<<

REVIEWS

OLIGOMERIZATION OF G-PROTEIN- COUPLED TRANSMITTER RECEPTORS

Michel Bouvier Examples of G-protein-coupled receptors that can be biochemically detected in homo- or heteromeric complexes are emerging at an accelerated rate. Biophysical approaches have confirmed the existence of several such complexes in living cells and there is strong evidence to support the idea that dimerization is important in different aspects of biogenesis and function. While the existence of G-protein-coupled-receptor homodimers raises fundamental questions about the molecular mechanisms involved in transmitter recognition and , the formation of heterodimers raises fascinating combinatorial possibilities that could underlie an unexpected level of pharmacological diversity, and contribute to cross-talk regulation between transmission systems. Because G-protein-coupled receptors are major pharmacological targets, the existence of dimers could have important implications for the development and screening of new . Here, we review the evidence supporting the existence of G-protein-coupled-receptor dimerization and discuss its functional importance.

BIOGENIC AMINES As one of the largest gene families, G-protein-coupled a wealth of information on the biochemical events A series of molecules that can receptors (GPCRs) represent the most commonly used underlying cellular signalling by GPCRs. act as and signal-transduction system in the animal kingdom. In The proposed membrane topology of the receptors include noradrenaline and humans, it is estimated that ~1,000 distinct members consists of a hydrophobic core of seven transmembrane . direct responses to a wide variety of chemical transmit- α-helices that interact to form a three-dimensional bar- 1 ALLOSTERIC ters, including BIOGENIC AMINES, amino acids, , rel within the cytoplasmic membrane , an extracellular A term to describe proteins that lipids, nucleosides and large polypeptides. These trans- amino-terminal segment bearing amino-linked glyco- have two or more receptor sites, membrane receptors are key controllers of such diverse sylation sites and a cytoplasmic carboxy-terminal tail. one of which (the active site) binds the principal substrate, physiological processes as neurotransmission, cellular Their binding to specific ligands involves multiple inter- whereas the other(s) bind(s) metabolism, secretion, cellular differentiation and actions between functional groups on the ligands and effector molecules that can growth, as well as inflammatory and immune responses. specific amino acids within the extracellular domains influence its biological activity. The GPCRs therefore represent important targets for the and/or the hydrophobic transmembrane core of the development of new candidates with potential receptor2. Classically, the basic transduction unit com- applications in all clinical fields. Many therapeutic agents prises two elements in addition to the receptor: first, a Department of Biochemistry used at present act by either activating () or trimeric (αβγ) ; and second, an effector com- and Groupe de Recherche sur blocking (antagonists) GPCRs; widely used examples ponent. Binding of a transmitter promotes ALLOSTERIC le système Nerveux β Autonome, Faculté de are - receptor agonists for and interactions between the receptor and the trimeric G Médecine, Université de antagonists for and failure, hista- protein, leading to the release of GDP and the binding Montréal, α mine H1- and H2-receptor antagonists for allergies and of GTP to the -subunit. This destabilizes the trimeric P.O. Box 6128, Down-Town duodenal ulcers, agonists as analgesics, complex, allowing dissociation of the Gα•GTP and βγ Station, Montréal, Quebec, • H3C 3J7 Canada. receptor antagonists as and dimer . The ‘activated’ G protein, through its Gα GTP e-mail: Michel.Bouvier@ serotonin receptor agonists for . The results of chain, the βγ dimer or both, in turn interacts with and umontreal.ca studies pursued over the past two decades have provided modulates the effector component. Termination of the

274 | APRIL 2001 | VOLUME 2 www.nature.com/reviews/neuro © 2001 Macmillan Magazines Ltd REVIEWS

signal is achieved via hydrolysis of GTP to GDP by a by site–site interactions among receptors within dimeric 16,18–22 GTPase activity intrinsic to Gα. or oligomeric complexes . Atypical binding proper- Effector systems known to be modulated by GPCRs ties of dopamine antagonists such as [3H]-, using the scheme described above include enzymes such which detects only half of the maximal binding sites seen as , phospholipases C and D and cyclic by ligands of the benzamide family, have also been inter- GMP phosphodiesterase, as well as ion channels and preted as evidence for receptor dimers23. Biochemical antiporters such as the and channels studies, including photo-affinity labelling of the mus- and the Na+/H+ exchanger. Recently, additional effector carinic receptor24, radiation inactivation of α-adrenergic systems that were classically believed to be activated by and β-adrenergic25–27, gonadotropin28, - growth-factor receptors via kinase activation releasing hormone29, dopamine30 and A1 were also shown to be modulated by GPCRs3–8. In par- (REF.31) receptors, crosslinking of the receptor32, ticular, the ERK, p38 and JNK MAP (mitogen-activated and hydrodynamic properties of cardiac muscarinic protein) kinase signalling pathways were shown to be receptors33, also supported the idea that GPCRs might

activated by stimulation of G proteins of the Gq,Gi and form oligomeric structures. Gs families. Depending on the system considered, tyro- Despite these observations, the idea that GPCRs sine kinases9, phosphatidylinositol-3-OH kinase10, could function as dimers or oligomers never gained gen- Akt/ (REF.7),Src11 and Ras12 have all been eral acceptance and the prevailing model remained that implicated in this pathway. In addition, G-protein-inde- of a single receptor molecule interacting with a single G pendent signalling has been documented. For instance, protein. This dogma remained unchallenged until the β direct interaction of the 2- with the mid-1990s, when trans-complementation studies and Na+/H+-exchanger regulatory factor, NHERF, was new biochemical data reopened the question of GPCR shown to modulate the activity of the Na+/H+ exchanger dimerization. Most of the evidence taken to support the type-3 (REF.13). Other examples include the chemokine14 existence of GPCR dimers would also be consistent with and angiotensin II 15 receptors, where direct recruitment the existence of higher-order oligomers. As available and activation of the Janus kinases (JAK) was suggested. techniques do not allow these possibilities to be readily Generally, it was believed that distinct sets of intra- distinguished, the term dimer is often used, being the molecular interactions within the receptors would char- simplest form of oligomer that can explain the observa- acterize the active and inactive conformations upon tions. It is in this context that we use the word dimer binding of the ligands. However, recent data indicate throughout this review. that, in addition to the specific intramolecular interac- tions that could define the activation states of the recep- Complementation and immunoprecipitation tor, intermolecular interactions might also be important. One of the first studies that renewed interest in the pos- Receptor dimerization as well as interactions with acces- sibility that GPCRs could function as dimers was the 34 α sory proteins have been documented and proposed as elegant study by Maggio et al. , using chimeric 2- important determinants of GPCR activity. The following adrenergic/M3 muscarinic receptors composed of the sections review the biochemical and biophysical evi- first five transmembrane domains of one receptor and dence supporting the existence of GPCR homo- and the last two transmembrane domains of the other. heterodimers. The potential roles and implications of the When either chimera was expressed alone, no binding formation of such receptor dimers are discussed in the or signalling could be detected, but coexpression of the light of the most recent data, which indicate that dimer- two chimeras restored binding and signalling to both ization and oligomeric assemblies might represent the muscarinic and adrenergic ligands. Similarly, coexpres- rule rather than the exception for this important class of sion of two binding-defective angiotensin II receptor receptors. In several instances, the formation of oligo- point mutants rescued the binding affinity for the pep- meric complexes larger than dimers could explain the tide35, whereas coexpression of calcium receptors har- data as well as, or in some cases even better16 than, dimers. bouring inactivating in distinct domains was shown to partially rescue calcium-mediated signalling36. History of GPCR dimerization Such functional trans-complementations were inter- The concept that dimerization participates in the activa- preted as intermolecular interactions between inactive tion of transmembrane receptors is well accepted for receptors in a way that restored both -binding and many growth-factor and cytokine receptors, such as the signalling domains within a dimeric complex. epidermal growth factor (EGF), -derived growth Also consistent with the idea of GPCR dimer forma- factor (PDGF), interferon-γ and growth- recep- tion was the observation that several receptor mutants tors17. By contrast, until very recently, the conventional behave as DOMINANT-NEGATIVE mutants when expressed assumption for GPCRs was that monomeric receptors with their cognate wild-type receptor37–41. In these interacted allosterically with a single heterotrimeric G cases, dimerization between the wild-type and the inac- protein. However, as early as the mid-1970s, several indi- tive receptor was invoked to explain the blunted rect pharmacological observations led investigators to response observed. This was suggested to be potentially DOMINANT-NEGATIVE propose that GPCRs might also function as dimers. For clinically relevant for the calcium-sensing receptor, as A mutant protein that can form a heteromeric complex with the instance, complex binding curves for both agonists and some mutants with dominant-negative properties for normal molecule, knocking out antagonists to GPCRs were interpreted as evidence for this receptor are associated with inherited human the activity of the entire complex. negative or positive cooperativity that could be explained hypocalcaemic disorders37.

NATURE REVIEWS | NEUROSCIENCE VOLUME 2 | APRIL 2001 | 275 © 2001 Macmillan Magazines Ltd REVIEWS

Although these trans-complementation results indi- groups took advantage of biophysical assays based on cate that, at least in some conditions, GPCRs can func- light resonance energy transfer (BOX 1). tion as dimers, several investigators argued that this was Using fusion constructs between a GPCR and biolu- most probably the case only when mutant receptors were minescent (luciferase) and/or fluorescent (green fluores- considered. At about the same time, however, new bio- cent) proteins, bioluminescence resonance energy trans- chemical data were starting to support the idea that wild- fer (BRET) and fluorescence resonance energy transfer type GPCRs also existed as dimers. A co-immunoprecip- (FRET) were originally used to show homodimerization β 54 α itation approach using differentially epitope-tagged of the human 2-adrenergic receptor and the yeast - receptors provided direct biochemical evidence to sup- mating factor41 in living cells. More recently, the exis- β δ 55 port the existence of 2-adrenergic receptor homo- tence of -opioid and thyrotropin-releasing hormone 42 β 56 dimers .When Myc- and HA-tagged 2-adrenergic receptor oligomers was also confirmed in intact cells receptors were coexpressed, detection of HA immunore- using BRET. FRET between fluorescently conjugated activity in fractions immunoprecipitated with the anti- antibodies recognizing differentially epitope-tagged Myc antibody was taken as evidence of intermolecular receptors also allowed the detection of homodimers of interactions between the two differentially tagged recep- the SSTR5-somatostatin57 and δ-opioid receptors55 in tors. The selectivity of the interaction was illustrated by whole cells. Finally, derivatization of luteinizing hor- the lack of co-immunoprecipitation of the distantly mone (LH) with two different fluorophores (fluorescein related Myc-tagged M2 muscarinic receptor with isothiocyanate and tetramethylrhodamine isothio- β HA– 2-adrenergic receptor. Similar co-immunoprecipi- cyanate) representing a good FRET pair permitted the tation approaches have since been used to document the detection of LH receptor dimers in cells58.

dimerization of several GPCRs, including the GABAB Detection of BRET and FRET, even in the absence of (REFS 43–45), mGluR5 (REF.46), δ-opioid47, calcium48 and added , unambiguously shows that many GPCRs M3 muscarinic49 receptors. can form constitutive homodimers in intact living cells An interesting feature of many of these dimers is and that GPCR dimerization is not a biochemical arte- their relative resistance to sodium dodecyl sulphate fact. The presence of receptor dimers in the absence of (SDS) denaturation. Upon SDS–PAGE, they often migrate receptor activation by ligands raises the question of the as molecular species corresponding to twice the expect- role of dimerization in the activation process. It has ed monomeric receptor molecular mass. This intriguing been proposed56 that the constitutive presence of dimers resistance of the dimers to SDS is not unique to GPCRs could explain the constitutive activity that has been and is common to several proteins that form hydro- described for many GPCRs59. To our knowledge, how- phobic intermolecular interactions50. This might explain ever, no study has systematically assessed the effect of the recurrent observations, in western-blot analyses, of inverse agonists, which are known inhibitors of the con- immunoreactive receptor bands that could correspond stitutive ‘agonist-independent’ activity of the receptors, to oligomeric complexes. Covalent crosslinking before on constitutive dimerization. Alternatively, constitutive solubilization was also found to increase the proportion dimerization could be the reflection of a more funda- of dimers observed upon western blotting, and this mental role of GPCR dimerization in receptor ontology. β was used to document GPCR dimerization of the 2- adrenergic receptor (REF.42), the δ-opioid receptor51, the Dimerization in chaperoning and transport metabotropic glutamate mGluR5 receptor46 and the One of the most striking observations to indicate that calcium receptor48. GPCR dimerization might be important in receptor In most instances, detection of GPCRs dimers using folding and transport to the cell surface came from

co-immunoprecipitation, crosslinking and western-blot studies of the metabotropic GABAB receptor. Several approaches was achieved in heterologous systems over- groups simultaneously reported that coexpression of

expressing the receptor under investigation. However, two isoforms of the GABAB receptor, GABABR1 (a or b) 52 dimers of the A1 adenosine , dopamine D2 (REF.53) and (GBR1) and GABABR2 (GBR2), is a prerequisite for the metabotropic GABAB (REF. 45) receptors were also formation of a functional GABA receptor at the cell sur- observed in situ in tissue, indicating that the phe- face43–45,60,61 (FIG. 1). Detailed analysis of this phenome- nomenon is not simply an artefact due to anomalously non revealed that, when expressed alone in mammalian high levels of expression. cells, the GBR1 isoforms are retained intracellularly as immature glycoproteins62. By contrast, GBR2 is trans- Detecting dimers in living cells ported to the cell surface even when expressed alone but Although fairly convincing, co-immunoprecipitation cannot bind GABA or promote intracellular signalling43. and western-blot analyses require receptor solubiliza- When both receptors were coexpressed, the two pro- tion, raising the possibility that the observed dimers teins reached the cell surface as mature proteins and a could be solubilization artefacts. This is a putatively functional GABA receptor ensued. These data were SDS–PAGE important caveat when considering proteins such as interpreted as an indication that heterodimerization (Sodium dodecyl GPCRs that are composed of seven hydrophobic between GBR1 and GBR2 receptors is necessary for the sulphate–polyacrylamide gel transmembrane domains. Incomplete solubilization proper cell-surface expression of a functional GABA electrophoresis). A method for B (FIG. 1) resolving a protein into its could easily lead to aggregation that could be mistak- receptor . In agreement with this hypothesis, the subunits and determining their enly interpreted as dimerization. In an effort to assess existence of heterodimers could be demonstrated in separate molecular weights. the existence of GPCR dimers in living cells, several the same studies by co-immunoprecipitation of two

276 | APRIL 2001 | VOLUME 2 www.nature.com/reviews/neuro © 2001 Macmillan Magazines Ltd REVIEWS

Box 1 | Light resonance energy transfer approaches Light resonance energy transfer approaches are based on the non-radiative transfer of excitation energy between the electromagnetic dipoles of an energy donor and acceptor. In the case of fluorescence resonance energy transfer (FRET), both the donor and acceptor are fluorescent molecules, whereas for bioluminescence resonance energy transfer VIIII VII (BRET), the donor is bioluminescent and the acceptor is fluorescent. A prerequisite for these processes is that the emission spectrum of the donor and the excitation spectrum of the acceptor must overlap and that the donor and acceptor be in close proximity. BRET95 is a phenomenon occurring naturally in several marine animals such as the sea Rluc GFP pansy Renilla reniformis and the jellyfish Aequorea victoria.In R. reniformis,the luminescence resulting from the catalytic degradation of coelenterazine by luciferase Addition of 1µM (Rluc) is transferred to green fluorescent protein (GFP), which, in turn, emits fluorescence coelenterazine at its characteristic wavelength on dimerization of the two proteins. The strict dependence on the molecular proximity between donors and acceptors for energy transfer makes it a system of choice to monitor protein–protein interactions in living cells. As shown in the figure, one can take advantage of this phenomenon to study VII I I VII dimerization of G-protein-coupled receptors (GPCRs). Fusion proteins that link GFP and Bioluminescence Rluc to the carboxyl terminus of individual GPCRs are constructed and coexpressed. In Rluc GFP 400 500 600 the absence of dimerization, the addition of coelenterazine H should lead to a Wavelength (nm) characteristic broad bioluminescence signal with an emission peak at 470 nm, consistent Emission of blue light at 470 nm by Rluc with the spectral properties of Rluc. If homodimerization occurs, the energy transfer between Rluc and GFP (resulting from the proximity between the bioluminescent and the Within 100 Å, fluorescent fusion proteins) should lead to the appearance of an additional fluorescence BRET occurs signal with an emission peak at 530 nm that is characteristic of the GFP used (namely the red-shifted YFP)54. FRET can be used in the same way, using GPCRs fused to GFPs that have overlapping spectral properties (typically the CFP and the YFP). In this case, the initial energy is IIVII VII provided by direct excitation of the fluorescent donor with a light source41. Both the fluorescence emission of the acceptor and the quenching of the fluorescence of the donor 57 58 Bioluminescence can be used to quantitate the energy transfer. Antibodies or ligands that bind to the Rluc GFP 400 500 600 receptors can also be coupled to fluorophores that can be used as FRET pairs. Other Wavelength (nm) variations of the FRET technique that have been used to study GPCR dimerization Emission of green light at 520 nm by GFP include photo-bleaching FRET57 and time-resolved FRET55. In photo-bleaching FRET,the efficacy of energy transfer is indirectly determined by measuring the photo-bleaching time of the energy donor (upon sustained excitation) in the presence and absence of the energy acceptor. The energy transfer between the donor and the acceptor results in a slowing down of the photo-bleaching. Time-resolved FRET takes advantage of the long-lived fluorescence of fluorophores such as the lanthanide chelate Europium3+, which allow delayed FRET measurements while reducing the background resulting from the short-lived autofluorescence96.

receptors bearing different immunological tags. The The role of dimerization as an early event involved in observation that the transcripts of the two receptor sub- receptor maturation and transport is further supported types are coexpressed in many regions of the brain44,45, by the observation that the dominant-negative effect of and that endogenous GBR1 and GBR2 could be co- truncated forms of the V2 resulted immunoprecipitated from a cortex membrane prepa- from the interaction of the truncated mutants with full- ration derived from brain45, adds support to the length receptors, leading to the intracellular retention of physiological relevance of this phenomenon. the complex38. The early onset of dimer formation is also The idea that emerged from these studies is that GBR2 confirmed by the fact that mutants of the vasopressin serves as a MOLECULAR CHAPERONE that is essential for the receptors that cause nephrogenic diabetes because they proper folding and cell-surface transport of GBR1 and of are retained in the ER and never reach the cell surface64 a functional metabotropic GABA receptor. Whether, also form dimers (J. P. Morello, D. Bichet and M. B., once at the cell surface, the dimer is the functional recep- unpublished observations). The dominant-negative MOLECULAR CHAPERONE A protein that assists in the non- tor, or whether the maturation and transport of GBR1 to effects of a truncated form of the CCR5 chemokine covalent assembly of a protein the cell surface is sufficient, is discussed more extensively receptor (CCR5∆32) over its wild-type counterpart were complex but does not participate in the next section. In any case, the idea that GBR2 serves also attributed to its propensity to dimerize with the in its function. as a chaperone and escort protein for GBR1 has been wild-type receptor in the ER, thereby promoting intra- 39 COILED-COIL INTERACTION further supported by a recent study identifying an endo- cellular retention of the heterodimer . As the wild-type 65 A type of protein–protein plasmic reticulum (ER) retention signal within the receptor is a major co-receptor for HIV entry , it has interaction that involves carboxyl tail of GBR1 (REF.63).According to the model pro- been proposed that this dominant-negative effect of interlacing of two helical posed by the authors, GBR1–GBR2 dimerization involv- CCR5∆32 could explain the slow onset of AIDS in domains. ing a COILED-COIL INTERACTION of the carboxyl tail would patients who are heterozygous for this mutation66,67.The

VASOPRESSIN serve to hide the ER retention signal, thereby allowing ER intracellular retention of the wild-type dopamine D3 Antidiuretic hormone. export and plasma membrane targeting of the dimer. receptor upon coexpression of a splice variant form,

NATURE REVIEWS | NEUROSCIENCE VOLUME 2 | APRIL 2001 | 277 © 2001 Macmillan Magazines Ltd REVIEWS

GBR1GBR2 GBR1–GBR2 dimer

Cell-surface Cell-surface expression expression

Receptor Transport Transport stays in ER vesicle vesicle

Golgi Golgi Golgi

ER ER ER

Nucleus Nucleus Nucleus

Figure 1 | Role of homo- and heterodimerization in the transport of G-protein-coupled receptors. When expressed alone,

the GABABR1 (GBR1) receptor is retained as an immature protein in the (ER) of cells and never reaches the cell surface. By contrast, the GBR2 isoform is transported normally to the plasma membrane but is unable to bind GABA and thus to signal. When coexpressed, the two receptors are properly processed and transported to the cell surface as a stable dimer, where

they act as a functional metabotropic GABAB receptor.

D3nf, which is truncated before the sixth transmem- inhibits both dimerization and receptor-stimulated brane domain, has also been attributed to early dimer- adenylyl cyclase activity42. This result can easily be recon- ization in the ER68. The potential regulatory role of this ciled with a two-state receptor model, assuming that the naturally occurring variant in func- monomer and dimer represent the inactive and active tion remains to be investigated, but the overlap in the conformations respectively. Obviously, these results distribution of wild-type and D3nf in pyramidal alone do not prove that the dimer represents the active of rat brains69 certainly makes it worth exploring. form of the receptor or that dimerization is even required for activation. Indeed, it could be proposed that Role of dimerization in signal transduction the prevents signalling by disrupting intramole- The strongest evidence supporting a role for GPCR cular interactions within the receptor monomer, and dimerization in signal transduction once the receptor that loss of dimers is a consequence rather than the has reached the cell surface also comes from work car- cause of receptor inactivation. Supporting this possibili-

ried out on the GABAB receptor. As mentioned above, ty is the observation that a similar peptide derived from of the ER retention signal within the car- the dopamine D1 receptor inhibits dopamine signalling boxyl tail of GBR1 results in the transport of GBR1 to without affecting dimerization70. However, the demon- β the cell surface, but for this mutant receptor to respond stration that bivalent anti- 2-adrenergic receptor anti- functionally to GABA, it had to be coexpressed with bodies, but not their monovalent FAB FRAGMENTS, function GBR2 (REF. 63). This strongly suggests that the forma- as agonists and stimulate receptor activity71 lends fur- tion of a GBR1–GBR2 dimer is essential for signalling ther support to the idea that activation might result and that the simple cell-surface targeting of GBR1 is from dimerization. not sufficient. Although entirely consistent with the In a slightly different context, antibody-promoted idea that the dimer represents the signalling unit, one dimerization had previously suggested a role in cannot exclude the possibility that GBR2 might be gonadotropin-releasing activity. required for the proper folding of GBR1 in the ER, and Crosslinking of gonadotropin-releasing hormone pep- that the mutant GBR1 expressed alone might not reach tide antagonists with specific antibodies converted the the correct conformation. antagonists to agonists, suggesting that induction of Additional evidence indicating a role for dimeriza- receptor dimerization/aggregation is sufficient for acti- tion in GPCR function comes from the observation that vation72,73. Similarly, dimerization of the occupied a peptide derived from the proposed dimerization inter- LHRH (LH-releasing hormone) receptor was proposed FAB FRAGMENT β The antigen-binding portion of face of the 2-adrenergic receptor (see the section on as the mechanism leading to LH release from pituitary an antibody. architecture and three-dimensional organization) cells74. In a more recent study, Carrithers and Lerner75

278 | APRIL 2001 | VOLUME 2 www.nature.com/reviews/neuro © 2001 Macmillan Magazines Ltd REVIEWS

showed that covalent dimerization of a peptidic α-MSH Alternatively, these heterodimers could serve as receptors (melanocyte-stimulating hormone) for as yet undiscovered endogenous opioid peptides. If transformed it into an agonist, also consistent with the heterodimerization is a general feature of the GPCR class view that dimerization of this GPCR might be sufficient of receptors, this would offer a theoretical basis for a to activate G-protein signalling. More recently, additional pharmacological complexity that was unanticipated. indirect evidence supporting a role for dimerization in Also, it would provide a molecular mechanism that receptor activity came from FRET studies. FRET could explain some aspects of the cross-talk regulation between fluorescent LH derivatives was observed in cells observed between different signalling systems. expressing a wild-type receptor, but not a receptor that Although the generality of the phenomenon remains can bind LH but is unable to transmit the signal, indicat- to be shown, recent studies suggest that heterodimer- ing that these signalling-deficient receptors were unable ization is most probably not restricted to the GBR1/ to form dimers58. GBR2 and κ/δ-opioid receptor cases. Co-immuno- Interestingly, an anti-CCR5 receptor antibody that precipitation studies revealed stable association of the promotes dimerization was found to activate receptor- angiotensin I and B2 (REF. 78), dopamine D1 40 µ δ 80 β promoted calcium mobilization and cell migration , and adenosine A1 (REF. 79), - and -opioid and 2- but to inhibit its function as an HIV co-receptor and adrenergic and δ-opioid receptors, whereas a combina- thereby prevent viral entry into cells76. This indicates tion of co-immunoprecipitation and FRET approaches that oligomerization might have both positive and nega- convincingly showed heterodimerization between tive influences on distinct receptor functions. A role in somatostatin SSTR5 and SSTR1 (REF. 57), and SSTR5 signal termination was also invoked for the dimerization and dopamine D2 (REF. 81) receptors. of the δ-opioid receptor, as truncation of the receptor In some of these cases, the functional consequences of carboxyl tail was found to inhibit both dimerization and the heterodimerization observed in heterologous expres- agonist-induced endocytosis, a process involved in sion systems might help to rationalize cross-talk regula- receptor desensitization51. tory processes that have been postulated in vivo.For Although increasing evidence supports the idea that instance, the functional antagonism between the vaso- GPCR dimerization might be an important aspect of constrictor and vasodilator actions of angiotensin II and receptor function, how it does so and whether dynamic bradykinin could involve mutual regulatory influence at regulation of the oligomeric state is involved in normal the receptor level. This is an intriguing concept, consider- receptor activity remain highly debated issues. ing that these two signalling systems are already inter- connected by the angiotensin-converting enzyme that Expanding receptor diversity releases angiotensin II from its precursor and inactivates The existence of GBR1–GBR2 dimers explicitly showed bradykinin. It was observed that coexpression of the that non-identical receptors can form stable dimers and angiotensin I and bradykinin B2 receptors in HEK-293 raised the intriguing possibility that additional het- cells increases the efficacy and potency of angiotensin II, erodimers between distinct receptor subtypes could but reduces the ability of bradykinin to stimulate inosi- exist. This possibility was rapidly confirmed in 1999 tol-phosphate production78. Whether this truly reflects when Jordan and Devi showed that, when coexpressed receptor heterodimerization remains to be investigated. in the same cells, Myc-tagged κ-opioid and flag-tagged However, the fact that similar regulatory influences δ-opioid receptors could be co-immunoprecipitated between the two receptor systems could also be observed and therefore existed as a stable complex47. By contrast, in the cell line A10 that endogenously no heterodimerization between κ- and µ-opioid recep- expresses both angiotensin I and bradykinin B2 recep- tors was observed. Interestingly, the pharmacological tors, and that these two receptors are coexpressed in sev- properties of the κ–δ heterodimer were found to be dif- eral tissues including smooth muscle, glomerular ferent, both from those of each receptor expressed indi- mesangium and renal medulla, lends more support to a vidually and from what would be expected from a sim- possible physiological role for heterodimerization. ple mixture. For instance, the heterodimer showed no For adenosine A1/dopamine D1 heterodimerization, significant affinity for either κ- or δ-selective agonists or the coexpression of these two receptors in cortical neu- antagonists but showed high affinity for partially selec- rons and their widely reported antagonistic interactions tive ligands. However, selective ligands were found to in the might offer a physiological bind the heterodimer synergistically when added simul- rationale. Interestingly, Ginés et al.79 found that in taneously. At the functional level, the more-than-addi- fibroblasts, in which they could co-immunoprecipitate tive effect of selective κ- and δ-agonists on the stimula- the A1 and D1 receptors, pretreatment with both tion of MAP kinase activity was interpreted as a adenosine and dopamine agonists, but not with either synergistic action of the drugs on the heterodimer. individually, reduced the signalling efficacy of the D1 As some of the properties of the heterodimer were receptor upon subsequent stimulation. Whether this similar to those reported for the proposed κ2-opioid mechanism contributes to the known A1-induced inhi- receptor subtype77, Devi and colleagues47 proposed that bition of D1 receptor function in the brain remains to heterodimerization between opioid receptor isoforms be formally tested. might account for some of the receptor subtypes that Similarly, colocalization of the dopamine D2 recep- have been identified pharmacologically but for which no tor and somatostatin SSTR5 in cortical and striatal neu- gene or cDNA has been found, despite large-scale efforts. rons, and the rich clinical and behavioural literature

NATURE REVIEWS | NEUROSCIENCE VOLUME 2 | APRIL 2001 | 279 © 2001 Macmillan Magazines Ltd REVIEWS

indicating interactions between the somatostatinergic As already indicated, a pharmacological diversity and systems, supports the idea that the that exceeds what can be accounted for by molecularly heterodimerization observed by FRET in heterologous defined and cloned receptors has been proposed for expression system might have physiological relevance81. several receptors. These include: the atypical β-adren- β In this case, the functionality of the heterodimer was ergic pharmacology often referred to as the 4-receptor, demonstrated by showing that coexpression of the D2 CALCITONIN-gene-related peptide 1 (CGRP1) and CGRP2 ∆ receptor with a SSTR5 mutant ( 318 SSTR5), which subtypes, subtypes, ETB receptor sub- binds somatostatin but does not signal when expressed types, receptor subtypes, additional metabo- alone, imparts a somatostatin response to the cells. tropic subtypes, a Y3 µ δ κ This somatostatin-mediated inhibition of adenylyl receptor, 1,2-, 1,2- and 1,2,3-opioid receptor subtypes, cyclase activity was blocked by a , platelet-activating-factor (PAF) receptor subtypes, indicating that SSTR5 and D2 heterodimerize to con- additional prostanoid receptor subtypes, additional stitute a functional receptor. Also, synergistic binding neurokinin receptor subtypes and subtypes of the

of dopaminergic and somatostatinergic agonists was vasoactive intestinal peptide PAC1 receptor. Whether observed upon coexpression of wild-type SSTR5 and some of these unaccounted-for pharmacologically D2 receptors. As for the other examples reported above, identified subtypes could be explained by heterodimer- further work is now required to determine if such ization between already cloned receptors or between functional heterodimers can provide an explanation such receptors and accessory proteins remains an open for the reported reciprocal enhancement of each of question. Obviously, other explanations such as alter- these signalling systems upon administration of both native splicing, post-translational modifications or the somatostatin and dopamine in vivo. existence of additional genes coding for these receptors Despite increasing evidence that GPCR hetero- could also be responsible for this diversity. dimerization might be a general phenomenon, a word A definitive demonstration that heterodimerization of caution is provided by a recent study by McVey and does indeed contribute to the pharmacological diversity colleagues55. In their study, these authors clearly and regulation of GPCRs will require colocalization and β demonstrate that homodimerization of the 2-adren- simultaneous expression of potentially heterodimeriz- ergic and δ-opioid receptors can be detected by co- ing receptors to be documented in native tissues in each immunoprecipitation, BRET and time-resolved FRET case. Correlation between coexpression and the func- approaches. However, heterodimerization between tional consequences imparted by heterodimerization these two receptor types was detected only after co- will remain the ultimate criteria to judge the general immunoprecipitation, and no significant BRET or physiological importance of this phenomenon. FRET signals were observed upon their coexpression. Given that co-immunoprecipitation requires solubi- Dynamic regulation of GPCR dimerization lization of the receptor, and that BRET and FRET were Given the potential role that dimerization could play in carried out in living cells, the authors concluded that receptor transport, function and pharmacological this heterodimerization might represent a biochemical specificity, the question of the dynamic regulation of artefact resulting from nonspecific aggregation of these dimer formation becomes central in understanding hydrophobic proteins in the co-immunoprecipitation receptor activation and regulation processes. However, conditions. This conclusion contrasts sharply with that when the effects of agonist stimulation were considered of Jordan et al.82, who also observed co-immunoprecip- for various receptor homo- and heterodimers, different β δ itation of the 2-adrenergic and -opioid receptors but groups obtained somewhat different results. When co- β CALCITONIN additionally found that a selective 2- immunoprecipitation and/or western blots were used to A polypeptide hormone, promoted internalization of the δ-opioid receptor in assess the extent of dimerization, agonists were found to consisting of 32 amino-acid 42,76,81 β living cells, indicating that a functional heterodimer increase (for 2-adrenergic, SST5 and chemokine residues, that regulates was expressed at the surface of these cells. The apparent CCR5 homodimerization), decrease51,79 (for δ-opioid calcium and phosphate levels in the blood. contradiction between these two studies could be due homodimerization and dopamine D1/adenosine A1 to the inability of FRET and BRET to detect the hetero- heterodimerization) or to have no effect on (for κ-opi- HOMOTROPIC dimer as a result of distance constraints (BOX 1),or oid and M3 muscarinic homodimerization as well as Interaction between proteins of might indicate that the β -adrenergic receptor could angiotensin I/bradykinin B2 heterodimerization)78,83,84 the same class. 2 promote the internalization of the δ-opioid receptor the extent of dimerization.

ADRENOMEDULLIN by a process that is independent of heterodimeriza- Such variability could be attributed to the poor A hypotensive tion. In any case, it emphasizes that great care must be quantitative power of the co-immunoprecipitation and secreted by the medulla of the taken in the interpretation of data generated in this western-blot approaches. However, the more quantita- . emerging field. tive energy-transfer techniques also led to considerably δ 55 Stable association between distinct receptor subtypes variable results. In the cases of the -opioid and yeast A peptide consisting of 37 is not the only type of heterodimerization in which α-mating factor41 receptors, the BRET or FRET basal amino-acid residues that is GPCRs engage. Recent studies have indicated that inter- signals (indicative of constitutive dimerization) secreted with and might action with newly discovered accessory proteins as well remained unaffected by the addition of agonists, where- act to modulate its stimulatory β 54 effects on glucose metabolism in as with receptors of completely distinct classes can have as for the 2-adrenergic and thyrotropin-releasing hor- 56 muscle. Also known as islet important consequences for GPCR expression and mone receptors, agonists promoted an increase in amyloid peptide. function (BOX 2 and BOX 3). BRET signals above the basal levels. When considering

280 | APRIL 2001 | VOLUME 2 www.nature.com/reviews/neuro © 2001 Macmillan Magazines Ltd REVIEWS

Box 2 | Heterodimerization of CRLR and RAMP

HOMOTROPIC interactions between G-protein-coupled ab CGRP receptors (GPCRs) are not the only type of protein– protein interaction shown to influence their functional N N expression. Recently, a new class of membrane proteins that CRLR RAMP1 can interact with GPCRs and affect their activity profile has CRLR been identified. These new proteins were discovered while Cell surface C studying the expression of a complementary DNA that expression cAMP C encoded a putative GPCR, which did not lead to the CGRP expression of a functional receptor. Specifically, a cDNA Transport Terminal glycosylation named calcitonin-receptor-like receptor (CRLR), which vesicle showed 55% overall identity to the calcitonin-receptor gene, C was proposed to encode the receptor for the calcitonin-gene- C related peptide (CGRP)97. However, various attempts to Golgi N show that it was indeed the CGRP receptor failed because it CRLR + RAMP1 N was impossible to demonstrate any type of functional expression98. ER Several years later, using an expression-cloning approach ADM Core glycosylation in oocytes, McLatchie et al.99 isolated a new gene product that conferred CGRP signalling when expressed in oocytes. N C Nucleus Surprisingly, the new gene encoded a relatively small N protein containing only one putative transmembrane (TM) C domain and a short cytosolic tail. Obviously, the structure of this protein did not conform to the general seven-TM- CRLR + RAMP2/3 domain topology of GPCRs. Further studies showed that this single TM-domain protein had to be coexpressed with the CRLR gene product to confer CGRP binding and signalling99. The newly discovered protein was therefore considered as a co-receptor and was named receptor-activity-modifying protein (RAMP). Sequence data analysis and additional cloning experiments led to the identification of at least three isoforms of RAMP (1, 2 and 3). As the figure shows, although coexpression of CRLR with RAMP1 generated a CGRP receptor, coexpression with RAMP2 or RAMP3 produced a receptor with the pharmacological properties of the ADRENOMEDULLIN receptor, suggesting that the nature of the RAMP could determine the pharmacological properties of a receptor produced from a unique GPCR-encoding gene. Taken with the observation that CRLR expressed in the absence of RAMP is retained intracellularly and does not reach the cell surface, these results led to the suggestion that RAMPs act as chaperone/escort proteins that facilitate the maturation and targeting of the receptors, and also as direct activity modifiers through intermolecular interactions once the receptors have reached the cell surface. Coexpression of RAMP1 and RAMP3 with a cDNA encoding the (CTR) allows the expressed receptor to bind AMYLIN in addition to calcitonin, showing that the actions of RAMP are not limited to CRLR100. However, in this case, RAMP was not required for the transport of the CTR receptor to the cell surface and seemed to act solely as an activity modifier. Therefore, as is the case for dimerization among GPCRs, heterodimerization with RAMP is involved both in endoplasmic-reticulum (ER) export and transit to the plasma membrane, and in the modulation of receptor function at the cell surface. The relative importance of these roles seems to vary from case to case. Despite intense efforts to identify additional members of the RAMP family through databank analysis or sequence homology cloning, no additional genes or cDNAs have been found so far, indicating that this family of proteins has only a few members, or if additional members do exist, that they have important sequence differences. Supporting the latter possibility is the identification, in Caenorhabditis elegans, of a single transmembrane domain protein named ODR4 that shares no sequence homology with the RAMPs but is required for the cell-surface targeting and functional expression of the seven-TM , ODR10 (REF.101).

the heterodimerization of the D2 and SSTR5 receptors, K44A) that blocks receptor endocytosis via clathrin- Rocheville et al.81 found that FRET between D2 and coated vesicles54,56. Also, coexpression of two receptors VINBLASTIN An alkaloid that arrests mitosis SSTR5 fluorescently labelled antibodies could be that are known to be internalized via the clathrin-coated β in metaphase by binding to observed only in the presence of either somatostatin or pit pathway (the 2-adrenergic receptor–Rluc and the spindle microtubules. dopamine. Similarly, using fusion constructs between chemokine CCR5–YFP) did not result in any BRET, the gonadotropin-releasing hormone receptor and even in the presence of agonists for the two receptors54, CYTOCHALASIN 85 Any of a group of fungal green and red fluorescent proteins, Cornea et al. found indicating that clustering of receptors into clathrin-coated metabolites that interfere with a FRET signal that was entirely agonist-dependent. pits is not sufficient to promote BRET. the assembly and diassembly of Agonist-promoted energy transfer was distinguish- As the contribution of receptor clustering and inter- actin filaments. One of the able from macro-aggregation processes such as patch- nalization was excluded as a likely explanation for ago- consequences of treating cells ing, capping and internalization, as in the case of the nist-induced increase in energy transfer, several with these agents is that cleavage 56,81,85 of the cytoplasm after nuclear gonadotropin-releasing hormone receptor it was not authors concluded that agonists promote dimer division is prevented. inhibited by VINBLASTIN, CYTOCHALASIN or EGTA85.A con- formation. However, great care must be taken before tribution of the receptor internalization process to the jumping to that conclusion. Although an increase in the DYNAMIN agonist-mediated increase in BRET was also excluded number of dimers would lead to a larger BRET or FRET A protein involved in the β formation of microtubule for the 2-adrenergic and thyrotropin-releasing hor- signal, a different interpretation could account for the bundles and in membrane mone receptors as the BRET increase was not prevented changes in energy transfer. Both BRET and FRET effica- transport. by a dominant-negative mutant of DYNAMIN (dynamin cy vary with the sixth power of the distance between the

NATURE REVIEWS | NEUROSCIENCE VOLUME 2 | APRIL 2001 | 281 © 2001 Macmillan Magazines Ltd REVIEWS

with the addition of glutamate leading to a significant Box 3 | Heterodimerization of dopamine D5 and GABA receptors A conformational change of the pre-existing dimer86. G-protein-coupled receptors (GPCRs) and ion-channel receptors belong to two entirely Clearly, additional studies are required to determine different classes of protein and until recently, no data indicated that they could physically unambiguously if dimer assembly can be regulated by interact. However, as the two main classes of proteins involved in neurotransmission, it the activation state of the receptor. The role of post- would not be surprising if they did. In fact, the reported dopamine-receptor-mediated translational modification, as well as interaction with

modulation of GABAA-stimulated synaptic activity would support such an interaction. other proteins involved in signal transduction (for Addressing this possibility, Liu et al.102 provided convincing biochemical and functional example, G proteins, β-arrestins, receptor-activity-mod- data indicating that the dopamine D5 receptor physically interacts with the GABA- ifying protein (RAMP)), also remains to be determined. operated chloride channel GABAA receptor. Using hippocampal neurons and transfected These are crucial questions for understanding the fibroblasts, the authors demonstrated the direct binding of the carboxy-terminal portion mechanisms underlying receptor activation, and they of the D5 receptor to the second intracellular loop of the GABA receptor γ2 subunit. This A take on a special importance when considering the het- interaction was found to be dependent on the presence of agonists for both receptors, erodimers. Receptor selectivity would have to be consid- indicating that only the activated forms interacted. ered in an entirely new light if receptor monomers were In cells coexpressing the two receptors, the D5-mediated stimulation of adenylyl found to exchange between homodimers and different cyclase was inhibited by GABA, whereas the GABA-induced chloride current was heterodimers once they have reached the cell surface decreased by the activation of the dopamine receptor, indicative of reciprocal receptor and become activated. cross-talk. This functional cross-talk was shown to be dependent on the physical interaction between the two receptors, as it could be blocked by the expression of mini- γ Architecture of GPCR dimers genes expressing either the carboxyl tail of D5 or the 2 subunit of GABAA that both inhibited the formation of the complex. The physiological relevance of the interaction is The only available data that directly address the three- further supported by the observation that a D5 agonist decreased the amplitude of the dimensional structure of GPCR dimers is the recently solved structure of the extracellular ligand-binding GABAA-mediated miniature inhibitory postsynaptic current in cultured hippocampal neurons under conditions where the second-messenger-mediated pathways (PKA and (amino-terminal) region of the metabotropic glutamate PKC) were blocked. This regulatory effect was prevented by the addition of a peptide receptor mGluR1 (REF.86). The crystal structure shows derived from the carboxyl tail of the D5 receptor, supporting the idea that it resulted that a single disulphide bridge between Cys140 of each from a direct interaction between the channel and the GPCR. monomer connects the two PROTOMERS (FIG. 2).However,

As no interaction could be observed between GABAA and the D1 dopamine receptor, the authors of this study argue that this disulphide bond the ability of D5 to form a heterodimer with the ionotropic receptor might represent the cannot act as a scaffold because of its location within a first clearly defined functional role differentiating these two dopamine receptor disordered segment of the protein. The dimer interface subtypes. The fact that D5, but not D1, is preferentially targeted to the dendritic shafts was proposed to consist mainly of the helical packing and the cell soma/axon area of cortical and hippocampal neurons, where inhibitory between α-helices (helices B and C) in each monomer. 103 GABAergic neurons form major postsynaptic contacts , supports the physiological On the basis of co-immunoprecipitation and west- relevance of such a selective interaction. ern-blot studies, the formation of intermolecular disul- Overall, heterodimer formation was interpreted as a newly discovered mechanism by phide bonds within the extracellular amino-terminal which GPCRs and ligand-gated channels mutually regulate each other in the control of portion of the receptors has also been proposed to con- synaptic signalling efficacy. tribute to the formation of GPCR dimers. For mGluR1, mutation of Cys140 interfered with the detection of energy donor and acceptor, and they are also sensitive to dimers87,88. Similarly, disulphide bonding within the their dipole orientations (BOX 1). Therefore, conforma- large extracellular amino-terminal domains of mGluR5 tional changes promoted by the binding of an agonist to (REF.46) and the calcium-sensing48 receptor was found to a pre-existing dimer could lead to significant changes in be important for covalent dimerization. Finally, cys- energy-transfer efficacy. The extent to which the confor- teines located in extracellular loops two and three of the mational change imposed by a specific receptor agonist M3 muscarinic receptor were also invoked in the dimer- is communicated to the receptor domain where the flu- ization of this receptor83. For mGluR5 (REFS 87,89) and orophores have been attached could determine whether the calcium-sensing receptor90 , however, the disulphide a change in signal is observed. In some cases, the signal bond was found not to be the only point of contact, and could go from undetected to very significant, whereas in non-covalent interactions involving both the extracellu- others the high constitutive signal (reflecting an already lar and transmembrane domains were proposed. The optimal transfer) would not be affected by the binding idea that hydrophobic interactions involving trans- of the agonist. membrane domains could be involved in GPCR dimer- β Such an explanation is entirely compatible with the ization was first proposed for the 2-adrenergic chaperoning role evoked above for the dimerization of receptor42. The idea came from an elegant series of several GPCRs, as their dimerization occurs in the ER, experiments by Engleman and colleagues91,92 that com- long before the agonists can influence dimer formation. bined the use of synthetic peptides, site-directed muta- The recent crystallographic resolution of the three- genesis and biophysical techniques. They showed that dimensional structure of the extracellular amino-termi- specific residues located in the transmembrane domain nal domain of the metabotropic glutamate receptor is of GLYCOPHORIN A are essential for the formation of the also consistent with such a model of constitutive dimer- hydrophobic interactions that stabilize the protein

PROTOMERS ization. Indeed, this part of the receptor, constituting its dimers. On the basis of an analogy with this proposed Identical subunits in an ligand-recognition and binding domain, was found to motif, glycine and leucine residues located within the oligomeric protein complex. β exist as a dimer whether glutamate was present or not, sixth transmembrane segment of the 2-adrenergic

282 | APRIL 2001 | VOLUME 2 www.nature.com/reviews/neuro © 2001 Macmillan Magazines Ltd REVIEWS

Disulphide bond formation Although it lacks a consensus sequence for a coiled-coil δ SS domain, the carboxyl tail of the -opioid receptor was 51 S S also invoked as a determinant of dimerization . Clearly, results available so far are too fragmentary to VIIII VII establish a general molecular mechanism for GPCR dimerization. The different dimerization modes pro- posed in the different studies (FIG. 2), rather than reflect- ing different strategies used by receptors of different classes, most probably indicate that multiple sites of Calcium and glutamate receptors interaction are involved in the assembly and stabilization of receptor dimers. Coiled-coil interaction Computational studies led Gouldson et al.93 to pro- pose two alternative three-dimensional models that could describe GPCR dimers (FIG. 3). Molecular- I VII VII I dynamics simulations, correlated-mutation analysis and evolutionary-trace analysis all support the involve- ment of transmembrane helices five and six in the dimerization interface, but cannot distinguish between GABA receptor B the domain-swapped and contact-dimer models. Interestingly, the two models predict that the third intracellular loop originating from each monomer Transmembrane interaction would be parallel within the dimer. This could have important functional implications given the role pro- posed for this receptor domain in G-protein engage- ment and stimulation. Although some of the biochemi- IIVII VII cal and functional data might be more easily rationalized by one model than another, the validation of any of these models awaits additional studies. The most direct test will certainly come from the resolution β of the structure of a GPCR dimer crystal. Unfortunately, 2-adrenergic, dopamine, muscarinic and angiotensin receptors the recent crystallization of did not allow the issue of dimerization to be directly addressed. The crys- Figure 2 | Molecular determinants of G-protein-coupled- tal revealed an antiparallel dimer with respect to the receptor dimerization. Distinct intermolecular interactions plane of the membrane that certainly resulted from the were found to be involved for various G-protein-coupled receptors. Covalent disulphide bonds were found to be crystallization process. Most probably, solubilization led important for the dimerization of the calcium-sensing and to the disassembly of physiologically relevant dimers metabotropic glutamate receptors. A coiled-coil interaction and the antiparallel dimer formed to minimize packing involving the carboxyl tail of the GBR1 and GBR2 receptors is energy during crystallization. Nevertheless, the resolu- involved in the formation of their heterodimer. Finally, for tion of this structure still provided indirect information monoamine receptors such as the β -adrenergic and 2 that could be analysed in the context of the dimer dopamine receptors, interactions between transmembrane helices were proposed to be involved. hypothesis. Indeed, the size of the intracellular surface exposed to the cytosol is too small to account for the simultaneous interaction of the receptor with both α- receptor were suggested as part of the dimerization and βγ-subunits of the G protein. As it is generally interface for this receptor42. Similar results were obtained accepted that such concomitant interaction is required for the dopamine D2 receptor53. However, the fact that for function, it could suggest that a receptor dimer is peptides derived from several transmembrane domains needed for a complete and productive interaction with of D2 could also block dimerization suggests that trans- a single . Of course, this membrane α-helices might have a more general role in intriguing speculation will require a more formal receptor folding and oligomerization and might not be demonstration. limited to strict consensus sequences. As mentioned previously, yeast two-hybrid screens43 Concluding remarks pointed to coiled-coil domains within the carboxyl tails After 20 years as an ‘underground’ concept, in the past of GBR1 and GBR2 as important molecular determi- five years GPCR dimers have gone from being a contro-

nants of GABAB receptor heterodimerization (FIG. 2). versial idea to a widely accepted hypothesis. Although its However, mutagenesis studies revealed that, although it universality still needs to be established, the increasing was important for receptor function and to mask the ER number of reports that have used various approaches to GLYCOPHORIN A retention signal, the coiled-coil motif was not necessary describe GPCR dimers has led to the general acceptance A carbohydrate-rich sialoglycoprotein that is for dimer formation, as deletion of the carboxyl tail did that at least some of them exist as oligomeric assemblies 63 abundant in erythrocyte not abrogate it . It is therefore likely that the carboxyl involving more than one receptor. The use of biophysi- membranes. tail participates in but is not essential for dimerization. cal approaches that do not require cell disruption, such

NATURE REVIEWS | NEUROSCIENCE VOLUME 2 | APRIL 2001 | 283 © 2001 Macmillan Magazines Ltd REVIEWS abtems rather than systems expressing unique receptor Formation of 1–7 dimer 2 1 2 1 from inactive muscarinic M3 6 5 6 5 isoforms would have to be considered. Also, assays α 4 3 7 4 3 7 and adrenergic 2 7 3 4 7 3 4 aimed at monitoring the dimerization state of receptors chimeric monomers 5 6 12 5 6 12 could be imagined. The pharmacological reality of such considera- I3 hinge loop tions is supported by the recent report that the anti- convulsant, antihyperalgesic and ANXIOLYTIC AGENT 2 1 gabapentin is an agonist for the GBR1a/GBR2 hetero- Muscarinic M3 domain 6 Contact-dimer 7 5 4 3 formation dimer but is esssentially inactive on the GBR1b/GBR2 α Adrenergic 2 domain 5 3 4 complex94. In CA1 pyramidal neurons of rat hippo- 6 7 12 campal slices, gabapentin was found to activate post- + Rearrangement synaptic inward rectifier K currents, probably via the of 1–7 dimer GBR1a/GBR2 heterodimer, but it did not depress 2 1 2 1 2 1 GABA signalling presynaptically, indicating a selec- 4 3 A 4 3 7 4 3 7 7 tivity of action that relies in part on the expression of 5 6 5 6 5 6 a specific GABAB receptor heterodimer. Such selective 6 5 6 5 6 5 agonism might be part of its therapeutic advantage as 7 7 4 7 3 4 3 4 3 an anticonvulsant. Obviously, the possibilities become 12 12 12 staggering when the proposed interactions between GPCRs and other classes of receptor (for example, Active 5–6 domain- 5–6 contact dimer swapped dimer ionotropic), or accessory protein (for example, RAMP), are taken into account. Figure 3 | Alternative three-dimensional models showing dimers of G-protein-coupled Oligomeric assembly of proteins, allowing expanded receptors. Two models have been proposed for the general three-dimensional organization of diversity with a limited number of modular elements, is G-protein-coupled-receptor dimers. a | First is the domain-swapping model in which each the rule rather than the exception in biology. When con- functional unit within the dimer is composed of the first five transmembrane domains of one sidering the nervous system, the existence of homo- and polypeptide chain and the last two of the other. Such a model is useful to rationalize the functional heterodimers of GPCRs offers an complementation observed when mutant or chimeric receptors are coexpressed. b | Second is the contact model in which each polypeptide forms a receptor unit that touches the other through attractive hypothesis that could underlie the high degree interactions involving transmembrane domains five and six. of diversity and plasticity that is characteristic of such a highly organized and complex system. Despite the excitement raised by the discovery of as light energy transfer, has certainly helped to reassure GPCR homo- and heterodimers, many questions still us that they are not biochemical artefacts. Also, the need to be answered before we can fully appreciate absolute requirement of heterodimerization for the their real contribution to normal physiology. Among

transport of a functional metabotropic GABAB receptor them, the general importance of heterodimerization to the cell surface has strongly reinforced the idea that in explaining pharmacological diversity and cross-talk dimerization is functionally important. Whether the regulation processes is likely to attract considerable main role of dimerization will be in the chaperoning attention. Although technically more demanding, it and cell transport of receptors, or in the control of sig- will also be necessary to assess the occurrence and nalling specificity and efficacy, remains an open ques- roles of heterodimers in native tissues to validate each tion for most receptors. However, a growing body of of the observations made in heterologous expression evidence arguing in favour of important roles for systems. If heterodimerization is found to be a general dimerization in all GPCR classes largely justifies further process in the nervous system, understanding the investigation. This is particularly true when considering mechanisms that direct the selectivity of interactions the veritable revolution that it might trigger in the between receptors will become a central question in development and screening of drugs that target GPCRs for their therapeutic actions. For instance, defining the Links oligomerization interface and its molecular dynamics DATABASE LINKS EGF | PDGF | interferon-γ | muscarinic could offer new pharmacological targets. Compounds receptors | α-adrenegic receptors | β-adrenegic receptors | that modulate receptor dimerization without interfer- gonadotropin-releasing hormone receptor | dopamine ing with the hormone-binding pocket could represent receptors | | | a new class of non-competitive drugs with distinct α 2-adrenergic receptor | M3 muscarinic receptor | selectivity and activity profiles. angiotensin II receptor | β -adrenergic receptor | One of the most intriguing promises is offered by the 2 M2 muscarinic receptor | GABAB receptor | mGluR5 | possibility that heterodimerization might be a common δ-opioid receptor | SSTR5 | CCR5 | κ-opioid receptor | phenomenon among GPCRs. In addition to exponen- µ-opioid receptor | SSTR1 | angiotensin I receptor | tially increasing the number of pharmacologically dis- bradykinin B2 receptor | RAMP | mGluR1 tinguishable targets, the definitive demonstration of FURTHER INFORMATION Domain swapping in G-protein- their physiological relevance will markedly change the ANXIOLYTIC AGENT coupled-receptor dimers A drug used to reduce anxiety, way that high-throughput screens are conceived for this ENCYCLOPEDIA OF LIFE SCIENCES G-protein-coupled such as benzodiazepines. class of receptor. For one, combinatorial expression sys- receptors

284 | APRIL 2001 | VOLUME 2 www.nature.com/reviews/neuro © 2001 Macmillan Magazines Ltd REVIEWS

neuropharmacology. One extensively studied aspect receptor subtypes, is already a highly debated issue. will certainly be the potential dynamic regulation of The prospect of such dynamic regulation has many dimer formation during the activation/inactivation potential implications for our understanding of the cycle of the receptors. The question of whether dimers neurotransmission process, and this will undoubtedly are constitutive and stable throughout the life of the lead many researchers to investigate the mechanisms receptor, or can undergo rounds of monomeriza- underlying oligomeric assembly of the most important tion/dimerization with potential exchanges between class of neurotransmitter receptors.

1. Suryanarayana, S., Von Zastrow, M. & Kobilka, B. K. muscarinic receptors expressed in Chinese hamster ovary 40. Rodriguez-Frade, J. M. et al. The chemokine monocyte Identification of intramolecular interactions in adrenergic cells. J. Biol. Chem. 269, 26127–26135 (1994). chemoattractant protein-1 induces functional responses receptors. J. Biol. Chem. 267, 21991–21994 (1992). 23. Seeman, P. et al. The cloned dopamine D2 receptor reveals through dimerization of its receptor CCR2. Proc. Natl Acad. 2. Bockaert, J. & Pin, J. P. Molecular tinkering of G protein- different densities for dopamine receptor antagonist ligands. Sci. USA 96, 3628–3633 (1999). coupled receptors: an evolutionary success. EMBO J. 18, Implications for human brain positron emission tomography. 41. Overton, M. C. & Blumer, K. J. G Protein coupled 1723–1729 (1999). Eur. J. Pharmacol. 227, 139–146 (1992). receptors function as oligomers in vivo. Curr. Biol. 10, 3. Crespo, P., Cachero, T. G., Xu, N. & Gutkind, J. S. Dual 24. Avissar, S., Amitai, G. & Sokolovsky, M. Oligomeric 341–344 (2000). effect of β-adrenergic receptors on mitogen-activated structure of muscarinic receptors is shown by photoaffinity Using non-disruptive bioluminescence and protein kinase. J. Biol. Chem. 270, 25259–25265 (1995). labeling: subunit assembly may explain high- and low-affinity fluorescence resonance energy transfer approaches, 4. Bogoyevitch, M. A. et al. Adrenergic receptor stimulation of agonist states. Proc. Natl Acad. Sci. USA 80, 156–159 this paper and references 54 and 57 all demonstrated the mitogen-activated protein kinase cascade and cardiac (1983). simultaneously and independently that G-protein- hypertrophy. Biochem. J. 314, 115–121 (1996). 25. Fraser, C. M. & Venter, J. C. The size of the mammalian lung coupled receptors form dimers in living cells. 5. Yamamoto, J., Nagao, M., Kaziro, Y. & Itoh, H. Activation of β-2-adrenergic receptor as determined by target size 42. Hebert, T. E. et al. A peptide derived from a β2-adrenergic p38 mitogen-activated protein kinase by signaling through analysis and immunoaffinity chromatography. Biochem. receptor transmembrane domain inhibits both receptor G protein-coupled receptors. Involvement of Gi and Gq/11 Biophys. Res. Commun. 109, 21–29 (1982). dimerization and activation. J. Biol. Chem. 271, subunits. J. Biol. Chem. 272, 27771–27777 (1997). 26. Venter, J. C., Schaber, J. S., U’Prichard, D. C. & Fraser, 16384–16392 (1996). 6. Williams, N. G., Zhong, H. & Minneman, K. P. Differential C. M. Molecular size of the human platelet α2-adrenergic Using differential epitope tagging and coupling of α1-, α2-, and β-adrenergic receptors to receptor as determined by radiation inactivation. Biochem. co-immunoprecipitation, this study provided direct mitogen-activated protein kinase pathways and Biophys. Res. Commun. 116, 1070–1075 (1983). biochemical evidence that wild-type differentiation in transfected PC12 cells. J. Biol. Chem. 273, 27. Venter, J. C., Horne, P., Eddy, B., Greguski, R. & Fraser, G-protein-coupled receptors exist and might function 24624–24632 (1998). C. M. Alpha 1-adrenergic receptor structure. Mol. as dimers. 7. Gerhardt, C. C., Gros, J., Strosberg, A. D. & Issad, T. Pharmacol. 26, 196–205 (1984). 43. White, J. H. et al. Heterodimerization is required for the Stimulation of the extracellular signal-regulated kinase 1/2 28. Crine, P., Aubry, M. & Potier, M. Incorporation of radiolabeled formation of a functional GABA(B) receptor. Nature 396, pathway by human β-3 adrenergic receptor: new amino acids into protein subunits of the rat leydig cell 679–682 (1998). pharmacological profile and mechanism of activation. : application to the study of receptor 44. Jones, K. A. et al. GABA(B) receptors function as a Mol. Pharmacol. 55, 255–262 (1999). structure and turnover. Ann. NY Acad. Sci. 438, 224–236 heteromeric assembly of the subunits GABA(B)R1 and 8. Soeder, K. J. et al. The β-3 adrenergic receptor activates (1984). GABA(B)R2. Nature 396, 674–679 (1998). mitogen-activated protein kinase in adipocytes through a 29. Conn, P. M. & Venter, J. C. Radiation inactivation (target size 45. Kaupmann, K. et al. GABA(B)-receptor subtypes assemble

Gi-dependent mechanism. J. Biol. Chem. 274, analysis) of the gonadotropin-releasing hormone receptor: into functional heteromeric complexes. Nature 396, 12017–12022 (1999). evidence for a high molecular weight complex. 683–687 (1998). 9. van Biesen, T. et al. Receptor-tyrosine-kinase- and Gβγ- Endocrinology 116, 1324–1326 (1985). References 43–45 showed simultaneously and mediated MAP kinase activation by a common signalling 30. Bouvier, C. et al. Solubilization and characterization of D2- independently that not only could heterodimerization

pathway. Nature 376, 781–784 (1995). dopamine receptors in an estrone-induced, - between GABABR1 and GABABR2 receptors occur,

10. Zou, Y. et al. Both Gs and Gi proteins are critically involved in secreting rat pituitary adenoma. J. Neurochem. 47, but that it is essential to the formation of a functional isoproterenol-induced cardiomyocyte hypertrophy. J. Biol. 1653–1660 (1986). receptor expressed at the cell surface. Chem. 274, 9760–9770 (1999). 31. Frame, L. T., Yeung, S. M., Venter, J. C. & Cooper, D. M. 46. Romano, C., Yang, W. L. & O’Malley, K. L. Metabotropic 11. Luttrell, L. M., Della, R. G., van Biesen, T., Luttrell, D. K. & Target size of the adenosine R1 receptor. Biochem. J. 235, glutamate receptor 5 is a disulfide-linked dimer. J. Biol. Lefkowitz, R. J. Gβγ subunits mediate Src-dependent 621–624 (1986). Chem. 271, 28612–28616 (1996). phosphorylation of the epidermal growth factor receptor. A 32. Herberg, J. T., Codina, J., Rich, K. A., Rojas, F. J. & Iyengar, Biochemical demonstration that dimerization of the scaffold for G protein-coupled receptor-mediated Ras R. The hepatic glucagon receptor. Solubilization, metabotropic glutamate receptor involves the activation. J. Biol. Chem. 272, 4637–4644 (1997). characterization, and development of an affinity adsorption formation of disulphide bridges between their large 12. Luttrell, L. et al. Beta-arrestin-dependent formation of β-2- assay for the soluble receptor. J. Biol. Chem. 259, extracellular amino-terminal domains. adrenergic receptor-Src protein kinase complexes. Science 9285–9294 (1984). 47. Jordan, B. A. & Devi, L. A. G-protein-coupled receptor 283, 655–661 (1999). 33. Peterson, G. L., Rosenbaum, L. C., Broderick, D. J. & heterodimerization modulates receptor function. Nature 13. Hall, R. A. et al. The β-2 adrenergic receptor interacts with Schimerlick, M. I. Physical properties of the purified cardiac 399, 697–700 (1999). the Na/H-exchanger regulatory factor to control NA/H muscarinic . Biochemistry 25, A combination of co-immunoprecipitation and exchange. Nature 329, 626–630 (1998). 3189–3202 (1986). pharmacological analysis of coexpressed δ- and 14. Mellado, M. et al. The chemokine monocyte chemotactic 34. Maggio, R., Vogel, Z. & Wess, J. Co-expression studies with κ-opioid receptors suggested for the first time that protein 1 triggers Janus kinase 2 activation and tyrosine mutant muscarinic/adrenergic receptors provide evidence dimerization between distinct receptor subtypes phosphorylation of the CCR2B receptor. J. Immunol. 161, for intermolecular ‘cross-talk’ between G-protein-linked could lead to the formation of a receptor with unique 805–813 (1998). receptors. Proc. Natl Acad. Sci. USA 90, 3103–3107 pharmacological properties. 15. Ali, M. S. et al. Dependence on the motif YIPP for the (1993). 48. Bai, M., Trivedi, S. & Brown, E. M. Dimerization of the α physical association of Jak2 kinase with the intracellular Using 2-adrenergic/M3 muscarinic chimeric extracellular calcium-sensing receptor (CaR) on the cell carboxyl tail of the angiotensin II AT1 receptor. J. Biol. Chem. proteins, this study documented the occurrence of surface of CaR-transfected HEK293 cells. J. Biol. Chem. 272, 23382–23388 (1997). intermolecular functional complementation indicating 273, 23605–23610 (1998). 16. Wreggett, K. A. & Wells, J. W. Cooperativity manifest in the that G-protein-coupled receptors can function as 49. Zeng, F. Y. & Wess, J. Identification and molecular binding properties of purified cardiac muscarinic receptors. dimeric entities. characterization of m3 muscarinic receptor dimers. J. Biol. J. Biol. Chem. 270, 22488–22499 (1995). 35. Monnot, C. et al. Polar residues in the transmembrane Chem. 274, 19487–19497 (1999). 17. Heldin, C. H. Dimerization of cell surface receptors in signal domain of the type 1 angiotensin II receptor are required for 50. Furthmayr, H. & Marchesi, V. T. Subunit structure of human transduction. Cell 80, 213–223 (1995). binding and coupling. Reconstitution of the binding site by erythrocyte glycophorin A. Biochemistry 15, 1137–1144 18. Limbird, L. E., Meyts, P. D. & Lefkowitz, R. J. Beta- co-expression of two deficient mutants. J. Biol. Chem. 271, (1976). adrenergic receptors: evidence for negative cooperativity. 1507–1513 (1996). 51. Cvejic, S. & Devi, L. A. Dimerization of the δ-opioid receptor: Biochem. Biophys. Res. Commun. 64, 1160–1168 (1975). 36. Bai, M., Trivedi, S., Kifor, O., Quinn, S. J. & Brown, E. M. implication for a role in receptor internalization. J. Biol. 19. Potter, L. T. et al. Evidence for paired M2 muscarinic Intermolecular interactions between dimeric calcium- Chem. 272, 26959–26964 (1997). receptors. Mol. Pharmacol. 39, 211–221 (1991). sensing receptor monomers are important for its normal 52. Ciruela, F. et al. Immunological identification of A1 20. Limbird, L. E. & Lefkowitz, R. J. Negative cooperativity function. Proc. Natl Acad. Sci. USA 96, 2834–2839 (1999). adenosine receptors in brain cortex. J. Neurosci. Res. 42, among β-adrenergic receptors in frog erythrocyte 37. Bai, M. et al. Expression and characterization of inactivating 818–828 (1995). membranes. J. Biol. Chem. 251, 5007–5014 (1976). and activating mutations in the human Ca2+-sensing 53. Ng, G. Y. et al. Dopamine D2 receptor dimers and receptor- 21. Mattera, R., Pitts, B. J., Entman, M. L. & Birnbaumer, L. receptor. J. Biol. Chem. 271, 19537–19545 (1996). blocking peptides. Biochem. Biophys. Res. Commun. 227, Guanine nucleotide regulation of a mammalian myocardial 38. Zhu, X. & Wess, J. Truncated V2 vasopressin receptors as 200–204 (1996). muscarinic receptor system. Evidence for homo- and negative regulators of wild-type V2 receptor function. 54. Angers, S. et al. Detection of β2-adrenergic receptor heterotropic cooperativity in ligand binding analysed by Biochemistry 37, 15773–15784 (1998). dimerization in living cells using bioluminescence resonance computer-assisted curve fitting. J. Biol. Chem. 260, 39. Benkirane, M., Jin, D. Y., Chun, R. F., Koup, R. A. & Jeang, energy transfer (BRET). Proc. Natl Acad. Sci. USA 97, 7410–7421 (1985). K. T. Mechanism of transdominant inhibition of CCR5- 3684–3689 (2000). 22. Hirschberg, B. T. & Schimerlik, M. I. A kinetic model for mediated HIV-1 infection by ccr5delta32. J. Biol. Chem. 55. McVey, M. et al. Monitoring receptor oligomerization using oxotremorine M binding to recombinant porcine m2 272, 30603–30606 (1997). time-resolved fluorescence resonance energy transfer and

NATURE REVIEWS | NEUROSCIENCE VOLUME 2 | APRIL 2001 | 285 © 2001 Macmillan Magazines Ltd REVIEWS

bioluminescence resonance energy transfer: The human δ- receptors. Biochem. Biophys. Res. Commun. 133, 91. Lemmon, M. A. et al. Glycophorin A dimerization is driven by opioid receptor displays constitutive oligomerization at the 449–456 (1985). specific interactions between transmembrane alpha-helices. cell surface which is not regulated by receptor occupancy. 74. Gregory, H., Taylor, C. L. & Hopkins, C. R. Luteinizing J. Biol. Chem. 267, 7683–7689 (1992). J. Biol. Chem. (in the press). hormone release from dissociated pituitary cells by 92. Lemmon, M. A. & Engelman, D. M. Specificity and 56. Kroeger, K. M., Hanyaloglu, A. C., Seeber, R. M., Miles, L. E. dimerization of occupied LHRH receptors. Nature 300, promiscuity in membrane helix interactions. FEBS Lett. 346, C. & Eidne, K. A. Constitutive and agonist-dependent 269–271 (1982). 17–20 (1994). homo-oligomerizationof the thyrotropin-releasing hormone 75. Carrithers, M. D. & Lerner, M. R. Synthesis and 93. Gouldson, P. R. et al. Dimerization and domain swapping in receptor; detection in living cells using bioluminescence characterization of bivalent peptide ligands targeted to G-protein-coupled receptors. A computational study. resonance energy transfer. J. Biol. Chem. (in the press). G-protein-coupled receptors. Chem. Biol. 3, 537–542 23, S60–S77 (2000). 57. Rocheville, M. et al. Subtypes of the (1996). 94. Ng, G. Y. et al. Gamma-aminobutyric acid type B receptors assemble as functional homo-and heterodimers. J. Biol. 76. Vila-Coro, A. J. et al. HIV-1 infection through the CCR5 with specific heterodimer composition and postsynaptic Chem. 275, 7862–7869 (2000). receptor is blocked by receptor dimerization. Proc. Natl actions in hippocampal neurons are targets of 58. Roess, D. A., Horvat, R. D., Munnelly, H. & Barisas, B. G. Acad. Sci. USA 97, 3388–3393 (2000). anticonvulsant gabapentin action. Mol. Pharmacol. 59, receptors are self-associated in the 77. Zukin, R. S., Eghbali, M., Olive, D., Unterwald, E. M. & 144–152 (2001). plasma membrane. Endocrinology 141, 4518–4523 (2000). Tempel, A. Characterization and visualization of rat and This paper describes the formation of heterodimers κ κ κ 59. Bond, R. A. & Bouvier, M. Receptor-based Drug Design (ed. guinea pig brain -opioid receptors: evidence for 1 and 2 between GABABR2 and different splice variants of the

Leff, P.) 363–377 (Marcel Dekker, New York, 1998). opioid receptors. Proc. Natl Acad. Sci. USA 85, 4061–4065 GABABR1, leading to receptors with different

60. Ng, G. Y. et al. Identification of a GABAB receptor subunit, (1988). pharmacological selectivity to the action of

gb2, required for functional GABAB receptor activity. J. Biol. 78. AbdAlla, S., Lother, H. & Quitterer, U. AT1-receptor anticonvulsant drugs. Chem. 274, 7607–7610 (1999). heterodimers show enhanced G-protein activation 95. Xu, Y., Piston, D. W. & Johnson, C. H. A bioluminescence

61. Kuner, R. et al. Role of heteromer formation in GABAB and altered receptor sequestration. Nature 407, 94–98 resonance energy transfer (BRET) system: application to receptor function. Science 283, 74–77 (1999). (2000). interacting circadian clock proteins. Proc. Natl Acad. Sci.

62. Couve, A. et al. Intracellular retention of recombinant GABAB 79. Gines, S. et al. Dopamine D1 and adenosine A1 receptors USA 96, 151–156 (1999). receptors. J. Biol. Chem. 273, 26361–26367 (1998). form functionally interacting heteromeric complexes. Proc. 96. Hovius, R., Vallotton, P., Wohland, T. & Vogel, H. 63. Margeta-Mitrovic, M., Jan, Y. N. & Jan, L. Y. A trafficking Natl Acad. Sci. USA 97, 8606–8611 (2000). Fluorescence techniques: shedding light on ligand-receptor checkpoint controls GABA(B) receptor heterodimerization. 80. George, S. R. et al. Oligomerization of µ- and δ-opioid interactions. Trends Pharmacol. Sci. 21, 266–273 27, 97–106 (2000). receptors. Generation of novel functional properties. J. Biol. (2000). Identification of an endoplasmic retention signal Chem. 275, 26128–26135 (2000). 97. Njuki, F. et al. A new calcitonin-receptor-like sequence in rat

within the carboxyl tail of the GABABR1 that is masked 81. Rocheville, M. et al. Receptors for dopamine and pulmonary blood vessels. Clin. Sci. 85, 385–388 (1993).

by heterodimerization with GABABR2. In addition to somatostatin: formation of hetero-oligomers with enhanced 98. Fluhmann, B., Muff, R., Hunziker, W., Fischer, J. A. & Born, W. highlighting the importance of dimerization for the functional activity. Science 288, 154–157 (2000). A human orphan calcitonin receptor-like structure. Biochem. transport of G-protein-coupled receptors, this paper 82. Jordan, B. A., Trapaidze, N., Gomes, I., Nivarthi, R. & Devi, Biophys. Res. Commun. 206, 341–347 (1995). β also shows that GABAB receptor heterodimerization is L. A. Oligomerization of opioid receptors with 2-adrenergic 99. McLatchie, L. M. et al. RAMPs regulate the transport and required for function once it has reached the cell receptors: a role in trafficking and mitogen-activated protein ligand specificity of the calcitonin-receptor-like receptor. surface. kinase activation. Proc. Natl Acad. Sci. USA 98, 343–348 Nature 393, 333–339 (1998). 64. Morello, J. P. et al. Pharmacological chaperones rescue cell- (2001). The first demonstration that heterodimerization surface expression and function of misfolded V2 vasopressin 83. Jordan, B. A. & Devi, L. A. G-protein-coupled receptor between a G-protein-coupled receptor (the calcitonin receptor mutants. J. Clin. Invest. 105, 887–895 (2000). heterodimerization modulates receptor function. Nature receptor-like receptor) and accessory proteins known 65. Deng, H. et al. Identification of a major co-receptor for 399, 697–700 (1999). as the receptor-activity-modifying proteins (RAMPs) is primary isolates of HIV-1. Nature 381, 661–666 (1996). 84. Zeng, F. Y. & Wess, J. Identification and molecular involved both in the cell-surface transport and the 66. Samson, M. et al. Resistance to HIV-1 infection in caucasian characterization of m3 muscarinic receptor dimers. J. Biol. pharmacological properties of the resulting individuals bearing mutant alleles of the CCR-5 chemokine Chem. 274, 19487–19497 (1999). calcitonin-gene-related peptide and adrenomedullin receptor gene. Nature 382, 722–725 (1996). 85. Cornea, A., Janovick, J. A., Maya-Nunez, G. & Conn, P. M. receptors. 67. Liu, R. et al. Homozygous defect in HIV-1 coreceptor Gonadotropin releasing hormone microaggregation: rate 100. Christopoulos, G. et al. Multiple amylin receptors arise from accounts for resistance of some multiply-exposed monitored by fluorescence resonance energy transfer. receptor activity-modifying protein interaction with the individuals to HIV-1 infection. Cell 86, 367–377 (1996). J. Biol. Chem. (in the press). calcitonin receptor gene product. Mol. Pharmacol. 56, 68. Karpa, K. D., Lin, R., Kabbani, N. & Levenson, R. The 86. Kunishima, N. et al. Structural basis of glutamate recognition 235–242 (1999). dopamine D3 receptor interacts with itself and the truncated by a dimeric metabotropic glutamate receptor. Nature 407, 101. Dwyer, N. D., Troemel, E. R., Sengupta, P. & Bargmann, C. I. D3 splice variant d3nf: D3-D3nf interaction causes 971–977 (2000). Odorant receptor localization to olfactory cilia is mediated by mislocalization of D3 receptors. Mol. Pharmacol. 58, The resolution of the three-dimensional structure of ODR-4, a novel membrane-associated protein. Cell 93, 677–683 (2000). the ligand-binding domain of the metabotropic 455–466 (1998). 69. Nimchinsky, E. A., Hof, P. R., Janssen, W. G. M., Morrison, glutamate receptor revealed that it is a dimer both in 102. Liu, F. et al. Direct protein–protein coupling enables cross- J. H. & Schmauss, C. Expression of dopamine D3 receptor the presence and absence of glutamate, indicating talk between dopamine D5 and γ-aminobutyric acid A dimers and tetramers in brain and in transfected cells. that G-protein-coupled receptors might be receptors. Nature 403, 274–280 (2000). J. Biol. Chem. 272, 29229–29237 (1997). constitutive dimers. This paper reports a physical association between a 70. George, S. R. et al. A transmembrane domain-derived 87. Tsuji, Y. et al. Cryptic dimer interface and domain seven-transmembrane-domain dopamine receptor

peptide inhibits D1 dopamine receptor function without organization of the extracellular region of metabotropic and a channel GABAA receptor, which leads to affecting receptor oligomerization. J. Biol. Chem. 273, glutamate receptor subtype 1. J. Biol. Chem. 275, reciprocal regulation of the two classes of receptors 30244–30248 (1998). 28144–28151 (2000). on co-activation. 71. Mijares, A., Lebesgue, D., Wallukat, G. & Hoebeke, J. From 88. Ray, K. & Hauschild, B. C. Cys-140 is critical for 103. Bergson, C. et al. Regional, cellular, and subcellular agonist to antagonist: Fab fragments of an agonist-like metabotropic glutamate receptor-1 dimerization. J. Biol. variations in the distribution of D1 and D5 dopamine monoclonal anti-β2-adrenoceptor antibody behave as Chem. 275, 34245–34251 (2000). receptors in primate brain. J. Neurosci. 15, 7821–7836 antagonists. Mol. Pharmacol. 58, 373–379 (2000). 89. Romano, C. et al. Covalent and noncovalent interactions (1995). 72. Conn, P. M., Rogers, D. C., Stewart, J. M., Niedel, J. & mediate metabotropic glutamate receptor mGlu(5) Sheffield, T. Conversion of a gonadotropin-releasing dimerization. Mol. Pharmacol. 59, 46–53 (2001). Acknowledgements hormone antagonist to an agonist. Nature 296, 653–655 90. Zhang, Z., Sun, S., Quinn, S. J., Brown, E. M. & Bai, M. The The author is grateful to Stephane Angers, Ali Salahpour, Jean- (1982). extracellular calcium-sensing receptor dimerizes through François Mercier, Sandrine Hilairet, Lynda Adam and Momique 73. Hazum, E. & Keinan, D. Gonadotropin releasing hormone multiple types of intermolecular interactions. J. Biol. Chem. Lagacé for the numerous discussions and their critical reading of activation is mediated by dimerization of occupied (2000). the manuscript.

286 | APRIL 2001 | VOLUME 2 www.nature.com/reviews/neuro © 2001 Macmillan Magazines Ltd