Elsevier

Brain Research Reviews

Volume 58, Issue 2, August 2008, Pages 475-482
Brain Research Reviews

Review
Novel pharmacological targets based on receptor heteromers

https://doi.org/10.1016/j.brainresrev.2008.06.002Get rights and content

Abstract

Studies performed in the last 10 years have provided solid evidence indicating that G-protein-coupled receptors are expressed on the plasma membrane as homo and heterodimers. The first consequence of this fact is that homo and heterodimers are the true targets of natural (hormones, neurotransmitters) and synthetic drugs. Furthermore a given receptor in a heteromer may display a different functional and/or pharmacological profile than the same receptor characterized as monomer or as homodimer. Recent evidence indicates that receptor heteromers are sensors that lead to a fine-tuning in neurotransmission or hormone regulation; mainly this is achieved by a modification of the signaling pathways activated via a given receptor when it is forming a given heteromer. Quite often antagonists display variable affinities when a given receptor is expressed with different heteromeric partners. This fact should be taken into account in the development of new drugs. Finally it should be pointed out that radioligand binding data has to be analyzed by a model that considers receptors as dimers and not as monomers. This model provides a novel approach to characterize drugs interacting with the orthosteric center (agonists/antagonists) or with allosteric centers (allosteric regulators).

Introduction

An increasing number of G-protein-coupled heptaspanning-membrane receptors (GPCRs) are reported to be expressed on the plasma membrane as dimers (Carriba et al., 2007, Bouvier, 2001, Devi, 2001, Rios et al., 2001, Agnati et al., 2003, Franco, 2003, Terrillon and Bouvier, 2004, Bulenger et al., 2005, Prinster et al., 2005, Herrick-Davis et al., 2006, Milligan, 2006, Milligan, 2007, Milligan and Kostenis, 2006, Pin et al., 2007). Quite often dimers are constitutively expressed in a given cell and a given cell can express both homodimers and a variety of heterodimers. From our work and the work of other laboratories on homo and heteromerization of receptors present in striatal neurons it has been feasible to demonstrate some consequences of homo and heteromerization in terms of both, ligand pharmacology and receptor function.

Key roles for receptor homo and heterodimerization include effective quality control of protein folding prior to plasma membrane delivery and interactions with heterotrimeric G-proteins (Bulenger et al., 2005, Milligan, 2007). At the plasma membrane level, there are several examples showing that cells sense neurotransmitters (or hormones) in a different way when they express one or another set of receptors that assemble into heterodimers (see for instance Gines et al., 2000, Canals et al., 2003). One of such examples is given by the lack of desensitization of D1 receptors (D1R) interacting with adenosine A1 receptors (A1R) in striatal GABAergic neurons unless both receptors in the D1R/A1R receptor heteromer are activated simultaneously (Fig. 1, Gines et al., 2000). Another example is at the biochemical level. An antagonistic A2AR–D2R interaction have been discovered in which the stimulation of A2AR decreases the affinity of D2R for agonists in membrane preparations (Ferré et al., 1991). The intramembrane A2AR–D2R interaction implies a close physical interaction between the two receptors. In fact, the pharmacology of D2R is affected by adenosine analogs activating A2AR (Canals et al., 2003). Signaling and/or desensitization may vary depending on whether one or the two receptors in the heteromer are activated and the resulting signal is very often not simply the addition of the signals given by individual activation of the receptors; even there are instances in which heteromer-mediated signaling becomes qualitatively different (see below).

Characterization of GPCRs by radioligand binding needs the use of a model considering that receptors are dimers. This model has been devised in two different laboratories (Durroux, 2005, Albizu et al., 2006, Franco et al., 2005, Franco et al., 2006) and, recently, simple equations useful to fit radioligand biding data to receptors dimers have been released (Casadó et al., 2007). Using these equations one can calculate the dissociation constants (KD values) for the binding of the first and the second molecule to the dimer but also a newly defined dimer cooperative index (DC). These parameters will likely be very useful to quantitatively determine receptor regulation, for instance by allosteric regulators (see below).

A variety of techniques have been used to detect heteromers, among them are co-immunoprecipitation and, more recently, energy transfer biophysical assays such as fluorescence resonance energy transfer (FRET) and bioluminescence energy transfer (BRET) which allow the demonstration of receptor heteromers in the natural environment of the living cell. However, these techniques are not appropriate to be applied in tissues, making more difficult the demonstration of neurotransmitter receptor heteromers in the brain. It is quite important to be able to detect receptor heteromers in natural tissues. For this purpose one has to look for the “dimer fingerprint” (Ferré et al., 2007a) which can be functional or biochemical. Significant examples of identification of dimer fingerprints are given below.

Section snippets

GPCR homo and heterodimers as targets for hormones, neurotransmitters and drugs

Neurotransmitter and hormone GPCRs cannot only be considered as single functional units, but as forming part of multimolecular aggregates localized in the plane of the plasma membrane (also called “horizontal molecular networks”), which can contain other interacting proteins, including receptors for the same or other neurotransmitters (Franco, 2003, Agnati et al., 2003, Agnati et al., 2005). The hypothesis on the existence of intramembrane receptor–receptor interactions was introduced in the

Pharmacology of GPCR heteromers

The easier detection of pharmacological profile modification of receptor heteromers has been achieved in the case of opioid receptors (Waldhoer et al., 2004, Rios et al., 2001) Different strategies have been considered for making opiates such as morphine more effective, while curbing its liability to be abused. In particular, delta opioid receptor ligands have been useful in enhancing morphine's potency. The underlying molecular basis is a physical association between mu and delta opioid

Fitting data to GPCR homo and heteromers

There are reasons for equivocal interpretations of the data fitting to receptor dimers assuming they are monomers. In this regard new models considering receptor dimers have been recently developed (Durroux, 2005, Albizu et al., 2006, Franco et al., 2005, Franco et al., 2006). Until now the approaches for fitting ligand binding data have been based on the existence of receptor monomers. From a recently devised model for receptor dimers, there exists a new approach for fitting data that gives

Quantifying allosteric effects

The possibility to calculate the dissociation constants for the binding of the first agonist/antagonist molecule (KD1) and the second molecule (KD2) and the cooperativity index (DC) for a homodimer allows easy quantitation of the effect of allosteric regulators. These are natural or synthetic molecules that interact with an allosteric center of the receptor altering the binding of agonists to the orthosteric center and subsequently regulating receptor activation. It should be noted that in the

Receptor–heteromer-specific drugs

Heteromerization of opioid receptors has been shown to alter opioid analog effects and affect receptor trafficking in cell culture model systems (Gomes et al., 2004, Waldhoer et al., 2004, Gupta et al., 2006). Waldhoer et al. (2005) demonstrated that 6′-guanidinonaltrindole has the unique property of selectively activating only opioid receptor heteromers but not homomers. The assayed compound induced analgesia in vivo depending on the place of administration. This study constitutes a proof of

Conclusions

Receptors occur as homodimers and/or heterodimers on the cell surface and therefore dimers/oligomers are the real targets for agonists/antagonists and for drugs interacting with these receptors at the orthosteric site. This is a concept that is currently overlooked by pharmaceutical companies, which concentrate in a single receptor whose pharmacological characterization is frequently performed using single transfected cells in which receptor heteromers cannot occur. The physiology of heteromers

Acknowledgments

This research was supported by grants SAF2006–00170 SAF2005–00170 from the Spanish Ministerio de Ciencia y Tecnología, grant 060110 from Fundació La Marató de TV3 and from Intramural Funds of the NIDA, NIH.

References (54)

  • GaoZ.-G. et al.

    Differential allosteric modulation by amiloride analogues of agonist and antagonist binding at A1 and A3 adenosine receptors

    Biochem. Pharmacol.

    (2003)
  • HeplerJ.R. et al.

    G proteins

    Trends Biochem. Sci.

    (1992)
  • Herrick-DavisK. et al.

    Serotonin 5-HT2C receptor homodimer biogenesis in the endoplasmic reticulum: real-time visualization with confocal fluorescence resonance energy transfer

    J. Biol. Chem.

    (2006)
  • HillionJ. et al.

    Coaggregation, cointernalization and codesensitization of adenosine A2A receptors and dopamine D2 receptors

    J. Biol. Chem.

    (2002)
  • LeeS.P. et al.

    Dopamine D1 and D2 receptor co-activation generates a novel phospholipase C-mediated calcium signal

    J. Biol. Chem.

    (2004)
  • MilliganG.

    G-protein-coupled receptor heterodimers: pharmacology, function and relevance to drug discovery

    Drug Discov. Today.

    (2006)
  • MilliganG.

    G protein-coupled receptor dimerisation: molecular basis and relevance to function

    Biochim. Biophys. Acta.

    (2007)
  • RiosC.D. et al.

    G-protein-coupled receptor dimerization: modulation of receptor function

    Pharmacol. Ther.

    (2001)
  • AgnatiL.F. et al.

    Aspects on receptor regulation and isoreceptor identification

    Med. Biol.

    (1980)
  • AgnatiL.F. et al.

    New vistas on synaptic plasticity: the receptor mosaic hypothesis of the engram

    Med. Biol.

    (1982)
  • AgnatiL.F. et al.

    Molecular mechanisms and therapeutical implications of intramembrane receptor/receptor interactions among heptahelical receptors with examples from the striatopallidal GABA neurons

    Pharmacol. Rev.

    (2003)
  • AgnatiL.F. et al.

    Allosteric modulation of dopamine D2 receptors by homocysteine

    J. Proteome Res.

    (2006)
  • AlbizuL. et al.

    Probing the existence of G protein-coupled receptor dimers by positive and negative ligand-dependent cooperative binding

    Mol. Pharmacol.

    (2006)
  • BouvierM.

    Oligomerization of G-protein-coupled transmitter receptors

    Nat. Rev. Neurosci.

    (2001)
  • CanalsM. et al.

    Homodimerization of adenosine A2A receptors: qualitative and quantitative assessment by fluorescence and bioluminescence energy transfer

    J Neurochem.

    (2004)
  • CarribaP. et al.

    Striatal adenosine A(2A) and cannabinoid CB(1) receptors form functional heteromeric complexes that mediate the motor effects of cannabinoids

    Neuropsychopharmacology.

    (2007)
  • CiruelaF. et al.

    Presynaptic control of striatal glutamatergic neurotransmission by adenosine A1–A2A receptor heteromers

    J. Neurosci.

    (2006)
  • Cited by (28)

    • Pharmacological potential of varinic-, minor-, and acidic phytocannabinoids

      2020, Pharmacological Research
      Citation Excerpt :

      CB1R and CB2R receptors exhibit complex pharmacology, and their signaling (which is also complex) and potential as therapeutic targets have been addressed in several reviews [4–9]. It is already known that these receptors may form heteroreceptor complexes between themselves, as well as with other GPCRs [10–13]. By definition, these complexes display properties other than those that are displayed by individual receptors [14].

    • L-DOPA disrupts adenosine A<inf>2A</inf>-cannabinoid CB<inf>1</inf>-dopamine D<inf>2</inf> receptor heteromer cross-talk in the striatum of hemiparkinsonian rats: Biochemical and behavioral studies

      2014, Experimental Neurology
      Citation Excerpt :

      Several reports have demonstrated the localization of CB1 and D2 receptors in the BG (Egertová and Elphick, 2000; Mátyás et al., 2006; Pickel et al., 2006; Uchigashima et al., 2007; Yin and Lovinger, 2006), predominantly in the soma and dendrites of striato-pallidal GABA neurons and in cortico-striatal glutamate terminals where A2A receptors are also present (Carriba et al., 2007; Egertová and Elphick, 2000; Ferré et al., 2007; Pickel et al., 2006; Uchigashima et al., 2007; Yin and Lovinger, 2006). Co-immunoprecipitation and co-localization assays (Hillion et al., 2002), as well as bioluminescence resonance energy transfer (BRET) and fluorescence resonance energy transfer (FRET) techniques (Canals et al., 2003), have shown that A2A–D2 receptors can form homo- and heteromers (Franco et al., 2008; Hillion et al., 2002). Functional interactions between the CB1 receptor and the A2A or D2 receptors, relevant for striatal function, have also been reported (Carriba et al., 2007; Marcellino et al., 2008).

    • Challenges in the development of heteromer-GPCR-based drugs

      2013, Progress in Molecular Biology and Translational Science
      Citation Excerpt :

      This certitude demands that new models be developed for interpreting ligand-binding assays to identify allosteric modulators. Recently, attempts have been made to incorporate heteromer formation into new models for ligand binding.58–62 Further, refinement of these models may be required, but they clearly demonstrate that classical models of ligand binding are less relevant when studying receptor heteromers.

    • Neuromuscular transmission modulation by adenosine upon aging

      2012, Neurobiology of Aging
      Citation Excerpt :

      In these conditions blockade of the A1 receptor molecule could induce a conformational change in the heteromer that does not allow either A2A agonist binding or effective coupling to the A2A transducing system. Indeed, heteromerization of G protein coupled receptors may affect several aspects of receptor functioning (see Franco et al., 2008), so that monomers and heteromers can have distinct pharmacological and signaling properties. Therefore, age-dependent changes in the proportion of adenosine receptors in a heteromeric or monomeric form may, at least in part, be responsible for the lost of excitatory effects in aged rats(2).

    View all citing articles on Scopus
    View full text