound neither muscarinic or α2-adrenergicligands. However, the co-expression of the two chimeras resulted in regen-eration of some level of high-affinity binding for both muscarinic and α2-adrenergic antagonists (22). These workers were also able to reconstituteagonist-mediated signal transduction when pairs of distinct, functionally im-paired mutant muscarinic receptors were co-expressed (22). Related studieson the angiotensin II type 1 receptor showed that forms of this GPCR withspecific mutations in either transmembrane region III or transmembrane seg-ment V were unable to bind ligands. However, ligand binding was observedupon co-expression (23). These studies were central in providing evidence infavor of molecular interactions between GPCRs. Despite these conclusions,the number of ligand-binding sites produced following co-expression waslow, and it is unclear if this simply reflects that the individual mutations arevery poorly expressed compared to the wild-type protein, or if the efficiencyof “reconstitution” is poor. One potential explanation is that if the reconstitu-tion of high-affinity ligand binding requires a domain exchange mechanism,then this may be thermodynamically feasible but uncommon compared withdimerization based on linear packing, which would not be anticipated toregenerate ligand binding on co-expression of the two mutants.

The first definitive evidence of GPCR hetero-dimerization was the rec-ognition that the γ-aminobutyric acid (GABA)BR1 polypeptide could bindagonist (although with lower than the anticipated affinity) and antagonistligands but could not generate signals or be trafficked effectively to the cellsurface unless co-expressed with the GABABR2 polypeptide (24,25). Simi-larly to other family C GPCRs, the long extracellular N-terminal extensionbinds the ligand, but the architecture of the prototypic seven-transmembranecore defines the signal transduction unit. Because such GPCRs can be con-sidered bifunctional polypeptides, chimeras between the extracellular re-gion and the transmembrane segments and intracellular elements have beengenerated to aid understanding of the mechanisms of signal propagation,from ligand binding to G protein activation (26).

Several family A GPCRs that respond to large glycoprotein hormonesalso have long extracellular N-termini and, therefore, are amenable to re-lated approaches. One well-studied example is the luteinizing hormone re-ceptor. This GPCR has been used to generate differentially defective mutantpairs—one in the extracellular exodomain that lacks binding of the ligandhuman chorionic gonadotropin and a second that can bind the ligand but isunable to signal because of a mutation in the endodomain (27,28). Their co-expression resulted in the restoration of ligand generation of the secondmessenger cyclic adenosine monophosphate (cAMP). Again, although this

Page 4: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

270 Milligan, Carrillo, and Pascal

provided a persuasive case for intermolecular interactions among GPCRs,the amplification mechanisms resulting in cAMP generation (which alsooften result in the characteristic of “spare receptors”) do not allow for easyassessment of the effectiveness of the complementation process.

It has been suggested that unlike many other GPCRs, the luteinizing hor-mone receptor does not form stable dimers. Such functional complementa-tion studies can provide evidence for both cis-activation (i.e., ligand boundto the exodomain, causing activation on the same endodomain) or trans-activation, where the ligand activates the partner endodomain within a dimer(29). It is noteworthy that such a model may also explain the complex inter-actions between activated protease-activated receptor (PAR)3 and PAR4GPCRs (30) and, potentially, other complex interactions between PAR sub-types (31). The glycoprotein hormones and PARs are unusual examples offamily A, rhodopsin-like GPCRs. For the bulk of these, the agonist ligandsbind at least partially within the cavity created by the seven-transmembranehelix architecture. We wished to develop a generic functional complemen-tation strategy that would be suitable to study both homo- andheterodimerization of family A GPCRs and that would provide quantitativeassays for the effectiveness of complementation. Therefore, we employedpairs of nonfunctional, but potentially complementary, GPCR–G protein α-subunit fusion proteins.

3. FUNCTIONAL COMPLEMENTATION OF PAIRSOF GPCR–G PROTEIN FUSIONS

3.1. GPCR–G Protein Fusions

The first GPCR–G protein fusion was constructed between the β2-adrenoceptor and the α-subunit of the long isoform of the G protein Gs.When this was expressed in S49 cyc– cells lacking expression of Gαs, the β-adrenoceptor agonist isoprenaline was able to stimulate adenylyl cyclase(32), thus confirming the functionality of both elements of the fusion con-struct. Although the fusion construct had several interesting characteristics(32), it was initially viewed as little more than a curiosity. However, overthe intervening period, a wide range of GPCRs and G proteins have beenused to generate similar fusions (33,34). These have been used to addressissues such as the selectivity of GPCRs in activating different G proteins(35), the role and regulation of posttranslational acylation in the functionand cellular location of GPCRs and G proteins (36–38), as reagents to screenfor agonists at “orphan” GPCRs (39,40) and, most recently, to explore thebasis and selectivity of GPCR dimerization (41).

Page 5: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

Functional Complementation 271

GPCR–G protein fusions can also be considered as bifunctional polypep-tides because, although they are generated from a single open frame, theycontain the sequence and functionality of both GPCR and G protein. There-fore, mutants that eliminate the ability of agonists to generate a signal corre-sponding to activation of the G protein can be produced by alterations in thesequence of either the GPCR or the G protein element. This allows for theproduction of distinct pairs of fusions that are individually inactive but havethe potential to complement function if they interact. Two obvious regionscan be targeted for the G protein segment of the fusion. All G protein α-subunits have a conserved Val-Gly-Gly-Gln-Arg sequence (Table 1), wheremutation of the second Gly to Ala generates a form of the G protein that isunable to exchange guanosine diphosphate (GDP) for guanosine triphos-phate (GTP) and hence become activated. This provides a simple strategy togenerate GPCR–G protein fusion proteins that are unable to respond to ago-nist ligands even though the GPCRs linked to such mutants are wild-typeand able to bind ligands. Equally, because it is well established that the ex-treme C-terminal region of most G protein α-subunits is a key contact do-main for GPCR-mediated activation (42), judicious mutation in this regioncan produce forms of the G protein that are not responsive to GPCRs. Forexample, because pertussis toxin-catalyzed adenosine diphosphate (ADP)-ribosylation of the Cys residue four amino acids from the C-terminus of allwidely expressed Gi-family G protein α-subunits prevents GPCR-mediatedactivation of these G proteins, Bahia et al. (43) replaced this residue in Gαi1with each of the other naturally occurring amino acids and then assessed theability of each of the mutants to be activated by agonist occupancy of theα2A-adrenoceptor. Substitution of the Cys with more hydrophilic amino ac-ids reduced coupling effectiveness, and there was no significant activationwith either positively or negatively charged amino acids at this position.Similar data have been produced for activation of equivalently mutatedforms of Gαi3 by the 5-HT1A receptor (44). Several of the Gαi1 mutationshave been constructed into fusion proteins with the α2A-adrenoceptor andhave been used to monitor the effects on these alterations on informationtransfer from GPCR to G protein, which was measured as alterations in ago-nist potency and relative efficacy (45,46). Similarly, mutation of the Tyrfour amino acids from the C-terminal of Gα11 resulted in reduction of itsability to be activated by the α1b-adrenoceptor (47). With Asp at this posi-tion, no activation was observed by the agonist-occupied α1b-adrenoceptor,both in co-expression studies and when the modified G protein was con-structed into a fusion with this GPCR (47). Although it has not been exploredin such detail, it is well known that the lack of function of Gαs in the S49

Page 6: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

272 Milligan, Carrillo, and Pascal

unc cell line reflects an Arg to Pro substitution six amino acids from the C-terminus of this G protein (48). Therefore, although the details of contactsbetween different GPCRs and a specific G protein differ subtly (49), alter-ations in the C-terminal tail of the G protein can produce forms than cannotbe activated by the GPCR of choice.

Table 1A Generic Strategy to Generate Inactive G Protein α-Subunits

G α-subunit Species Sequence

Gs Human VGGQRDERRKGolf Human VGGQRDERRKGi1 Human VGGQRSERKK

Rat VGGQRSERKKGi2 Human VGGQRSERKK

Mouse VGGQRSERKKGi3 Human VGGQRSERKK

Rat VGGQRSERKKGo1 Human VGGQRSERKK

Bovine VGGQRSERKKGo2 Human VGGQRSERKKGz Rat VGGQRSERKK

Bovine VGGQRSERKKTransducin1 Human VGGQRSERKKTransducin2 Rat VGGQRSERKKGustducin Human VGGQRSERKKGq Mouse VGGQRSERKK

Human VGGQRSERKKG11 Rat VGGQRSERKK

Human VGGQRSQRQKG12 Rat VGGQRSQRQK

Human VGGQRSERKRG13 Human VGGQRSERRKG14 Mouse VGGQRSERRKG15 Human VGGQRSERKKG16

G protein α-subunits are highly conserved between mammalian spe-cies, particularly in core domains involved in the binding and hydroly-sis of guanine nucleotides. Alteration of the highlighted glycine residueto alanine results in forms of the G protein that are unable to bind GTP.This provides a generic means to generate G proteins that are unable tobe activated by G protein-coupled receptors (GPCRs) and thus to pro-duce one of the pair of inactive GPCR–G protein fusion proteins used inthe functional complementation strategy.

Page 7: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

Functional Complementation 273

A range of potential strategies are available to produce inactive GPCR–Gprotein fusions via mutation of the GPCR. However, the most generic (atleast, for many rhodopsin-like class A GPCRs) appears to be via mutation ofone or more highly conserved hydrophobic residues in the second intracel-lular loop of the GPCR to acidic residues. The first of these is frequentlythree amino acids downstream of the highly conserved DRY domain, andthe second is four amino acids further downstream (Table 2).

3.2. Complementation of GPCR–G Protein Fusionsin Cell Membranes

A useful feature of GPCR–G protein fusions is that the defined 1:1 stoichi-ometry ensures that saturation ligand-binding studies define the expressionlevels of the G protein as well as the GPCR. Agonist stimulation of the bind-ing of [35S]GTPγS (50) to such fusion proteins can then provide a proximal,and potentially quantitative, assay of G protein activation which directexperiments have shown to be linear over a good range of membrane amounts(38). It is important to demonstrate that agonist-mediated binding of[35S]GTPγS is truly to the G protein element of the fusion rather than to en-dogenously expressed G proteins. This is an important issue because—at least,for some fusion constructs—it is clear that activation of endogenously ex-pressed G proteins can occur (51,52). However, generally, this seems to be aserious concern only with high-level expression of the GPCR–G protein fu-sion or when a free G protein is also co-expressed at high levels (53). Forfusions incorporating members of the Gi family of G proteins, mutation of thepertussis toxin-sensitive Cys residue to a hydrophobic amino acid, such asIle, prevents pertussis toxin-catalyzed ADP-ribosylation without inhibitingGPCR-mediated activation of the G protein (45,51). Therefore, followingtransfection of cells to express a GPCR–G protein fusion containing such apertussis toxin-resistant mutant, treatment of the cells with pertussis toxinresults in ADP-ribosylation of only the endogenously expressed forms of Gi.Binding of [35S]GTPγS to these expressed forms of Gi hence cannot be stimu-lated by agonist. Therefore, following immunoprecipitation with an anti-Gαiantiserum, agonist-enhanced binding of the nucleotide must reflect loading ofthe G protein element of the fusion protein. For example, membranes pre-pared from pertussis toxin-treated HEK293 cells expressing a δ-opioid pep-tide (DOP)-opioid receptor–Cys351IleGαi1 fusion protein (15 fmol of[3H]antagonist binding sites) were subjected to a [35S]GTPγS binding assayin the absence or presence of the agonist D-ala2, D-leu5-enkephalin (DADLE)and were then subjected to immunoprecipitation. Radioactivity in the pres-ence of the agonist was approximately sixfold higher than in its absence, con-

Page 8: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

274 Milligan, Carrillo, and Pascal

Table 2A Generic Strategy to Generate GPCR Variants Unable to Activate G Proteins

GPCR Specie G protein Sequence

5-HT1A receptor Human Gi/Go DRYWAITDPID5-HT1B receptor Mouse Gi/Go DRYWAITDAVE5-HT1D receptor Rabbit Gi/Go DRYWAITDALE5-HT2A receptor Rat Gq/G11 DRYVAIQNPIH5-HT2C receptor Rat Gq/G11 DRYVAIRNPIE5-HT4 receptor Mouse Gs DRYYAICCQPL5-HT6 receptor Rat Gs DRYLLILSPLRα1a-adrenoceptor Bovine Gq/G11 DRYIGVSYPLRα1b-adrenoceptor Hamster Gq/G11 DRYIGVRYSLQα2b-adrenoceptor Rat Gi/Go DRYWAVSRALEβ1-adrenoceptor Human Gs DRYLAITSPFRβ2-adrenoceptor Bovine Gs DRYLAITSPFKβ3-adrenoceptor Mouse Gs DRYLAVTNPLRA1 adenosine receptor Human Gi/Go DRYLRVKIPLRA3 adenosine receptor Human Gi/Go DRYLRVKLTVRAcetylcholine M1 receptor Mouse Gq/G11 DRYFSVTRPLSAcetylcholine M2 receptor Human Gi/Go DRYFCVTKPLTAcetylcholine M3 receptor Mouse Gq/G11 DRYFSITRPLTAngiotensin AT1A receptor Human Gq/G11 DRYLAIVHPMKAngiotensin AT1B receptor Rat Gq/G11 DRYLAIVHPMKBradykinin B2 receptor Human Gq/G11 DRYLALVKTMSChemokine CXCR3 Mouse Gi/Go DRYLSIVHATQChemokine CXCR4 Human Gi/Go DRYLAIVHATNDopamine D2 receptor Mouse Gi/Go DRYTAVAMPMLDopamine D3 receptor Rat Gi/Go DRYTAVVMPVHFSH receptor Bovine Gs ERWHTITHAMQGnRH receptor Mouse Gq/G11 DRSLAITQPLAHistamine H1 receptor Mouse Gq/G11 DRYRSVQQPLRHistamine H2 receptor Human Gs DRYCAVMDPLRLH receptor Mouse Gs ERWHTITYAVQMelanocortin 2 receptor Human Gs DRYITIFHALRDOP-opioid receptor Rat Gi/Go, Gz DRYIAVCHPVKKOP-opioid receptor 1 Mouse Gi/Go DRYIAVCHPVKMOP-opioid receptor 1 Rat Gi/Go, Gz DRYIAVCHPVKRhodopsin Bovine Gt ERYVVVCKPMSOxytocin receptor Rat Gq/G11 DRCLAICQPLRP2U purinoceptor 1 Rat Gq/G11 HRCLGVLRPLHProstaglandin D2 receptor Mouse Gs ECWLSLGHPFFProstaglandin E2 receptor Rat Gq/G11 ERCVGVTQPLISomatostatin receptor 2 Human Gi/Go Gq DRYLAVVHPIKThyrotropin-releasing Rat Gq/G11 ERYIAICHPIK

hormone receptor-1

The vast majority of class A, rhodopsin-like G protein-coupled receptors (GPCRs) possess a pairof hydrophobic residues (highlighted) in the second intracellular loop, downstream of the highly

continued

Page 9: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

Functional Complementation 275

firming the functionality of this construct. When similar experiments wereperformed with a version of this fusion protein that also incorporated aGly203Ala mutation in the G protein, DADLE did not enhance incorporationof the nucleotide. This was also true for a version of the fusion that incorpo-rated Val150Glu and Val154Asp mutations into the second intracellular loop ofthe GPCR element but that was wild-type for Gαi1, other than the pertussistoxin-resistant Cys351Ile mutation. However, when Val150Glu,Val154AspDOP–Cys351IleGαi1 was co-expressed with DOP–Gly203Ala,Cys351IleGαi1membranes prepared after treatment with pertussis toxin and containing 15fmol of DOP-opioid receptor binding sites displayed marked reconstitution of[35S]GTPγS binding in response to DADLE (Fig. 1). Assuming that theintracellular loop 2 mutations do not interfere with DOP–DOP interactions,that GPCRs function as dimers, that dimers bind two ligands, and thatVal150Glu,Val154Asp DOP–Cys351IleGαi1 and DOP–Gly203Ala,Cys351IleGαi1express to equal extents in the cotransfections, simple arithmetic defines thatthe heterodimer (containing one copy of each of Val150Glu,Val154Asp DOP–Cys351IleGαi1 and DOP–Gly203Ala,Cys351IleGαi1) should represent only 50%of the ligand-binding sites. Of the other 50%, 25% would be expected to beVal150Glu,Val154Asp DOP–Cys351IleGαi1 homodimers, and 25% would beexpected to be DOP-Gly203Ala,Cys351IleGαi1 homodimers.

As demonstrated earlier, neither the Val150Glu,Val154Asp DOP–Cys351IleGαi1 nor the DOP–Gly203Ala,Cys351IleGαi1 homodimers bind[35S]GTPγS in response to agonists. Therefore, conceptually, following co-expression, membranes containing 30 fmol of antagonist binding sites couldbe anticipated to bind the same level of [35S]GTPγS in response to agonist asmembranes expressing 15 fmol of the DOP–Cys351Ile Gαi1 fusion protein.Reconstitution was not this effective (Fig. 1). This may imply that the qua-ternary structure of the GPCR–G protein fusion is a higher order oligomerrather than a dimer or that there is a complex mixture of species that mayalso include monomers. Further analyses are required to test these ideas.

Table 2 (From opposite page) conserved ’DRY’ domain at the bottom of transmembraneregion III. In the examples we tested, alteration of one or both of these to acidic residuesresulted in forms of the GPCR that are unable to activate G protein. This provides a poten-tially generic means to form an inactive GPCR–G protein fusion protein. Such mutations donot affect the binding of antagonist ligands. The effect on the binding of agonist ligands ismore complex and generally reflects whether the GPCR in question shows a significant alter-ation in agonist binding in response to addition of guanine nucleotides. For example, manyGq/G11-coupled GPCRs display only a marginal effect on agonist binding affinity; however,for Gs and Gi/Go-coupled receptors, this alteration in affinity can be substantial.

Page 10: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

276 Milligan, Carrillo, and Pascal

Fig. 1. Functional complementation of agonist-mediated [35S]GTPγS binding byco-expression of pairs of inactive DOP-opioid receptor–Gαi1 fusion proteins. A fu-sion protein was created between the human DOP-opioid receptor and a pertussistoxin-resistant Cys351Ile mutant of Gαi1. Further modifications to this fusion intro-duced either Val150Glu,Val154Asp mutations into the receptor element or Gly203Alainto the G protein. Each of these three fusion proteins was transfected into HEK293cells that were then treated with pertussis toxin to cause ADP-ribosylation ofendogenously expressed forms of Gαi/Gαo. Expression levels of the fusion proteinswere measured by the specific binding of the opioid receptor antagonist[3H]diprenorphine and membrane amounts containing 15 fmol of each fusion pro-tein used in [35S]GTPγS binding studies, which were conducted in the absence orpresence of the synthetic enkephalin DADLE (10 μM). At the termination of assay,samples were immunoprecipitated with an antiserum directed to the C-terminaldecapeptide of Gαi1 and were then counted. Equivalent studies were also performedon membranes of cells into which Val150Glu,Val154Asp DOP-opioid receptor–Cys351IleGαi1 and DOP-opioid receptor–Gly203Ala,Cys351IleGαi1 were co-expressed. Co-expression reconstituted DADLE-stimulated [35S]GTPγS binding, butthis was not produced when membrane preparations individually expressing eitherVal150Glu,Val154Asp DOP-opioid receptor–Cys351IleGαi1 or DOP-opioid receptor–Gly203Ala,Cys351IleGαi1 were mixed.

Page 11: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

Functional Complementation 277

Importantly, however, co-expression is required to reconstitute function.Membranes prepared from cells each expressing one of the inactive fusionconstructs did not allow agonist-stimulated binding of [35S]GTPγS whenmixed. Equivalent fusion constructs incorporating the μ-opioid peptide(MOP)-opioid receptor have similar characteristics, and reconstitution ofagonist function is again achieved following co-expression of the pair ofindividually inactive fusion constructs. Co-expression of one inactive fusioncontaining the MOP-opioid receptor, with the partner containing the DOP-opioid receptor, also results in reconstitution of function and is consistentwith the formation of a MOP–DOP heterodimer (54,55).

Other G proteins are not targets for pertussis toxin-catalyzed ADP-ribosylation, but are widely expressed. Therefore, other strategies are re-quired to ensure that enhanced binding of [35S]GTPγS truly reflectsactivation of the G protein element of the fusion, rather than endogenous Gprotein. The α1-adrenoceptor agonist phenylephrine elevates binding of[35S]GTPγS following expression of a fusion protein between the α1b-adrenoceptor and Gα11 in HEK293 cells and membrane preparation (53).However, nucleotide binding in the absence of agonist is high, thus limitingthe agonist-induced signal to background. The vast majority of the agonist-independent binding is not to either the G protein within the fusion constructor to endogenously expressed Gα11 and/or Gαq. When samples were immu-noprecipitated with an anti-Gα11/Gαq antiserum, virtually all the agonist-independent binding was lost, but because the agonist-mediated signal wasmaintained, the signal to background ratio was increased greatly (53). Thisdid not establish whether all (or any) of the [35S]GTPγS was bound to the Gprotein element of the fusion protein, because both the fusion construct andthe endogenously expressed G proteins were pulled down. However, fol-lowing expression of an α1b-adrenoceptor–Gly208AlaGα11 fusion protein,virtually no agonist-stimulated binding of [35S]GTPγS was observed in suchimmunoprecipitates (53). These results imply that at the levels of expres-sion achieved, the receptor in this fusion cannot access and activate the endo-genous pools of Gα11/Gαq to any significant degree. Therefore, theagonist-stimulation of [35S]GTPγS binding produced in membranes express-ing the wild-type α1b-adrenoceptor–Gα11 fusion is a direct measure of acti-vation of the fusion protein. A fusion between Leu151Aspα1b-adrenoceptorand wild-type Gα11 also generated very little agonist-dependent [35S]GTPγSbinding (41), defining the poor ability of this mutated receptor to activate Gprotein. Similarly to the opioid receptor constructs discussed earlier, co-expression of Leu151Aspα1b-adrenoceptor–Gα11 and α1b-adrenoceptor–Gly208AlaGα11 resulted in reconstitution of agonist function consistent with

Page 12: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

278 Milligan, Carrillo, and Pascal

Fig. 2. Detection of α1b-adrenoceptor dimerization by functional complementa-tion of a pair of inactive α1b-adrenoceptor–Gα11 fusion proteins using single-cellCa2+ imaging studies. A fusion protein was constructed between the hamster α1b-adrenoceptor and Gα11. This construct was transfected into a mouse embryo fibro-blast cell line (EF88 cells) derived from a double Gαq/Gα11 knockout mouse alongwith green fluorescent protein (GFP). Imaging of these cells (top panel left) showsonly a single positive cell in the field, and this was the only cell that responded tophenylephrine (basal vs peak [Ca2+]). Because these cells do not express any Gprotein able to induce Ca2+ signaling, these data demonstrate the functionality of

Page 13: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

Functional Complementation 279

dimerization of the α1b-adrenoceptor bringing the wild-type G protein andwild-type receptor into contact (41). Similar data have been obtained forfusion proteins between the histamine H1 receptor and Gα11.

As mentioned earlier, the binding affinity of both antagonist and agonistligands for the α1b-adrenoceptor–Gα11 fusion protein was unaffected byintroduction of the Leu151Asp mutation that blocked agonist activation. Thismay seem rather surprising, because the affinity of agonist ligands is oftenmodulated by the interaction status of GPCR and G protein, and such effectshave been observed for other GPCR–G protein fusions (56). However, gua-nine nucleotide-induced shifts in agonist affinity are well established to bevery small for α1-adrenoceptors. Significant alterations in agonist affinitymight be observed with the introduction of equivalent mutations in otherGPCR–G protein fusions.

3.3. Complementation Between GPCR–G Protein Fusionsin Intact Cells

When the α1b-adrenoceptor–Gα11 fusion protein was expressed in EF88cells, addition of phenylephrine resulted in elevation of [Ca2+]i (41,47). Thisreflects functionality of the fusion protein because EF88 cells are a line ofmouse embryo fibroblasts derived from an animal in which the genes for bothof the widely expressed Ca2+ mobilizing G proteins, Gα11 and Gαq had beeninactivated (57). Earlier studies demonstrated the requirement for co-expres-sion of both a suitable GPCR and an appropriate G protein in these cells togenerate function (38). Furthermore, only cells that are positively transfectedrespond to the agonist. The poor transfection efficiency of these cells whenusing cationic lipid-based methods means that analysis has generally requiredthe use of single-cell Ca2+ imaging (38,41). However, retrovirally basedinfection or the development of other effective means of transfection wouldallow the use of standard, cuvet-based measurements of Ca2+ levels and easiergeneration of concentration–response curves. As suggested under Subhead-ing 3.2., transfection of these cells with a fusion protein between the wild-type α1b-adrenoceptor and Gly208AlaGα11 failed to generate an elevation in

Fig. 2 (From opposite page) the fusion protein. The detailed time-course of [Ca2+]regulation in response to phenylephrine is shown in the bottom panel (top-mostline). The fusion was modified to introduce a Leu151Asp mutation into the receptorelement (bottom-most line) or a Gly208Ala mutation into the G protein (second linefrom bottom). When expressed in EF88 cells, neither of these mutants responded tophenylephrine; however, agonist-mediated signaling was reconstituted when theywere co-expressed (second line from top).

Page 14: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

280 Milligan, Carrillo, and Pascal

[Ca2+]i in response to phenylephrine (41). This was also the case forLeu151Aspα1b-adrenoceptor–Gα11. Similarly to the membrane-based assays,co-expression of Leu151Aspα1b-adrenoceptor–Gα11 and α1b-adrenoceptor–Gly208AlaGα1111 resulted in a restoration of phenylephrine-mediated eleva-tion of [Ca2+]i (Fig. 2). Because there are no appropriate endogenous Gproteins in these cells, such data require interactions between the complemen-tary fusions. Similar results were obtained when equivalent fusion proteinsthat incorporated the histamine H1 receptor rather than the α1b-adrenoceptorwere used (41). When Leu151Aspα1b-adrenoceptor–Gα11 and histamine H1–Gly208AlaGα11 were co-expressed, histamine, but not phenylephrine, wasable to elevate [Ca2+]i. The reverse was true when α1b-adrenoceptor–Gly208AlaGα11 was co-expressed with Leu133Asp histamine H1–Gα11 (41).

A weakness of single-cell imaging studies is the impossibility of moni-toring expression levels of each construct. However, when the wild-typehistamine H1 receptor–Gα11 fusion was co-expressed with the isolatedLeu151Aspα1b-adrenoceptor, the capacity of histamine to elevate [Ca2+]i wasreduced. Although these studies are more difficult to interpret than the par-allel experiments performed in membranes of HEK293 cells (in which theexpression levels of both the histamine H1 receptor–Gα11 fusion andLeu151Aspα1b-adrenoceptor could be measured directly by [3H]ligand-bind-ing studies; see Subheading 3.2.), their results are consistent with theLeu151Aspα1b-adrenoceptor forming a nonfunctional heterodimer with thehistamine H1 receptor–Gα11 fusion protein and thus reducing levels of func-tional histamine H1 receptor–Gα11/histamine H1 receptor–Gα11homodimer. This type of strategy should be generally applicable for GPCRsthat couple to the Ca2+ mobilizing G proteins Gq and G11, and may be appro-priate to measure the relative interaction affinities among GPCRs.

4. FUTURE PERSPECTIVES

Potentially, although the subject has yet to be directly explored, studiesakin to those described earlier could be expanded. Many GPCRs can inter-act with the so-called “promiscuous” G proteins G15 and G16 (58). Fusionproteins that incorporate either G15 or G16 with several GPCRs that do notroutinely elevate [Ca2+]i have been generated and have been used to allowligand screening and detailed pharmacological characterization using plat-forms such as the fluorescence imaging plate reader system, which is widelyemployed by the pharmaceutical industry (59–61). Incorporation of aGly211Ala form of Gα16 into a fusion should render it unresponsive to ago-nists and allow equivalent single-cell reconstitution assays, as detailed underSubheading 3.3. Equally, chimeric G proteins containing the backbone of a

Page 15: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

Functional Complementation 281

Ca2+-mobilizing G protein (with the extreme C-terminal tail modified toprovide interaction with GPCRs that couple selectively to Gi-family or Gs Gproteins) have been widely employed (62–64) to channel signal to a com-mon and easy-to-measure assay endpoint. GPCR–G protein fusions con-taining chimeric G proteins have also been generated and have been shownto be functional (65,66). Selectivity of GPCR interactions can be quantita-tively measured from the extent of reconstitution of [35S]GTPγS binding,with expression of known amounts of two GPCR fusions monitored by satu-ration ligand-binding studies. Equally, the domains and amino acids thatprovide the interfaces of GPCR dimerization should be amenable to analy-sis via mutagenesis studies followed by reconstitution.

ACKNOWLEDGMENTS

Work in the authors’ laboratory in this area is supported by the MedicalResearch Council, the Biotechnology and Biological Sciences ResearchCouncil, and the Wellcome Trust.

REFERENCES

1. Salahpour A, Angers S, Bouvier M. Functional significance of oligomerizationof G-protein-coupled receptors. Trends Endocrinol Metab 2000;11:163–168.

2. Hebert TE, Moffett S, Morello JP, et al. A peptide derived from a beta2-adren-ergic receptor transmembrane domain inhibits both receptor dimerization andactivation. J Biol Chem 1996;271:16,384–16,392.

3. Ng GYK, O’Dowd BF, Lee SP, et al. Dopamine D2 receptor dimers and recep-tor-blocking peptides. Biochem Biophys Res Comm 1996;227:200–204.

4. Nimchinsky EA, Hof PR, Janssen WG, Morrison JH, Schmauss C. Expressionof dopamine D3 receptor dimers and tetramers in brain and in transfected cells.J Biol Chem 1997;272:29,229–29,237.

5. Cvejic S, Devi LA. Dimerization of the delta opioid receptor: implication for arole in receptor internalization. J Biol Chem 1997;272:26,959–26,964.

6. George SR, O’Dowd BF, Lee SP. G-protein-coupled receptor oligomerizationand its potential for drug discovery. Nat Rev Drug Discov 2002;1:808–820.

7. Devi LA. Heterodimerization of G-protein-coupled receptors: pharmacology,signaling and trafficking. Trends Pharmacol Sci 2001;22:532–537.

8. Salim K, Fenton T, Bacha J, et al. Oligomerization of G-protein-coupled re-ceptors shown by selective co-immunoprecipitation. J Biol Chem2002;277:15,482–15,485.

9. Angers S, Salahpour A, Joly E, et al. Detection of beta 2-adrenergic receptordimerization in living cells using bioluminescence resonance energy transfer(BRET). Proc Natl Acad Sci USA 2000;97:3684–3689.

10. McVey M, Ramsay D, Kellett E, et al. Monitoring receptor oligomerizationusing time-resolved fluorescence resonance energy transfer and biolumines-cence resonance energy transfer. The human delta-opioid receptor displays

Page 16: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

282 Milligan, Carrillo, and Pascal

constitutive oligomerization at the cell surface, which is not regulated by re-ceptor occupancy. J Biol Chem 2001;276:14,092–14,099.

11. Mercier JF, Salahpour A, Angers S, Breit A, Bouvier M. Quantitative assessmentof beta 1- and beta 2-adrenergic receptor homo- and heterodimerization by biolu-minescence resonance energy transfer. J Biol Chem 2002;277:44,925–44,931.

12. Overton MC, Blumer KJ. The extracellular N-terminal domain and transmem-brane domains 1 and 2 mediate oligomerization of a yeast G protein-coupledreceptor. J Biol Chem 2002;277:41,463–41,472.

13. Ramsay D, Kellett E, McVey M, Rees S, Milligan G. Homo- and hetero-oligo-meric interactions between G-protein-coupled receptors in living cells moni-tored by two variants of bioluminescence resonance energy transfer (BRET):hetero-oligomers between receptor subtypes form more efficiently thanbetween less closely related sequences. Biochem J 2002;365:429–440.

14. Stanasila L, Perez J-B, Vogel H, Cotecchia S. Oligomerization of the α1a- andα1β-adrenergic receptor subtypes. J Biol Chem 2003;278:40,239–40,251.

15. Eidne KA, Kroeger KM, Hanyaloglu AC. Applications of novel resonanceenergy transfer techniques to study dynamic hormone receptor interactions inliving cells. Trends Endocrinol Metabol 2002;13:415–421.

16. Milligan G. Applications of bioluminescence and fluorescence resonanceenergy transfer to drug discovery at G protein-coupled receptors. Eur J PharmSci 2004;21:397–405.

17. Kroeger KM, Pfleger KDG, Eidne KA. Biophysical and biochemical methods tostudy G protein-coupled receptor oligomerization. In Devi LA, ed., The G Pro-tein-Coupled Receptors Handbook. Humana, Totowa, NJ: 2005; pp. 217–241.

18. Martin NP, Leavitt LM, Sommers CM, Dumont ME. Assembly of G protein-coupled receptors from fragments: identification of functional receptors withdiscontinuities in each of the loops connecting transmembrane segments. Bio-chemistry 1999;38:682–695.

19. Dean MK, Higgs C, Smith RE, et al. Dimerization of G-protein-coupled recep-tors. J Med Chem 2001;44:4595–4614.

20. Gouldson PR, Dean MK, Snell CR, Bywater RP, Gkoutos G, Reynolds CA.Lipid-facing correlated mutations and dimerization in G-protein coupled re-ceptors. Protein Eng 2001;14:759–767.

21. Maggio R, Scarselli M, Novi F, Millan MJ, Corsini GU. Potent activation ofdopamine D3/D2 heterodimers by the antiparkinsonian agents, S32504,pramipexole and ropinirole. J Neurochem 2003;87:631–641.

22. Maggio R, Vogel Z, Wess J. Coexpression studies with mutant muscarinic/adrenergic receptors provide evidence for intermolecular “cross-talk” betweenG-protein-linked receptors. Proc Natl Acad Sci USA 1993;90:3103–3107.

23. Monnot C, Bihoreau C, Conchon S, Curnow KM, Corvol P, Clauser E. Polarresidues in the transmembrane domains of the type 1 angiotensin II receptorare required for binding and coupling. Reconstitution of the binding site by co-expression of two deficient mutants. J Biol Chem 1996;271:1507–1513.

24. Mohler H, Fritschy JM. GABAB receptors make it to the top—as dimers.Trends Pharmacol Sci 1999;20:87–89.

Page 17: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

Functional Complementation 283

25. Marshall FH, Jones KA, Kaupmann K, Bettler B. GABAB receptors—the first7TM heterodimers. Trends Pharmacol Sci 1999;20:396–399.

26. Parmentier ML, Prezeau L, Bockaert J, Pin JP. A model for the functioning offamily 3 GPCRs. Trends Pharmacol Sci 2002;23:268–274.

27. Lee C, Ji I, Ryu K, Song Y, Conn PM, Ji TH. Two defective heterozygousluteinizing hormone receptors can rescue hormone action. J Biol Chem.2002;277:15,795–15,800.

28. Lee C, Ji IJ, Ji TH. Use of defined-function mutants to access receptor–recep-tor interactions. Methods 2002;27:318–323.

29. Ji I, Lee C, Song Y, Conn PM, Ji TH. Cis- and trans-activation of hormonereceptors: the LH receptor. Mol Endocrinol 2002;16:1299–1308.

30. Nakanishi-Matsui M, Zheng YW, Sulciner DJ, Weiss EJ, Ludeman MJ,Coughlin SR. PAR3 is a cofactor for PAR4 activation by thrombin. Nature2000;404:609–613.

31. O’Brien PJ, Prevost N, Molino M, et al. Thrombin responses in humanendothelial cells. Contributions from receptors other than PAR1 include thetransactivation of PAR2 by thrombin-cleaved PAR1. J Biol Chem2000;275:13,502–13,509.

32. Bertin B, Freissmuth M, Jockers R, Strosberg AD, Marullo S. Cellular signal-ing by an agonist-activated receptor/Gs alpha fusion protein. Proc Natl AcadSci USA 1994;91:8827–8831.

33. Milligan G. Insights into ligand pharmacology using receptor–G protein fusionproteins. Trends Pharmacol Sci 2000;21:24–28.

34. Milligan G. Construction and analysis of function of GPCR–G protein fusionproteins. Methods Enzymol 2002;343:260–273.

35. Moon HE, Cavalli A, Bahia DS, Hoffmann M, Massotte D, Milligan G. Thehuman δ opioid receptor activates Gi1α more efficiently than Go1α. JNeurochem 2001;76:1805–1813.

36. Ugur O, Onaran HO, Jones TL. Partial rescue of functional interactions of anonpalmitoylated mutant of the G-protein G alpha s by fusion to the beta-adr-energic receptor. Biochemistry 2003;42:2607–2615.

37. Loisel TP, Ansanay H, Adam L, et al. Activation of the beta(2)-adrenergicreceptor-Galpha(s) complex leads to rapid depalmitoylation and inhibition ofrepalmitoylation of both the receptor and Galpha(s). J Biol Chem1999;274:31,014–31,019.

38. Stevens PA, Pediani J, Carrillo JJ, Milligan G. Coordinated agonist regulationof receptor and G protein palmitoylation and functional rescue ofpalmitoylation-deficient mutants of the G protein G11alpha following fusion tothe alpha1b-adrenoreceptor: palmitoylation of G11alpha is not required for in-teraction with beta/gamma complex. J Biol Chem 2001;276:35,883–35,890.

39. Hosoi T, Koguchi Y, Sugikawa E, et al. Identification of a novel humaneicosanoid receptor coupled to G(i/o). J Biol Chem 2002;277:31,459–31,465.

40. Takeda S, Yamamoto A, Okada T, et al. Identification of surrogate ligands fororphan G protein-coupled receptors. Life Sci 2003;74:367–377.

41. Carrillo JJ, Pediani J, Milligan G. Dimers of class A G protein-coupled recep-tors function via agonist-mediated trans-activation of associated G proteins. JBiol Chem 2003;278:42,578–42,587.

Page 18: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

284 Milligan, Carrillo, and Pascal

42. Gether U. Uncovering molecular mechanisms involved in activation of G pro-tein-coupled receptors. Endocr Rev 2000;21:90–113.

43. Bahia DS, Wise A, Fanelli F, Lee M, Rees S, Milligan G. Hydrophobicity ofresidue351 of the G-protein Gi1α determines the extent of activation by the α2A-adrenoceptor. Biochemistry 1998;37:11,555–11,562.

44. Dupuis DS, Wurch T, Tardif S, Colpaert FC, Pauwels PJ. Modulation of 5-HT(1A) receptor activation by its interaction with wild-type and mutantg(alphai3) proteins. Neuropharmacology 2001;40:36–47.

45. Jackson VN, Bahia DS, Milligan G. Modulation of the relative intrinsic activ-ity of agonists at the α2A-adrenoceptor by mutation of residue351 of the G pro-tein Gi1α. Mol Pharmacol 1999;55:195–201.

46. Milligan G. The use of receptor-G protein fusion proteins for the study of ligandactivity. Receptors Channels 2002;8:309–317.

47. Liu S, Carrillo JJ, Pediani J, Milligan G. Effective information transfer fromthe α1b-adrenoceptor to G11α requires both β/γ interactions and an aromaticgroup 4 amino acid from the C-terminus of the G protein. J Biol Chem2002;277:25,707–25,714.

48. Sullivan KA, Miller RT, Masters SB, Beiderman B, Heideman W, Bourne HR.Identification of receptor contact site involved in receptor–G protein coupling.Nature 1987;330:758–760.

49. Waldhoer M, Wise A, Milligan G, Freissmuth M, Nanoff C. Kinetics of ter-nary complex formation with fusion proteins composed of the A1-adenosinereceptor and G protein α-subunits. J Biol Chem 1999;274:30,571–30,579.

50. Milligan G. Principles: extending the utility of [35S]GTPγS binding assays.Trends Pharmacol Sci 2003;24:87–90.

51. Burt AR, Sautel M, Wilson MA, Rees S, Wise A, Milligan G. Agonist-occupa-tion of an α2A-adrenoceptor–Gi1α fusion protein results in activation of bothreceptor -linked and endogenous G proteins: comparisons of their contribu-tions to GTPase activity and signal transduction and analysis of receptor–Gprotein activation stoichiometry. J Biol Chem 1998;273:10,367–10,375.

52. Molinari P, Ambrosio C, Riitano D, Sbraccia M, Gro MC, Costa T. Promiscu-ous coupling at receptor–Galpha fusion proteins. The receptor of one covalentcomplex interacts with the alpha-subunit of another. J Biol Chem2003;278:15,778–15,788.

53. Carrillo JJ, Stevens PA, Milligan G. Measurement of agonist-dependent and -independent signal initiation of alpha(1b)-adrenoceptor mutants by directanalysis of guanine nucleotide exchange on the G protein γalpha(11). JPharmacol Exp Ther 2002;302:1080–1088.

54. George SR, Fan T, Xie Z, et al. Oligomerization of μ- and δ-opioid receptors.J Biol Chem 2000;275:26,128–26,135.

55. Jordan BA, Devi LA. G-protein-coupled receptor heterodimerization modu-lates receptor function. Nature 1999;399:697–700.

56. Wenzel-Seifert K, Seifert R. Molecular analysis of beta(2)-adrenoceptor cou-pling to G(s)-, G(i)-, and G(q)-proteins. Mol Pharmacol 2000;58:954–966.

57. Yu R, Hinkle PM. Signal transduction and hormone-dependent internalizationof the thyrotropin-releasing hormone receptor in cells lacking Gq and G11. JBiol Chem 1999;274:15,745–15,750.

Page 19: [Contemporary Clinical Neuroscience] The G Protein-Coupled Receptors Handbook Volume 215 || Functional Complementation and the Analysis of GPCR Dimerization

Functional Complementation 285

58. Milligan G, Marshall F, Rees S. G16 as a universal G protein adapter: implica-tions for agonist screening strategies. Trends Pharmacol Sci 1996;17:235–237.

59. Pauwels PJ, Colpaert FC. Disparate ligand-mediated Ca(2+) responses by wild-type, mutant Ser(200)Ala and Ser(204)Ala alpha(2A)-adrenoceptor:G(alpha15) fusion proteins: evidence for multiple ligand-activation bindingsites. Br J Pharmacol 2000;130:1505–1512.

60. Pauwels PJ, Tardif S, Finana F, Wurch T, Colpaert FC. Ligand-receptor inter-actions as controlled by wild-type and mutant Thr(370)Lys alpha2B-adrenoceptor–Galpha15 fusion proteins. J Neurochem 2000;74:375–384.

61. Martin RS, Reynen PH, Calixto JJ, et al. Pharmacological comparison of arecombinant CB1 cannabinoid receptor with its G(alpha 16) fusion product. JBiomol Screen 2002;7:281–289.

62. Milligan G, Rees S. Chimaeric G alpha proteins: their potential use in drugdiscovery. Trends Pharmacol Sci 1999;20:118–124.

63. Coward P, Chan SD, Wada HG, Humphries GM, Conklin BR. Chimeric Gproteins allow a high-throughput signaling assay of Gi-coupled receptors. AnalBiochem 1999;270:242–248.

64. Liu AM, Ho MK, Wong CS, Chan JH, Pau AH, Wong YH. Galpha(16/z) chi-meras efficiently link a wide range of G protein-coupled receptors to calciummobilization. J Biomol Screen 2003;8:39–49.

65. Feng GJ, Cavalli A, Milligan G. Engineering a V(2) vasopressin receptor ago-nist- and regulator of G-protein-signaling-sensitive G protein. Anal Biochem2002;300:212–220.

66. Fong CW, Milligan G. Analysis of agonist function at fusion proteins betweenthe IP prostanoid receptor and cognate, unnatural and chimaeric G-proteins.Biochem J 1999;342:457–463.-