<<

View metadata, citation and similar papers at core.ac.uk brought to you by CORE

provided by Elsevier - Publisher Connector

Biochimica et Biophysica Acta 1808 (2011) 1245–1255

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta

journal homepage: www.elsevier.com/locate/bbamem

Review containing oligomers: Their role in the control of dopamine and glutamate neurotransmission in the brain☆

Francisco Ciruela a,⁎, Maricel Gómez-Soler a, Diego Guidolin b, Dasiel O. Borroto-Escuela c, Luigi F. Agnati d, Kjell Fuxe c, Víctor Fernández-Dueñas a

a Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina, Universitat de Barcelona, 08097 L'Hospitalet de Llobregat, Spain b Department of Human Anatomy and Physiology, University of Padova, Padova, Italy c Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden d IRCCS San Camillo, Lido Venezia, Italy

article info abstract

Article history: While the G protein-coupled receptor (GPCR) oligomerization has been questioned during the last fifteen years, Received 22 November 2010 the existence of a multi-receptor complex involving direct receptor–receptor interactions, called receptor Received in revised form 2 February 2011 oligomers, begins to be widely accepted. Eventually, it has been postulated that oligomers constitute a distinct Accepted 5 February 2011 functional form of the GPCRs with essential receptorial features. Also, it has been proven, under certain Available online 18 February 2011 circumstances, that the GPCR oligomerization phenomenon is crucial for the receptor biosynthesis, maturation, trafficking, plasma membrane diffusion, and pharmacology and signalling. Adenosine receptors are GPCRs that Keywords: G protein-coupled receptors mediate the physiological functions of adenosine and indeed these receptors do also oligomerize. Accordingly, Adenosine receptors oligomers may improve the molecular mechanism by which extracellular adenosine signals Protein–protein interaction are transferred to the G proteins in the process of receptor transduction. Importantly, these adenosine receptor- GPCR oligomerization containing oligomers may allow not only the control of the adenosinergic function but also the fine-tuning modulation of other neurotransmitter systems (i.e. dopaminergic and glutamatergic transmission). Overall, we underscore here recent significant developments based on adenosine receptor oligomerization that are essential for acquiring a better understanding of neurotransmission in the central nervous system under normal and pathological conditions. This article is part of a Special Issue entitled: “Adenosine Receptors”. © 2011 Elsevier B.V. All rights reserved.

Contents

1. Introduction ...... 1245 2. Adenosine receptor containing oligomers ...... 1246

2.1. The adenosine A1 and A2A receptors heterodimer (A1R/A2AR) and the control of glutamate release in the central nervous system . . . 1247

3. A1R containing oligomers ...... 1249 3.1. A1R and dopamine D1 receptor (D1R) oligomers ...... 1249

3.2. A1R and metabotropic glutamate type 1 (mGlu1) receptor oligomers ...... 1249

4. Adenosine A2 receptor (A2R) containing oligomers ...... 1250

4.1. A2AR and dopamine D2 receptor (D2R) oligomers ...... 1250 4.2. A2AR and metabotropic glutamate type 5 (mGlu5)receptoroligomers...... 1251 5. The striatal spine module as a paradigm of ARs integrative functioning in the brain ...... 1251 6. Concluding remarks ...... 1252 Acknowledgements ...... 1252 References ...... 1252

☆ This article is part of a Special Issue entitled: “Adenosine Receptors”. 1. Introduction ⁎ Corresponding author at: Unitat de Farmacologia, Dept. de Patologia i Terapèutica Experimental, Facultat de Medicina-Bellvitge, Pavelló de Govern, Av. Feixa Llarga, s/n, Adenosine is an endogenous nucleoside mostly formed as a 08907 L'Hospitalet del Llobregat, Barcelona, Spain. Tel.: +34 934024280, +34 934035820; fax: +34 934029082. degradation product of (ATP) and to a lesser E-mail address: [email protected] (F. Ciruela). degree from S-adenosyl-L-homocysteine (SAH) metabolism. As soon

0005-2736/$ – see front matter © 2011 Elsevier B.V. All rights reserved. doi:10.1016/j.bbamem.2011.02.007 1246 F. Ciruela et al. / Biochimica et Biophysica Acta 1808 (2011) 1245–1255 as adenosine has been generated it can be either intracellularly physiological or behavioral responses remains quite difficult because phosphorylated to form AMP or eliminated out of the cells by means of the lack of good A2BR selective agonists or antagonists [12]. of ubiquitous nitrobenzylthioinosine-sensitive equilibrative nucleo- Ultimately, the A3R has also been found to inhibit adenylyl cyclase, side transporters (ENTs). Additionally, adenosine can react with L- and to promote mobilization probably by direct G protein βγ- homocysteine to form SAH. Finally, both intra- and extracellular subunits PLC activation (for review see [3])(Table 1). Interestingly, in adenosine can be deaminated to form by the action of intra- humans the A3R is a high affinity receptor antagonized by xanthines and ecto-adenosine deaminase, respectively. Interestingly, as an [13,14]. Finally, it is important to mention here that different classes of intermediate metabolite in several biochemical pathways, adenosine proteins, other than G proteins, have been shown to interact with plays a key regulatory role in numerous physiologic processes, adenosine receptors and to eventually modify their functioning [15]. including platelet function, coronary and systemic vascular tone, and lipolysis in adipocytes. In the nervous system, since the 2. Adenosine receptor containing oligomers extracellular concentration of adenosine raises as a function of the neuronal activity, it acts as an energy dependent neuromodulator The concept of GPCRs oligomerization was first postulated in the through combined presynaptic, postsynaptic and non-synaptic 1980s by Luigi F. Agnati and Kjell Fuxe, who proposed that an actions [1,2]. Therefore, extracellular adenosine regulates several intramembrane neuropeptide and –receptor functions in the brain, including neuronal viability, neuronal mem- interaction was responsible for the functional cross-talk observed brane potential, propagation of action potentials, astrocytic functions, between these two neurotransmitter systems [16,17]. The initial microglia reactivity, primary metabolism in both neurons and indirect evidence pointing to the existence of GPCR oligomers astrocytes, and blood flow [2]. Consequently, the physiological roles containing neuropeptide and monoamine receptors was established and potential therapeutic use of adenosine have been extensively by means of radioligand-binding experiments. Thus, by using this revised over the last years [3,4]. approach the existence of interactions between neurokinin NK1 and

Adenosine mediates its actions through the activation of specific 5-HT1 receptors, CCK-2 and serotonin 5-HT2 receptors, vasoactive GPCRs, for which four subtypes have been identified (A1R, A2AR, A2BR intestinal peptide (VIP) and serotonin 5-HT1 receptors, cholecystoki- and A3R) [5]. These receptors have a distinctive pharmacological nin CCK-2 and dopamine D2 receptors, neurotensin NTS1 and profile, tissue distribution and effector coupling [6], and its function- dopamine D2 receptors, and neuropeptide Y (NPY) and α2 adrenergic ing has been extensively studied in the central nervous system (CNS) receptors were established (for review see [18]). In addition, some

(Table 1). Interestingly, A1Rs and A2ARs are largely responsible for the photo-affinity labelling and radiation inactivation experiments, central effects of adenosine [7]. The most abundant and homoge- together with hydrodynamic and cross-linking analysis, also sup- neously distributed adenosine receptor in the brain is the inhibitory ported the GPCR oligomerization idea [19–22]. Later on, as the

A1R, which is functionally coupled to members of the pertussis toxin- existence of some GPCR homodimers in native tissue was demon- sensitive family of G proteins (Gi/o) and whose activation regulates strated (i.e. the dopamine D2 and the the activity of membrane and intracellular proteins such as adenylate homodimers in the brain [23,24]), the GPCR oligomerization issue cyclase, Ca2+ channels, K+ channels and phospholipase C (Table 1) began to gain significance, given the potential physiological implica-

[8]. In contrast, the A2AR is expressed at high levels in only a few tions of this phenomenon. Nevertheless, the straight evidence for a regions of the brain, namely the striatum, the and direct receptor–receptor interaction within the GPCR oligomerization the nucleus accumbens [9]. The A2AR is mostly coupled to Gs in the phenomenon was achieved by means of biophysical techniques based peripheral systems but mediates its effects predominantly through on resonance energy transfer (RET), like bioluminescence-RET (BRET) activation of Golf in the striatum [10], thus activating adenylyl cyclase and fluorescence-RET (FRET) (for review see [25]). In addition, the which in turn converts ATP into cAMP (Table 1). Next, the A2BRis combination of some fluorescence-based approaches allowed the positively coupled to adenylyl cyclase and PLC through a Gs and Gq identification of higher-order GPCR oligomers or receptor mosaics protein, respectively [11] (Table 1). The A2BR is thought to be fairly (RMs) in living cells [26–32]. Finally, these powerful approaches have ubiquitous in the brain, but the association of the A2BR to specific been recently used to demonstrate the existence of GPCR oligomers in

Table 1 Adenosine receptors.

a b Receptor Adenosine affinity (EC50) G protein Transduction mechanisms Physiological actions in brain

c A1R ~50 nM Gi1,2,3 Inhibits AC (↓cAMP) Inhibits synaptic transmission;

Activates PLC (↑IP3/DAG) hyperpolarizes neurons. Activates PLA2 (↑AA) Activates PLD (↑PEtOH) Activates GIRKs Inhibits Ca2+ channels

Go c A2AR~1μMGS Activates AC (↑cAMP) Facilitates transmitter release;

Golf Activates AC (↑cAMP) regulation of sensorimotor d G15,16 ↑IP3 integration in basal ganglia. Inhibits Ca2+ channels c A2BR N10 μMGS Activates AC (↑cAMP) Increases in cAMP in brain slices Activates Ca2+ channels d Gq/11 Activates PLC (↑IP3/DAG) c A3R ~50 nM Gi2,3 Inhibits AC (↓cAMP) Uncouples A1R and mGlu

Activates PLC (↑IP3/DAG) receptors a Data are from a cyclic AMP functional assay in CHO cells expressing the corresponding adenosine receptor [55]. The assay is performed in the presence of the adenosine transport inhibitor nitrobenzylthioinosine (NBTI) to avoid adenosine uptake. b AC, adenylyl cyclase; PLC, phospholipase C; IP3, triphosphate; DAG, diacylglycerol; PLA2, phospholipase A2; PEtOH, phosphatidylethanol; GIRKs, G protein-dependent inwardly rectifying K+ channels; AA, arachidonic acid. c Main mechanism of coupling. d Receptor transfected cell system. F. Ciruela et al. / Biochimica et Biophysica Acta 1808 (2011) 1245–1255 1247 native tissue (e.g. the presence of dimers and/or The question that is still remaining focuses on whether the oligomers in mammary glands) [33]. formation of the A1R/A2AR heteromer might play a physiological role In the last years, a set of publications have demonstrated the in native brain preparations. This issue is of major relevance, since the existence of adenosine receptors containing oligomers. For instance, the involvement of GPCR oligomerization in pathophysiological condi-

A1R has been found to heteromerize with the dopamine D1 receptor tions remains unexplored. Interestingly, there is strong evidence (D1R), this phenomenon being essential for differential desensitization pointing to the existence of the A1R/A2AR heteromer in the brain areas mechanisms and for receptor trafficking [34]. Similarly, the heterodimer where the distribution of A1Rs and A2ARs clearly overlaps, for instance composed by the A1R and the metabotropic glutamate type 1α in the dopamine-rich regions [51,52]. Indeed, ultrastructural analysis (mGlu1α) receptor seems to play a key role in preventing glutamate by immunolelectron microscopy experiments in the rat striatum have excitotoxicity [35], whereas the physiological role for the proved revealed that these receptors codistribute and colocalize in striatal heteromerization of the A1RandthepurinergicP2Y1 receptor (P2Y1R) is glutamatergic synapses [47] (Fig. 2). In addition, co-immunoprecip- still under debate [36]. On the other hand, the A2AR also possesses the itation studies have clearly shown that the A1R and A2AR are co- ability to oligomerize with the dopamine D2 receptor (D2R) [37,38]. purified from rat striatal synaptosomes when specific antibodies are Interestingly, the latter receptor–receptor interaction underlies the used, thus suggesting the existence of A1R/A2AR heteromers in the molecular mechanism behind the antagonistic adenosine-dopamine striatal glutamatergic synapses and/or perisynaptic regions [47]. interactions that regulate the function of the GABAergic enkephalinergic In the excitatory glutamatergic synapses the most accepted role for neurons [39,40]. Finally, the A2AR also heteromerizes with the adenosine is the inhibition of synaptic transmission (i.e. inhibition of metabotropic glutamate type 5 (mGlu5) receptor [41],andinthis glutamate release) through the activation of presynaptic A1Rs. In fact, case, a synergistic functional interaction has been demonstrated at both the A1R constitutes the prototypical inhibitory Gi/o protein-coupled biochemical and behavioral levels [42–44]. receptor whose stimulation decreases the probability of neurotrans- mitter release [53]. On the other hand, the use of particular

2.1. The adenosine A1 and A2A receptors heterodimer (A1R/A2AR) and the stimulating conditions together with pharmacological tools with control of glutamate release in the central nervous system adequate selectivity to hamper A2AR functioning, has made possible the identification of a parallel capacity of adenosine to facilitate the

A1Rs and A2ARs operate through opposing signal transduction pathways (Table 1). Thus, they have been historically considered as independent entities in the framework of adenosine physiology. Interestingly, these adenosine receptors were shown to form homo- dimers in both ectopic expression systems and in native tissue [24,45]. It was then postulated that the functional receptor placed at the cell surface of cells was represented by a homodimer [45]. In addition, it was shown that A1Rs and A2ARs were coexpressed in a variety of cells like glutamatergic neurons of the hippocampus [46] and the striatum

[47]. Furthermore, heterodimers involving A1Rs and A2ARs were described in the context of the modulation of the glutamatergic neurotransmission in the central nervous system [47]. Nevertheless, the existence of these dimeric forms involving either A1Rs or A2ARs did not first alter the widespread acceptance that these two adenosine receptors with opposite physiological functions would signal inde- pendently. However, some evidence strongly indicated a potential interaction between A1Rs and A2ARs [48–50][48], suggesting the possibility that these receptors could establish a molecular and/or functional cross-talk, i.e. as if they were part of the same molecular transduction complex or signalosome, thus affording a logical and economical device to fine-tune adenosine neuromodulation. Initially, as described for the neuropeptide and monoamine receptor–receptor interaction studies (see above), the functional cross-talk between the A1R and the A2AR was assessed by means of radioligand-binding experiments. Thus, these approaches revealed that the formation of the A1R/A2AR heteromer per se did not modify the affinity of A1Rs or A2ARs for their respective agonists. But interestingly, when the A1R/A2AR heteromer was challenged with an A2AR agonist, the affinity of the A1R for its agonist decreased significantly. Conversely, the activation of the A1R/A2AR heteromer with an A R agonist did not alter the A R binding characteristics. 1 2A Fig. 1. Scheme of the proposed concentration-dependent adenosine receptor hetero-

Therefore, it could be concluded that, within the A1R/A2AR heteromer, dimer switch. Upper panel shows the efficacy of adenosine to stimulate A1Rs and A2ARs the agonist-mediated A1R responsiveness is modulated by the A2AR, within the A1R/A2AR heterodimer. Low concentrations of adenosine activate predom- whereas the binding properties of the latter were not altered by the inantly A1Rs, which inhibits glutamate release. High concentrations of adenosine also activate A2ARs, which, by means of the A1R/A2AR intramembrane interaction (blue-red A1R challenge. This ability of the A2AR to control the A1R within the fi arrow), antagonizes A1R function, therefore facilitating glutamate release. The A1R/A2AR heteromer has been further con rmed by functional schematic GPCRs diagrams were prepared using PyMOL (PyMOL Molecular Graphics experiments (i.e. determination of receptor-mediated second mes- System, DeLano Scientific, San Carlos, CA, USA), with the crystal structure of the sensory senger generation). For instance, upon the formation of the A1R/A2AR II (PDB: 1JGJ) as models. In the lower panel the proposed kinetics of the A1R/ A2AR heterodimer-mediated glutamate release control is shown. Thus, increasing heteromer, the A2AR challenge significantly decreased the A1R- concentrations of adenosine (0.1–100 μM) produced a biphasic effect, with low and mediated intracellular calcium mobilization [47]. Overall, these high concentrations inhibiting and stimulating the evoked release of glutamate in results suggest that under certain circumstances the A2AR can control striatal synaptosomes, respectively. the A1R functionality, whereas the reverse does not happen. Adapted from Ref. [47] 1248 F. Ciruela et al. / Biochimica et Biophysica Acta 1808 (2011) 1245–1255

evoked glutamate release via activation of presynaptic A2ARs [54] the A1R/A2AR heteromer is found in the glutamatergic terminals of the (Fig. 2). Therefore, one can easily ask the following question: how striatal spine module (SSM) (see below and Fig. 2). Thus, the does adenosine decide between A1Rs and A2ARs in the glutamatergic extracellular adenosine switches from inhibition to facilitation thanks nerve terminal in order to control glutamate release? To answer this to the A1R/A2AR heteromer [47]. It is important to keep in mind that question several facts need to be primarily established. First, that the this scenario might be true in brain and related to the control of

A1R and the A2AR have different affinities for adenosine with the neurotransmitter release, where a proper A1R and A2AR co-localiza- former showing a higher affinity for this nucleoside than the latter tion and stoichiometry allow the existence of a balanced A1R/A2AR (Table 1) [55]. Therefore, at low concentrations of adenosine the A1R oligomer. However, in the situation where either an unbalanced A1R/ will be primarily stimulated and glutamate release inhibited (Fig. 1). A2AR oligomer exist (i.e. existence of spare A2AR that sensitize the In contrast, at high concentrations of adenosine both A1Rs and A2ARs A2AR-mediated adenosine response) or that the A1R and the A2ARdo could potentially be activated, and thus the question arises how the not oligomerize (i.e. do not co-distribute), then the predominance of a control of glutamate release will be established under these distinctive adenosine receptor-mediated function (A1RorA2AR) will circumstances. How can the integration of A1Rs and A2ARs signals be observed, as it has been recently described [56]. Thus, more work is take place? The evidence described so far point to the fact that the needed to fully characterize the functionality of the A1R/A2AR A1R/A2AR heteromer located at the glutamatergic terminal makes oligomer outside the SSM and also the mechanism by which the possible this integration, since the A1R responsiveness within the oligomer couples to G protein(s) (i.e. do the A1R/A2AR oligomer heteromer is under the control of the A2AR. Therefore, at high couples to a single G protein or simultaneously to Gi and GS?). Overall, concentrations of adenosine the A2AR stimulation will abrogate the the A1R/A2AR heteromer confers a rationale for the existence of A1R functioning by means of a phenomenon of allosteric cross- heteromers of isoreceptors (receptors for the same neurotransmitter) inhibition, which will lead to predominant facilitation of glutamate and demonstrates that neurotransmitter heteromers composed of release (Fig. 1). Then, it can be concluded that A1R/A2AR heteromer- isoreceptors with different affinities for their endogenous neuro- ization provides a molecular framework that allows adenosine to transmitter can act as concentration-dependent processors that exert exert a fine-tune modulation of glutamate release in the striatum as a fine-tune modulation of neurotransmission (Fig. 1).

Fig. 2. Schematic representation of the striatal spine module (SSM) within the striatal spiny neuron (MSN). (a) Immunoreactivity of NECAB2, an A2AR interacting protein, in the rat striatum. As described for the A2AR the immunoreactivity was very strong in the caudate putamen (Cpu) [130] as revealed by a pre-embedding immunoperoxidase method at the light microscopic level. Ctx, cortex; LS, lateral septum; LV, lateral ventricle. Adapted from Ref. [130]. (b) High magnification picture showing immunoreactivity for NECAB2 in a

GABAergic neuron of the caudate putamen. Adapted from Ref. [130]. (c) Electron micrograph showing the presynaptic co-localization of A1Rs and A2ARs in rat striatum. Peroxidase reaction product (immunoreactivity for A1Rs) filled axon terminals -B- establishing asymmetrical synapses with spines -S-, in which immunoparticles (immunoreactivity for A2ARs) were localized along the extrasynaptic plasma membrane (arrows). Adapted from Ref. [47]. (d) Subcellular distribution of A2AR, D2R and mGlu5 receptor in rat striatum. Electron micrograph showing immunoreactivity for A2AR, D2R and mGlu5 receptor in rat striatum as revealed using a triple-labeling post-embedding immunogold technique.

Immunoparticles for A2AR (10 nm size, arrows), D2R (15 nm size, crossed arrows) and mGlu5 receptor (20 nm size, arrowheads) were detected along the extrasynaptic and perisynaptic plasma membrane of the same dendritic spine (s) establishing excitatory synaptic contact with axon terminals. Adapted from Ref. [129]. (e) Schematic representation of the SSM. The MSN is the most common neuron in the striatum and it receives two main inputs: glutamatergic afferents from cortical, limbic and thalamic areas, and dopaminergic afferents from the mesencephalon. The A1R/A2AR heterodimer is located in the glutamatergic terminal and controls the glutamate release. The A2AR/D2R/mGlu5 receptor oligomer is located in the dendritic spine of the enkephalin medium spiny neuron and controls the excitability of the MSN. The differential stimulation of the units of the receptor heteromers

(e.g. A1R/A2AR and A2AR/D2R/mGlu5 receptor) determines the predominant signalling pathway (see [42,43]). The extracellular adenosine levels are thought to be regulated by a glial- based adenosine cycle [131]. Glial cells can release ATP via direct release through hemichannels (h-ch) and/or by vesicular release. Also, ATP can be co-released with glutamate at the synaptic cleft. The ATP at the extracellular level is quickly degraded into adenosine (Ado) by ectonucleotidases. Ado can also be released directly via equilibrative nucleoside transporters (nt). At the intracellular level adenosine concentration is mainly controlled by adenosine kinase, which is part of a substrate cycle between adenosine and AMP and finally ATP [131]. F. Ciruela et al. / Biochimica et Biophysica Acta 1808 (2011) 1245–1255 1249

3. A1R containing oligomers changing the dissociation constants of the high- and the low affinity binding sites [68,71,72]. Thus, the high affinity form of the receptor

3.1. A1R and dopamine D1 receptor (D1R) oligomers would be partially or completely lost in the presence of agonists activating A1Rs, while in the absence of cross-modulation the binding Dopamine plays a key role in a large variety of functions in the CNS of dopamine to the D1R would involve both the low affinity (80-90%) including short-term memory and motor, attention and reward and the high affinity form of the receptor [72]. Similarly, it was control [57–59]. Imbalance in dopaminergic neurotransmission has reported that the adenosine A1R selective antagonist DPCPX had an been associated with drug addiction [60] and CNS alterations in stress effect on the Ki value of the D1R selective agonist SKF 38393 in A1R/ conditions [61]. Furthermore, dysfunction of dopaminergic neuro- D1R transfected cells, thus adenosine would exert a tonic inhibition of transmission has been also involved in psychiatric and neurological dopamine in the D1R mediated function through the A1R/D1R complex disorders including Parkinson's disease, schizophrenia and bipolar [73]. In addition, radioligand binding experiments performed in disorders [62,63]. Dopamine specifically acts on GPCRs located in the cotransfected A1R/D1R cells showed that the adenosine A1R-mediated plasma membrane of cells and, based on morphological and modulation of D1R induced a concentration-dependent decrease in pharmacological characteristics, these receptors are classified as D1- the affinity of the D1R agonist SKF38393 [74]. Overall, it is now well like, which include the D1R and the D5R, and D2-like receptors, which accepted that the simultaneous activation of the A1R/D1R heteromer include the long and short isoforms of the D2R (termed D2LR and D2SR, may allow the antagonistic intramembrane receptor–receptor inter- respectively), the D3R and the D4R [63]. action to take place, and it has been suggested that one functional The modulatory effects of adenosine on dopamine systems have meaning of this interaction is the uncoupling of the D1R from Gs been investigated in view of their relevance to human pathology such protein with the disappearance of the D1R high affinity state [75]. as schizophrenia and Parkinson's disease. Antagonistic adenosine/ On the other hand, the uncoupling from the downstream signaling dopamine interactions have been widely reported, showing that effectors may also be dependent of A1R/D1R heteromerization, which adenosine can inhibit several effects of dopamine in the cerebral in fact appears to control the temporal dynamics of receptor cortex and basal ganglia. In rat models of Parkinson's disease, for activation and down-regulation [69]. As described earlier, upon co- instance, it was shown that nonselective adenosine receptor antago- activation of A1Rs and D1Rs in co-transfected fibroblast cells and in nists (i.e. either or theophyllamine) enhanced the effect of cortical neurons in culture the heteromer complex was maintained, L-DOPA and other agonists increasing motor while during individual treatments heteromerization disappeared, activity [64]. Similarly, it was also described in unilaterally 6-OH- thus indicating that co-activation induced heteromer stabilization. dopamine (which specifically destroys the nigrostriatal dopamine Similarly, pretreatment with an A1R agonist caused co-clustering (co- pathway) lesioned rats that the A1RselectiveantagonistCPT aggregation) of A1Rs and D1Rs, which was blocked by combined enhanced the motor activating effects of the D1R selective agonist pretreatment with A1R and D1R agonists [69]. These results indicated SKF 38393 [65], while the A1R selective agonist CPA counteracted SKF that the movement of the heteromer and/or clusters of heteromers in 38393-induced grooming behavior [66]. Hence, in order to explain the the plasma membrane could be agonist-dependent, and that receptor results obtained in a large number of behavioral studies, it was trafficking may depend on the activity of the two receptors of the suggested that antagonistic adenosine/dopamine interactions were, at heteromer. As a result, it has been suggested that D1R desensitization least in part, caused by an intramembrane interaction between may be mainly caused by a prolonged allosteric change in the D1R specific subtypes of dopamine and adenosine receptors, in this case brought about by the A1R/D1R heteromeric complex; specifically A1Rs and D1Rs [67,68]. these changes could consist of phosphorylation events and/or The first evidence of a receptor–receptor interaction involving the association with beta-arrestin-like molecules that finally would lead

A1R and the D1R was provided by means of co-immunoprecipitation to a reduced Gs coupling of the D1R [76,77]. experiments and confocal microscopy analysis in co-transfected Consequently, the A1R/D1R receptor–receptor interaction in this fibroblast Ltk-cells and cortical neurons in culture [69]. Interestingly, heteromer is relevant not only for acute A1R antagonism of D1R this selective A1R/D1R heteromerization disappeared after pretreat- signaling but also for a persistent long-term antagonism of A1RtoD1R ment with a selective D1R agonist, but not after combined pretreat- signaling to the Gs protein. Therefore, it has been postulated that ment with D1R and A1R agonists. Likewise, the D1R uncoupling from novel therapeutic approaches, for instance in Parkinson's disease, may adenylyl cyclase only occurred when the A1R/D1R heteromer was co- be developed taking into account not only the particular character- activated, while the A1R agonist alone did not substantially reduce the istics of the receptors within the heteromer, but also the fact that the D1R agonist-induced accumulation of cAMP [69]. Similarly, by means pathology may induce changes in such receptor–receptor interactions of double immunofluorescence experiments with confocal microsco- involved in the function of neuronal networks of the brain, such as in py, a high degree of A1R and D1R co-localization was found in both co- the basal ganglia and prefrontal cortex [78]. transfected fibroblast cells and cortical neurons in culture [70]. In this study A1R/D1R coimmunoprecipitation was also demonstrated in 3.2. A1R and metabotropic glutamate type 1 (mGlu1) receptor oligomers native tissue, for instance in samples from rat nucleus accumbens.

Interestingly, in the native tissue A1R/D1R coimmunoprecipitation Glutamate is the main excitatory neurotransmitter in the mam- was reduced only when rats were treated with cocaine, while co- malian central nervous system and exerts its effects through specific activation of A1Rs prevented receptor uncoupling [70]. cell surface receptors, namely ionotropic glutamate (iGlu) receptors – The above-commented results may then explain the well- that form ion channels – and metabotropic glutamate (mGlu) documented antagonistic A1R/D1R receptor–receptor interactions receptors, which are coupled to G proteins [79,80]. To date, eight found in the neuronal networks of the brain, since they indicate members of the metabotropic family have been that A1R/D1R heteromerization could determine changes in the identified and categorized into three groups (I, II and III), based on pharmacological profile of the receptors, for instance, activation of their , agonist selectivity, and signal transduction the A1R could modulate D1R signaling. One of the mechanisms that pathways [79]. Group I contains the mGlu1 and mGlu5 receptor may explain this action is based on the ability of A1R activation within subtypes, which are coupled to phospholipase C in transfected cells the heteromer to change the binding characteristics of dopamine to and have quisqualic acid as their most potent agonist. Five splice the D1R. Accordingly, it was firstly described in crude membrane variants of the mGlu1 receptor have been described [79,81]. preparations from cell lines and rat striatum that activation of A1Rs Interestingly, these splice variants differ between them in the length reduced the proportion of D1Rs in the high-affinity state without of their C-terminal tail, which might play a role in the subcellular 1250 F. Ciruela et al. / Biochimica et Biophysica Acta 1808 (2011) 1245–1255

targeting of the receptor [82]. For instance, the C-terminal tail of the it is well accepted that the A2AR plays a modulatory role in the CNS in metabotropic glutamate type 1α (mGlu1α) receptor interacts with general and in the striatum in particular, and that this A2AR-mediated tubulin, and it has been proposed that the latter can regulate the cell neuromodulatory role is somehow related to the ability of this receptor surface expression of the receptor and its plasma membrane to heteromerize with other GPCRs. Indeed, in the last decade the anchoring [83,84]. Noteworthy, another cytoskeletal protein, 4.1 G, existence of a direct receptor–receptor interaction between A2ARs and has also been described as a binding partner of the mGlu1α receptor D2Rs has been demonstrated [38,92]. Thus, it has been postulated that [85]. This multifunctional protein, which is a critical component of the while the previously described A1R/D1R interaction would modulate the spectrin/actin cytoskeleton, plays an important structural and function of the GABAergic dynorphynergic neurons, the A2AR/D2R regulatory role in the stabilization and assembly of receptors into interaction would modulate the function of the GABAergic enkephali- the membranes. Interestingly, the 4.1 G protein has also been found to nergic neurons [67,68]. bind to the third intracellular loop of the A1R and to play a regulatory The A2AR/D2R receptor–receptor interaction takes place in both role in the trafficking and down-regulation of this receptor [85]. the somatodendritic area and in the nerve terminals of the GABAergic As described earlier, among the numerous neurophysiological enkephalinergic neurons [40,42,43,68]. At this level, the coexistence actions of adenosine, the inhibition of glutamate neurotransmission of two reciprocal antagonistic interactions between these receptors has been studied in several brain regions [7,86]. The activation of has been studied. First, it has been demonstrated that the stimulation presynaptic adenosine A1Rs, which leads to a direct inhibitory effect of of D2Rs in the globus pallidus produces a strong counteraction of 2+ G protein βγ-subunits on voltage-dependent Ca channels, has been A2AR-mediated GABA release [93]. Similarly, recent studies in animal described as one of the mechanisms involved in the decrease of the models of Parkinson's disease have suggested that the pallidal A2AR/ probability of glutamate release to the synaptic space [53,87–89]. D2R interaction may contribute to the antiparkinsonian effects of the Interestingly, the A1R-mediated fine-tuning modulation of glutama- co-administration of A2AR selective antagonists and L-DOPA or D2R tergic neurotransmission has been demonstrated to occur, at least in selective agonists. On the other hand, it has been shown that there part, by means of an interaction between the A1R and the mGlu1α exists a strong tonic activation of D2R blocking the ability of the A2AR receptor [35]. Thus, while A1R/mGlu1α receptor–receptor coupling to signal through the cAMP-PKA pathway. In this way, in vivo could occur through specific targeting proteins, such as protein 4.1 G, administration of a D2R selective antagonist in the rodent striatum a direct receptor–receptor interaction between the A1R and the produces a significant increase in the expression of c-fos and mGlu1α receptor would also be possible. In fact, the C-terminal tail of preproenkephalin , which depends on the ability of the D2Rto the mGlu1α receptor has been shown to be necessary to reach the A1R/ tonically block A2AR signaling activated by endogenous adenosine mGlu1α receptor heteromer formation, since other splice variants, as [94]. Overall, two reciprocal antagonistic A2AR/D2R interactions have the mGlu1β receptor with a shorter C-terminal tail or a deleted mutant been described, namely an intermembrane interaction in which the of the mGlu1α receptor lacking the C-terminal cytoplasmatic domain A2AR mediates the inhibition of the D2R, thus modulating neuronal did not interact with the adenosine A1R [35]. In these experiments excitability and neurotransmitter release, and an interaction at the biochemical and functional evidence was also provided for a A1R/ level of adenylyl-cyclase in which the D2R inhibits A2AR-mediated mGlu1α receptor receptor–receptor interaction. Co-immunoprecipi- protein phosphorylation and expression. tation experiments showed a close and subtype-specific interaction There are two possible mechanisms to explain the apparently between A1Rs and mGlu1α receptors in both rat cerebellar synapto- incompatible coexistence of two reciprocal antagonistic A2AR/D2R somes and co-transfected HEK-293 cells. Also, in primary cultures of interactions. Firstly, it has been suggested that there exists a different cortical neurons a high degree of co-localization was also observed G-protein coupling depending on the conformation of the D2R. [35]. In these rat cortical neurons, it was suggested by means of Accordingly, it has been proposed that the D2R couples to Gi/o, and NMDA-mediated excitotoxicity experiments that the timing in A1R therefore negatively to adenylyl-cyclase, when not being part of the and mGlu1α receptor activation is very important to achieve a A2AR/D2R heteromer, while it switches to Gq/11-PLC signaling when maximum effect in adenosine- and glutamate-mediated neuroprotec- heteromerization occurs. And secondly, the presence of a third tion/neurodegeneration. Similarly, A1Rs and mGlu1α receptors may partner has been proposed, namely the mGlu5 receptor, which form part of a signalling complex in vivo that could play a critical role could interact with the A2AR (see below). Interestingly, it has been in fine-tuning neurotransmission at glutamatergic synapses. This was shown both in transfected cells and in the striatum that upon co- substantiated by using transiently co-transfected HEK-293 cells, in stimulation of A2ARs and mGlu5 receptors a synergistic effect is 2+ which a synergy between both receptors in receptor-evoked [Ca ]i produced on c-fos expression related to changes at the adenyl-cyclase signaling was shown, as previously reported [90]. Overall, these and the MAPK levels [41,95]. In addition, in vivo experiments have results demonstrate that a functional cross-talk occurs between A1Rs demonstrated that co-stimulation A2ARs and mGlu5Rs in the presence and mGlu1α receptors. These results open new perspectives for the of a selective D2R agonist permits the A2AR to be liberated from the development of novel agents to treat neuropsychiatric disorders in tonic inhibitory effect of the D2RonA2A signals through the cAMP- which abnormal glutamatergic neurotransmission is involved. PKA pathway [96]. In the last years, we and others have demonstrated the existence of

4. Adenosine A2 receptor (A2R) containing oligomers the A2AR/D2R heteromer in living cells by biochemical (i.e. co- immunoprecipitation) and by biophysical (i.e. FRET and BRET)

4.1. A2AR and dopamine D2 receptor (D2R) oligomers approaches [38,40,97–99]. In addition, by computerized modeling, and pull-down and mass spectrometry techniques, we have also

In the striatum, more than the 95% of the neuronal population is demonstrated that the heteromerization between the A2AR and the comprised by the medium spiny GABAergic efferent neurons. There are D2R depends on a coulombic interaction between the third intracellular two subtypes of these GABAergic striatal neurons, the GABAergic loop (3IL) of the D2R and the C-terminal tail of the A2AR receptor striatopallidal neurons, which can be called enkephalinergic neurons, [38,92,100,101]. Therefore, a positively charged arginine rich motif since they express the peptide enkephalin, and the GABAergic located in the N-terminal part of the D2R-3IL may interact with two striatonigral-striatoentopeduncular neurons, called also as GABAergic different negatively charged motifs from the C-terminal tail of the A2AR, dynorphynergic neurons, since they express the peptide dynorphin namely the 388HELKGVCPEPPGLDDPLAQDGAVGS412 domain which (and also substance P). Interestingly, while the GABAergic enkephali- contains two adjacent aspartic residues or the 370SAQEpSQGNT378 nergic neuron predominantly expresses A2ARs and D2Rs, the GABAergic domain which contains a phosphorylable serine residue (S374), and dynorphynergic neuron expresses A1Rs and D1Rs [18,67,91]. Currently, thus forming electrostatic bonds of covalent-like strength [100,101]. F. Ciruela et al. / Biochimica et Biophysica Acta 1808 (2011) 1245–1255 1251

Interestingly, we have recently demonstrated that the point mutation of this receptor complex would assemble with receptor tyrosine kinases the serine 374 to alanine (S374A) reduces A2AR/D2R heteromerization or non-receptor tyrosine kinase Src, leading to ERK activation [113– and blocks the A2AR allosteric modulation of the dopamine D2R [102],a 117]. Altogether, it has been then suggested that the A2AR/mGlu5 result that has been further confirmed [103]. Also, when the S374A point receptor heteromeric complex may be involved in striatal neuron substitution was accompanied with the mutation of the two negatively plasticity (e.g. long-term potentiation and long-term depression) charged aspartates in the A2AR C-terminal tail (D401A/D402A, see [111,112]. In this way, it has been suggested the possibility of a above) a synergistic reduction in the physical A2AR/D2R interaction was synergic interaction within the A2AR/mGlu5 receptor oligomeric observed and in the loss of antagonistic allosteric modulation over the complex modulating mGlu5 receptor desensitization, which has A2AR/D2Rinterface[104]. Thus, these results further corroborate the been demonstrated for NMDA/mGlu5 receptor and group II mGlu/ existence of an electrostatic interaction between the C-terminal tail of mGlu5 receptor interactions [118,119]. However, this interaction the A2AR and the 3IL of the D2R. In addition, by using synthetic would be dependent on a synergistic interaction at the second- 2+ transmembrane (TM) α-helix peptides of the D2Rwehaverecently messenger level (Ca mobilization) [118,119], a situation that has explored the role of TM helix interactions within the A2AR/D2R not been possible to demonstrate in A2AR/mGlu5 receptor cotrans- heteromer interface [104]. Thus, we have evidence that the TM domains fected HEK-293 cells [41].

IV and V of the D2R play a critical role in the A2AR/D2Rheteromer Similar to the data obtained in cultured cells, studies in striatal interface since the incubation with peptides corresponding to these slices have shown that stimulation of the mGlu5 receptor potentiates domains significantly reduced the ability of the A2ARandtheD2Rto A2AR signalling in a MAPK-dependent manner [95]. Interestingly, heteromerize. Also, the incubation with TM-IV or TM-V blocked the MAPK activation is involved in the recruitment of ionotropic allosteric modulation normally found in the A2AR/D2R heteromer [104]. glutamate AMPA receptors to the postsynaptic density [120,121], Overall, the A2AR double aspartate mutation (D401A/D402A) together thus the A2AR/mGlu5 receptor heteromer would be able to modulate with the serine mutation (S374A) had deleterious consequences for the plastic changes in the striatal spine module (SSM, see below). In fact, allosteric communication within the A2AR/D2R heteromer, thus pointing it has been demonstrated that the pharmacological or genetic toward the relevance of these residues in the formation of an inactivation of A2ARs or mGlu5 receptors impairs corticostriatal electrostatic zip between the A2ARandtheD2R, which is regulated by long-term potentiation [122,123]. The A2AR/mGlu5 receptor syner- phosphorylation events, and thus necessary for the direct physical gism has also been demonstrated in the rat striatum, mediating c-fos receptor–receptor cross-talk. expression and thus counteracting phencyclidine induced motor activity [41]. Interestingly, it is well known that this motor activity is

4.2. A2AR and metabotropic glutamate type 5 (mGlu5) receptor oligomers dependent on dopamine D2R activity, thus it has been suggested, as commented above, that upon A2AR/mGlu5 receptor co-stimulation it In some behavioral animal models the treatment with mGlu5 is blocked D2R mediated transmission at the behavioral level. receptor agonists and antagonists produce similar effects to that Similarly, chronic but not acute treatment with a mGlu5 receptor described for the treatment with A2AR agonists and antagonists, antagonist reversed the akinetic deficit in a model of Parkinson's respectively, namely the selective modulation of D2R-mediated disease [124]. In this case, it would seem that the potentiation of D2R effects. Briefly, while a selective mGlu5 receptor agonist mainly activity could in part be caused by the internalisation and down- inhibits the D2R agonists-mediated motor activation [105], a mGlu5 regulation of the A2AR/mGlu5 receptor heteromeric receptor complex, receptor antagonist counteract the effects of D2R antagonists [106]. thus removing the D2R from inhibition of its signalling. In addition, it Thus, A2AR and mGlu5 receptor agonists and A2AR and mGlu5 receptor has also been shown that an acute effect of mGlu5 receptors on antagonists showed synergistic effects at the behavioral level dopamine D2R activity exists, since a mGlu5 receptor antagonist [105,107,108]. In addition, in the striopallidal complex the mGlu5 induced counteraction of the blockade with haloperidol of the D2R, receptor showed a very similar localization to that described for the thus observing in rats reduced rigidity and catalepsy [106], effects that

A2AR [52,109]. Also, the mGlu5 receptor immunoreactivity was may in part be exerted at the network level (e.g. at the level of the commonly found perisynaptically to asymmetric postsynaptic synap- subthalamic–nigral glutamate system). ses [109,110]. Overall, these studies provided behavioral and Overall, A2AR/mGlu5 receptor and also D2R/A2AR/mGlu5 receptor morphological basis for the possible existence of functional interac- (see below) interactions provide the rationale for the application of tions between striatal A2ARs and mGlu5 receptors. A2AR antagonists and the possible application of mGlu5 receptor Interestingly, it has been possible to demonstrate the existence of antagonists in Parkinson's disease [67,106,107,125,126]. heteromeric receptor complexes between A2ARs and mGlu5 receptors in both living cells (i.e. HEK-293 cells) and in native tissue 5. The striatal spine module as a paradigm of ARs integrative preparations (i.e. rat striatal membranes) [41]. Thus, it was initially functioning in the brain found that A2ARs and mGlu5 receptors were co-distributed at the membrane surface of co-transfected HEK-293 cells and also that they The striatum receives the densest dopamine innervation and were co-immunoprecipitated from cell extracts when specific anti- contains the highest concentration of dopamine receptors in the brain bodies were used [41]. Also, a strong A2AR and mGlu5 receptor co- [127,128]. As a consequence, dopamine plays a key role in motor distribution in rat striatal primary cultures was shown [40]. On the activity and goal-directed behaviors and also in the pathophysiology other hand, a number of studies have focused on the functionality of of diverse disorders, including Parkinson's disease and drug addiction the A2AR/mGlu5 receptor heteromeric complex, and it has been [39]. Moreover, in this brain area, apart of the pivotal role played by suggested that the former oligomer may play a major role on striatal dopamine, there are other neurotransmitters/neuromodulators that neuronal function and dysfunction. Accordingly, functional A2AR/ participate in the proper striatal functioning, for instance glutamate, mGlu5 receptor synergistic interactions have been demonstrated both acetylcholine, GABA, adenosine, etc., Therefore, interactions among in co-transfected cells and in the rat striatum. Thus, in HEK-293 cells it striatal neurotransmitters/neuromodulators determines the types of was shown that upon A2AR/mGlu5 receptor co-activation a synergistic plasticity taking place at the glutamatergic synapses onto striatal interaction occurred at the level of extracellular signal-regulated neurons, and thus complex interactions between the receptors for kinase 1/2 phosphorylation (ERK) and of c-fos expression [41]. these neurotransmitters/neuromodulators are currently established. Interestingly, this synergism was shown to occur at the level of the Interestingly, we have demonstrated the existence of oligomeric mitogen-activated protein kinase (MAPK) cascade [111,112]; alter- complexes that simultaneously contain adenosine, dopamine and natively, it could also happen that upon co-activation of the oligomer, glutamate receptors (e.g., A2AR/D2R/mGlu5 receptor) and that these 1252 F. Ciruela et al. / Biochimica et Biophysica Acta 1808 (2011) 1245–1255 aggregates might be relevant for the proper striatal function [129]. signalling mediated by single stimulation of any receptor within the Indeed, the recent fluorescence-based approaches on the study of oligomer may be, from a qualitative and/or quantitative point of view, protein-protein interactions [25] have allowed us to demonstrate the different from the one obtained upon the multiple stimulation of the existence of higher-order A2AR/D2R/mGlu5 receptor oligomers or oligomer, as a consequence in the CNS the oligomer may function as a RMs. Initially, by using bimolecular fluorescence complementation computational device that modulates information flow between

(BiFC), we visualized for the first time the occurrence of D2R/mGlu5 neurons. In this way, the adenosine receptors in general and the receptor heterodimers in living cells [129]. Furthermore, the combi- oligomers containing these receptors in particular constitute a clear nation of BiFC and BRET techniques allowed us to detect the existence example of integration of different modes of communication (i.e. of receptor oligomers containing more than two protomers, namely wiring transmission vs volume transmission) mediated by neuro-

A2AR/D2R/mGlu5 receptor higher-order oligomers or RMs [129]. Next, transmitters and neuromodulators (i.e. dopamine, glutamate, acet- by using triple-labeling post-embedding immunogold and detection hylcholine and adenosine). Consequently, the challenge now would at electron microscopic level the precise simultaneous distribution of be to fully characterize these adenosine containing oligomers in

A2AR, D2RandmGlu5 receptor in striatal neurons have been native tissue in order to determine how these oligomers impinge into performed. It is important to mention here that these three receptors neuronal functioning in both normal and pathological conditions. co-distributed in post-synaptic structures along the extra-synaptic Regarding this last issue, several concepts should be taken into and peri-synaptic plasma membrane of spines, establishing asym- account to fully understand the role of these oligomers: the metrical, putative glutamatergic, synapses with axon terminals [129] differential spatio-temporal expression pattern of each independent

(Fig. 2, inset panel). Overall, the A2AR/D2R/mGlu5 receptor oligomeric receptor within the oligomer, the stoichiometry and relative affinity of complex is located adjacent to the glutamatergic synapse of the these receptors within the RMs, and the functional and molecular dendritic spine of the enkephalin MSN, and their cross-talk within the cross-talk between the different receptors of a named oligomer. receptor heteromers might help in theory to modulate postsynaptic Overall, the knowledge of these issues will be necessary before the plastic changes at the glutamatergic synapse [42,43] (Fig. 2). design of any adenosine receptor oligomer-based therapeutic

The A2AR and group I glutamate metabotropic (mGlu) receptor strategy. agonists could synergistically reduce affinity of striatal D2R. Therefore, in membrane preparations from rat striatum, the stimulation of either Acknowledgements the A2AR or the mGlu5 receptor produces a decrease in the affinity of the D2R for agonists and a decrease in D2R agonist-mediated motor This work was supported by grants SAF2008-01462 and Consolider- activation [105,125]. In addition, co-stimulation of A2AR and mGlu5 Ingenio CSD2008-00005 from Ministerio de Ciencia e Innovación and receptor produces a synergistic antagonistic modulation of D2R ICREA Academia-2010 from the Catalan Institution for Research and binding and function that is significantly stronger than the reduction Advanced Studies to FC. FC, VFD and MGS belong to the “Neuropharma- induced by stimulation of either receptor alone [105]. Thus, co- cology and Pain” accredited research group (Generalitat de Catalunya, administration of A2AR and mGlu5 receptor agonists inhibits motor 2009 SGR 232). activation induced by D2R agonists [41,105]. Therefore, it is postulated that the formation of A2AR/D2R/mGlu5 receptor oligomer is a key step References for the correct shape of the SSM as a functional local module in the [1] A.M. Sebastiao, J.A. Ribeiro, Fine-tuning neuromodulation by adenosine, Trends striatum (Fig. 2). Interestingly, the SSM, which is constituted by the Pharmacol. Sci. 21 (2000) 341–346. dendritic spine of the MSN, its glutamatergic and dopaminergic [2] B.B. Fredholm, J.F. Chen, R.A. Cunha, P. Svenningsson, J.M. Vaugeois, Adenosine terminals and astroglial processes, could be considered the minimum and brain function, Int. Rev. Neurobiol. 63 (2005) 191–270. [3] K.A. Jacobson, Z.G. Gao, Adenosine receptors as therapeutic targets, Nat. Rev. portion that operates as an integrative independent unit in the Drug Discov. 5 (2006) 247–264. striatum and it provides an example of the extraordinary degree of [4] M.P. Abbracchio, G. Burnstock, A. Verkhratsky, H. Zimmermann, Purinergic computation that takes place in local modules (Fig. 2) [42,43]. Indeed, signalling in the nervous system: an overview, Trends Neurosci. 32 (2009) 19–29. the formation of glutamate, dopamine and adenosine receptors [5] B.B. Fredholm, M.P. Abbracchio, G. Burnstock, J.W. Daly, T.K. Harden, K.A. Jacobson, P. Leff, M. Williams, Nomenclature and classification of purinoceptors, oligomers in the striatum are behind the SSM performance as a Pharmacol. Rev. 46 (1994) 143–156. local module, thus the coordinate operation of these oligomers within [6] M.E. Olah, G.L. Stiles, Adenosine receptor subtypes: characterization and – the SSM is associated with particular elaborated functions in the local therapeutic regulation, Annu. Rev. Pharmacol. Toxicol. 35 (1995) 581 606. [7] T.V. Dunwiddie, S.A. Masino, The role and regulation of adenosine in the central module.IntheSSM,theA2AR/D2R/mGlu5 receptor oligomeric nervous system, Annu. Rev. Neurosci. 24 (2001) 31–55. complex is located extrasynaptically and adjacent to the glutamater- [8] T.M. Palmer, G.L. Stiles, Adenosine receptors, Neuropharmacology 34 (1995) gic synapse of the dendritic spine of enkephalin MSNs, where they are 683–694. [9] B.B. Fredholm, Purinoceptors in the nervous system, Pharmacol. Toxicol. 76 activated by volume transmission and their cross-talk within the (1995) 228–239. complex helps to modulate postsynaptic plastic changes at the [10] R.B. Marala, S.J. Mustafa, Direct evidence for the coupling of A2-adenosine receptor to stimulatory -binding-protein in bovine brain glutamatergic synapse. On the other hand, the A1R/A2AR heteromeric striatum, J. Pharmacol. Exp. Ther. 266 (1993) 294–300. complex is found in glutamatergic terminals and the molecular cross- [11] S. Ryzhov, A.E. Goldstein, I. Biaggioni, I. Feoktistov, Cross-talk between Gs- and talk between the two receptors in the heteromer helps to modulate Gq-coupled pathways in regulation of interleukin-4 by A2B adenosine receptors glutamate release (Fig. 2). Overall, the formation of oligomeric in human mast cells, Mol. Pharmacol. 70 (2006) 727–735. [12] I. Feoktistov, I. Biaggioni, Adenosine A2B receptors, Pharmacol. Rev. 49 (1997) complexes in the SSM containing adenosine receptors allows a 381–402. much more elaborated tuning in the regulation of both presynaptic [13] C.A. Salvatore, M.A. Jacobson, H.E. Taylor, J. Linden, R.G. Johnson, Molecular and postsynaptic neuronal responses in the striatal local circuitry. cloning and characterization of the human A3 adenosine receptor, Proc. Natl Acad. Sci. USA 90 (1993) 10365–10369.

[14] F. Pedata, A.M. Pugliese, A.M. Sebastião, J.A. Ribeiro, Adenosine A3 receptor 6. Concluding remarks signaling in the central nervous system, in: P.A. Borea (Ed.), A3 Adenosine Receptors from Cell Biology to Pharmacology and Therapeutics, vol. 1, Springer – The existence of GPCR oligomers is now broadly accepted and the Science +Business Media B.V, Amsterdam, 2010, pp. 165 188. [15] F. Ciruela, C. Albergaria, A. Soriano, L. Cuffi, L. Carbonell, S. Sanchez, J. Gandia, V. functional meaning of receptor oligomerization is becoming revealed Fernandez-Duenas, Adenosine receptors interacting proteins (ARIPs): behind (http://data.gpcr-okb.org/gpcr-okb/). In the CNS the oligomerization the biology of adenosine signaling, Biochim. Biophys. Acta 1798 (2010) 9–20. of neurotransmitter/neuromodulator receptors confers functional [16] L.F. Agnati, K. Fuxe, I. Zini, P. Lenzi, T. Hokfelt, Aspects on receptor regulation and isoreceptor identification, Med. Biol. 58 (1980) 182–187. entities which possess different biochemical characteristics with [17] K. Fuxe, L.F. Agnati, F. Benfenati, M. Celani, I. Zini, M. Zoli, V. Mutt, Evidence for respect to the individual components of the heteromer. Thus, the the existence of receptor–receptor interactions in the central nervous system. F. Ciruela et al. / Biochimica et Biophysica Acta 1808 (2011) 1245–1255 1253

Studies on the regulation of monoamine receptors by neuropeptides, J. Neural [41] S. Ferre, M. Karcz-Kubicha, B.T. Hope, P. Popoli, J. Burgueno, M.A. Gutierrez, V. Transm. Suppl. 18 (1983) 165–179. Casado, K. Fuxe, S.R. Goldberg, C. Lluis, R. Franco, F. Ciruela, Synergistic

[18] L.F. Agnati, S. Ferre, C. Lluis, R. Franco, K. Fuxe, Molecular mechanisms and interaction between adenosine A2A and glutamate mGlu5 receptors: implications therapeutical implications of intramembrane receptor/receptor interactions for striatal neuronal function, Proc. Natl Acad. Sci. USA 99 (2002) 11940–11945. among heptahelical receptors with examples from the striatopallidal GABA [42] S. Ferre, F. Ciruela, C. Quiroz, R. Lujan, P. Popoli, R.A. Cunha, L.F. Agnati, K. Fuxe, A.S. neurons, Pharmacol. Rev. 55 (2003) 509–550. Woods, C. Lluis, R. Franco, Adenosine receptor heteromers and their integrative role [19] S. Avissar, G. Amitai, M. Sokolovsky, Oligomeric structure of muscarinic receptors in striatal function, Sci. World J. 7 (2007) 74–85. is shown by photoaffinity labeling: subunit assembly may explain high- and [43] S. Ferre, L.F. Agnati, F. Ciruela, C. Lluis, A.S. Woods, K. Fuxe, R. Franco, low-affinity agonist states, Proc. Natl Acad. Sci. USA 80 (1983) 156–159. Neurotransmitter receptor heteromers and their integrative role in “local [20] C.M. Fraser, J.C. Venter, The size of the mammalian lung beta 2-adrenergic modules”: the striatal spine module, Brain Res. Rev. 55 (2007) 55–67. receptor as determined by target size analysis and immunoaffinity chromatog- [44] S. Ferre, F. Ciruela, A.S. Woods, C. Lluis, R. Franco, Functional relevance of raphy, Biochem. Biophys. Res. Commun. 109 (1982) 21–29. neurotransmitter receptor heteromers in the central nervous system, Trends [21] G.L. Peterson, L.C. Rosenbaum, D.J. Broderick, M.I. Schimerlik, Physical properties Neurosci. 30 (2007) 440–446. of the purified cardiac muscarinic , Biochemistry 25 [45] M. Canals, J.Burgueno, D. Marcellino, N. Cabello, E.I. Canela, J. Mallol, L. Agnati, S. Ferre,

(1986) 3189–3202. M.Bouvier,K.Fuxe,F.Ciruela,C.Lluis,R.Franco,HomodimerizationofadenosineA2A [22] J.T. Herberg, J. Codina, K.A. Rich, F.J. Rojas, R. Iyengar, The hepatic glucagon receptors: qualitative and quantitative assessment by fluorescence and biolumines- receptor. Solubilization, characterization, and development of an affinity cence energy transfer, J. Neurochem. 88 (2004) 726–734. adsorption assay for the soluble receptor, J. Biol. Chem. 259 (1984) 9285–9294. [46] N. Rebola, R.J. Rodrigues, L.V. Lopes, P.J. Richardson, C.R. Oliveira, R.A. Cunha,

[23] P. Zawarynski, T. Tallerico, P. Seeman, S.P. Lee, B.F. O'Dowd, S.R. George, Dopamine Adenosine A1 and A2A receptors are co-expressed in pyramidal neurons and co- D2 receptor dimers in human and rat brain, FEBS Lett. 441 (1998) 383–386. localized in glutamatergic nerve terminals of the rat hippocampus, Neuroscience [24] F. Ciruela, V. Casado, J. Mallol, E.I. Canela, C. Lluis, R. Franco, Immunological 133 (2005) 79–83.

identification of A1 adenosine receptors in brain cortex, J. Neurosci. Res. 42 [47] F. Ciruela, V. Casado, R.J. Rodrigues, R. Lujan, J. Burgueno, M. Canals, J. Borycz, N. (1995) 818–828. Rebola, S.R. Goldberg, J. Mallol, A. Cortes, E.I. Canela, J.F. Lopez-Gimenez, G. [25] F. Ciruela, J.P. Vilardaga, V. Fernandez-Duenas, Lighting up multiprotein Milligan, C. Lluis, R.A. Cunha, S. Ferre, R. Franco, Presynaptic control of striatal

complexes: lessons from GPCR oligomerization, Trends Biotechnol. 28 (2010) glutamatergic neurotransmission by adenosine A1–A2A receptor heteromers, 407–415. J. Neurosci. 26 (2006) 2080–2087. [26] P. Carriba, G. Navarro, F. Ciruela, S. Ferre, V. Casado, L. Agnati, A. Cortes, J. Mallol, [48] R.A. Cunha, E. Milusheva, E.S. Vizi, J.A. Ribeiro, A.M. Sebastiao, Excitatory and K. Fuxe, E.I. Canela, C. Lluis, R. Franco, Detection of heteromerization of more inhibitory effects of A1 and A2A adenosine receptor activation on the electrically than two proteins by sequential BRET-FRET, Nat. Meth. 5 (2008) 727–733. evoked [3H]acetylcholine release from different areas of the rat hippocampus, [27] J. Gandia, J. Galino, O.B. Amaral, A. Soriano, C. Lluis, R. Franco, F. Ciruela, Detection J. Neurochem. 63 (1994) 207–214. of higher-order G protein-coupled receptor oligomers by a combined BRET-BiFC [49] L.V. Lopes, R.A. Cunha, J.A. Ribeiro, Cross talk between A(1) and A(2A) adenosine technique, FEBS Lett. 582 (2008) 2979–2984. receptors in the hippocampus and cortex of young adult and old rats, [28] D.J. Dupre, M. Robitaille, N. Ethier, L.R. Villeneuve, A.M. Mamarbachi, T.E. Hebert, J. Neurophysiol. 82 (1999) 3196–3203.

Seven transmembrane receptor core signaling complexes are assembled prior to [50] L.V. Lopes, R.A. Cunha, B. Kull, B.B. Fredholm, J.A. Ribeiro, plasma membrane trafficking, J. Biol. Chem. 281 (2006) 34561–34573. facilitation of hippocampal synaptic transmission is dependent on tonic A1 [29] W. Guo, E. Urizar, M. Kralikova, J.C. Mobarec, L. Shi, M. Filizola, J.A. Javitch, receptor inhibition, Neuroscience 112 (2002) 319–329.

Dopamine D2 receptors form higher order oligomers at physiological expression [51] T. Ochiishi, L. Chen, A. Yukawa, Y. Saitoh, Y. Sekino, T. Arai, H. Nakata, H. levels, EMBO J. 27 (2008) 2293–2304. Miyamoto, Cellular localization of adenosine A1 receptors in rat forebrain: [30] J.F. Lopez-Gimenez, M. Canals, J.D. Pediani, G. Milligan, The alpha1b- immunohistochemical analysis using adenosine A1 receptor-specific monoclonal adrenoceptor exists as a higher-order oligomer: effective oligomerization is antibody, J. Comp. Neurol. 411 (1999) 301–316. required for receptor maturation, surface delivery, and function, Mol. [52] B.D. Hettinger, A. Lee, J. Linden, D.L. Rosin, Ultrastructural localization of

Pharmacol. 71 (2007) 1015–1029. adenosine A2A receptors suggests multiple cellular sites for modulation of [31] G. Navarro, P. Carriba, J. Gandia, F. Ciruela, V. Casado, A. Cortes, J. Mallol, E.I. GABAergic neurons in rat striatum, J. Comp. Neurol. 431 (2001) 331–346.

Canela, C. Lluis, R. Franco, Detection of heteromers formed by cannabinoid CB1, [53] L.G. Wu, P. Saggau, Presynaptic inhibition of elicited neurotransmitter release, dopamine D2, and adenosine A2A G protein-coupled receptors by combining Trends Neurosci. 20 (1997) 204–212. bimolecular fluorescence complementation and bioluminescence energy trans- [54] S. Ferre, J. Borycz, S.R. Goldberg, B.T. Hope, M. Morales, C. Lluis, R. Franco, F. fer, Sci. World J. 8 (2008) 1088–1097. Ciruela, R. Cunha, Role of adenosine in the control of homosynaptic plasticity in

[32] P.A. Vidi, J. Chen, J.M. Irudayaraj, V.J. Watts, Adenosine A2A receptors assemble striatal excitatory synapses, J. Integr. Neurosci. 4 (2005) 445–464. into higher-order oligomers at the plasma membrane, FEBS Lett. 582 (2008) [55] B.B. Fredholm, E. Irenius, B. Kull, G. Schulte, Comparison of the potency of 3985–3990. adenosine as an agonist at human adenosine receptors expressed in Chinese [33] L. Albizu, M. Cottet, M. Kralikova, S. Stoev, R. Seyer, I. Brabet, T. Roux, H. Bazin, E. hamster ovary cells, Biochem. Pharmacol. 61 (2001) 443–448. Bourrier, L. Lamarque, C. Breton, M.L. Rives, A. Newman, J. Javitch, E. Trinquet, M. [56] P.A. Pousinha, A.M. Correia, A.M. Sebastiao, J.A. Ribeiro, Predominance of Manning, J.P. Pin, B. Mouillac, T. Durroux, Time-resolved FRET between GPCR adenosine excitatory over inhibitory effects on transmission at the neuromus- ligands reveals oligomers in native tissues, Nat. Chem. Biol. 6 (2010) 587–594. cular junction of infant rats, J. Pharmacol. Exp. Ther. 332 (2010) 153–163. [34] M. Torvinen, S. Gines, J. Hillion, S. Latini, M. Canals, F. Ciruela, F. Bordoni, W. [57] P.S. Goldman-Rakic, The cortical dopamine system: role in memory and Staines, F. Pedata, L.F. Agnati, C. Lluis, R. Franco, S. Ferre, K. Fuxe, Interactions cognition, Adv. Pharmacol. 42 (1998) 707–711.

among adenosine deaminase, adenosine A1 receptors and dopamine D1 [58] W. Schultz, Getting formal with dopamine and reward, Neuron 36 (2002) receptors in stably cotransfected fibroblast cells and neurons, Neuroscience 241–263. 113 (2002) 709–719. [59] J. Yelnik, Functional anatomy of the basal ganglia, Mov. Disord. 17 (Suppl 3) [35] F. Ciruela, M. Escriche, J. Burgueno, E. Angulo, V. Casado, M.M. Soloviev, E.I. (2002) S15–S21. Canela, J. Mallol, W.Y. Chan, C. Lluis, R.A. McIlhinney, R. Franco, Metabotropic [60] N.D. Volkow, J.S. Fowler, G.J. Wang, R.Z. Goldstein, Role of dopamine, the frontal

glutamate 1alpha and adenosine A1 receptors assemble into functionally cortex and memory circuits in drug addiction: insight from imaging studies, interacting complexes, J. Biol. Chem. 276 (2001) 18345–18351. Neurobiol. Learn. Mem. 78 (2002) 610–624. [36] H. Nakata, K. Yoshioka, T. Kamiya, H. Tsuga, K. Oyanagi, Functions of heteromeric [61] A.F. Arnsten, Stress impairs prefrontal cortical function in rats and monkeys: role

association between adenosine and P2Y receptors, J. Mol. Neurosci. 26 (2005) of dopamine D1 and norepinephrine alpha-1 receptor mechanisms, Prog. Brain 233–238. Res. 126 (2000) 183–192. [37] J. Hillion, M. Canals, M. Torvinen, V. Casado, R. Scott, A. Terasmaa, A. Hansson, S. [62] N.R. Swerdlow, G.F. Koob, Dopamine, schizophrenia, mania, and depression: Watson, M.E. Olah, J. Mallol, E.I. Canela, M. Zoli, L.F. Agnati, C.F. Ibanez, C. Lluis, R. toward a unified hypothesis of cortico-striato-palido-thalamic function, Behav. Franco, S. Ferre, K. Fuxe, Coaggregation, cointernalization, and codesensitization Brain Sci. 10 (1987) 197–245.

of adenosine A2A receptors and dopamine D2 receptors, J. Biol. Chem. 277 (2002) [63] C. Missale, S.R. Nash, S.W. Robinson, M. Jaber, M.G. Caron, Dopamine receptors: 18091–18097. from structure to function, Physiol. Rev. 78 (1998) 189–225. [38] M. Canals, D. Marcellino, F. Fanelli, F. Ciruela, P. de Benedetti, S.R. Goldberg, K. [64] K. Fuxe, U. Ungerstedt, Action of caffeine and theophyllamine on supersensitive Neve, K. Fuxe, L.F. Agnati, A.S. Woods, S. Ferre, C. Lluis, M. Bouvier, R. Franco, dopamine receptors: considerable enhancement of receptor response to

Adenosine A2A-dopamine D2 receptor–receptor heteromerization: qualitative treatment with DOPA and dopamine receptor agonists, Med. Biol. 52 (1974) and quantitative assessment by fluorescence and bioluminescence energy 48–54. transfer, J. Biol. Chem. 278 (2003) 46741–46749. [65] P. Popoli, L. Gimenez-Llort, A. Pezzola, R. Reggio, E. Martinez, K. Fuxe, S. Ferre,

[39] S. Ferre, F. Ciruela, M. Canals, D. Marcellino, J. Burgueno, V. Casado, J. Hillion, M. Adenosine A1 receptor blockade selectively potentiates the motor effects Torvinen, F. Fanelli, P. Benedetti Pd, S.R. Goldberg, M. Bouvier, K. Fuxe, L.F. Agnati, induced by dopamine D1 receptor stimulation in rodents, Neurosci. Lett. 218 C. Lluis, R. Franco, A. Woods, Adenosine A2A-dopamine D2 receptor–receptor (1996) 209–213. heteromers. Targets for neuro-psychiatric disorders, Parkinsonism Relat. Disord. [66] R. Rimondini, S. Ferre, L. Gimenez-Llort, S.O. Ogren, K. Fuxe, Differential effects

10 (2004) 265–271. of selective adenosine A1 and A2A receptor agonists on dopamine receptor [40] K. Fuxe, L.F. Agnati, K. Jacobsen, J. Hillion, M. Canals, M. Torvinen, B. Tinner- agonist-induced behavioural responses in rats, Eur. J. Pharmacol. 347 (1998) Staines, W. Staines, D. Rosin, A. Terasmaa, P. Popoli, G. Leo, V. Vergoni, C. Lluis, F. 153–158.

Ciruela, R. Franco, S. Ferre, Receptor heteromerization in adenosine A2A receptor [67] S. Ferre, B.B. Fredholm, M. Morelli, P. Popoli, K. Fuxe, Adenosine-dopamine signaling: relevance for striatal function and Parkinson's disease, Neurology 61 receptor–receptor interactions as an integrative mechanism in the basal ganglia, (2003) S19–S23. Trends Neurosci. 20 (1997) 482–487. 1254 F. Ciruela et al. / Biochimica et Biophysica Acta 1808 (2011) 1245–1255

[68] K. Fuxe, S. Ferre, M. Zoli, L.F. Agnati, Integrated events in central dopamine [96] A. Tozzi, A. Tscherter, V. Belcastro, M. Tantucci, C. Costa, B. Picconi, D. Centonze, P.

transmission as analyzed at multiple levels. Evidence for intramembrane Calabresi, F. Borsini, Interaction of A2A adenosine and D2 dopamine receptors adenosine A2A/dopamine D2 and adenosine A1/dopamine D1 receptor interac- modulates corticostriatal glutamatergic transmission, Neuropharmacology 53 tions in the basal ganglia, Brain Res. Brain Res. Rev. 26 (1998) 258–273. (2007) 783–789.

[69] S. Gines, J. Hillion, M. Torvinen, S. Le Crom, V. Casado, E.I. Canela, S. Rondin, J.Y. [97] T. Kamiya, O. Saitoh, K. Yoshioka, H. Nakata, Oligomerization of adenosine A2A Lew, S. Watson, M. Zoli, L.F. Agnati, P. Verniera, C. Lluis, S. Ferre, K. Fuxe, R. and dopamine D2 receptors in living cells, Biochem. Biophys. Res. Commun. 306 Franco, Dopamine D1 and adenosine A1 receptors form functionally interacting (2003) 544–549. heteromeric complexes, Proc. Natl Acad. Sci. USA 97 (2000) 8606–8611. [98] S. Genedani, D. Guidolin, G. Leo, M. Filaferro, M. Torvinen, A.S. Woods, K. Fuxe, S. [70] S. Toda, L.F. Alguacil, P.W. Kalivas, Repeated cocaine administration changes the Ferre, L.F. Agnati, Computer-assisted image analysis of caveolin-1 involvement in

function and subcellular distribution of adenosine A1 receptor in the rat nucleus the internalization process of adenosine A2A-dopamine D2 receptor hetero- accumbens, J. Neurochem. 87 (2003) 1478–1484. dimers, J. Mol. Neurosci. 26 (2005) 177–184. [71] S. Ferre, P. Popoli, L. Gimenez-Llort, U.B. Finnman, E. Martinez, A. Scotti de [99] L.F. Agnati, K. Fuxe, M. Torvinen, S. Genedani, R. Franco, S. Watson, G.G. Nussdorfer, G.

Carolis, K. Fuxe, Postsynaptic antagonistic interaction between adenosine A1 and Leo, D. Guidolin, New methods to evaluate colocalization of fluorophores in dopamine D1 receptors, NeuroReport 6 (1994) 73–76. immunocytochemical preparations as exemplified by a study on A2A and D2 receptors [72] S. Ferre, M. Torvinen, K. Antoniou, E. Irenius, O. Civelli, E. Arenas, B.B. Fredholm, in Chinese hamster ovary cells, J. Histochem. Cytochem. 53 (2005) 941–953.

K. Fuxe, Adenosine A1 receptor-mediated modulation of dopamine D1 receptors [100] A.S. Woods, S. Ferre, Amazing stability of the arginine-phosphate electrostatic in stably cotransfected fibroblast cells, J. Biol. Chem. 273 (1998) 4718–4724. interaction, J. Proteome Res. 4 (2005) 1397–1402.

[73] S. Le Crom, D. Prou, P. Vernier, Autocrine activation of adenosine A1 receptors [101] A.S. Woods, F. Ciruela, K. Fuxe, L.F. Agnati, C. Lluis, R. Franco, S. Ferre, Role of blocks D1A but not D1B dopamine receptor desensitization, J. Neurochem. 82 electrostatic interaction in receptor–receptor heteromerization, J. Mol. Neurosci. (2002) 1549–1552. 26 (2005) 125–132.

[74] Y. Cao, W.C. Sun, L. Jin, K.Q. Xie, X.Z. Zhu, Activation of adenosine A1 receptor [102] D.O. Borroto-Escuela, D. Marcellino, M. Narvaez, M. Flajolet, N. Heintz, L. Agnati, modulates dopamine D1 receptor activity in stably cotransfected human F. Ciruela, K. Fuxe, A serine point mutation in the adenosine A2AR C-terminal tail embryonic kidney 293 cells, Eur. J. Pharmacol. 548 (2006) 29–35. reduces receptor heteromerization and allosteric modulation of the dopamine

[75] R. Franco, C. Lluis, E.I. Canela, J. Mallol, L. Agnati, V. Casado, F. Ciruela, S. Ferre, K. D2R, Biochem. Biophys. Res. Commun. 394 (2010) 222–227. Fuxe, Receptor–receptor interactions involving adenosine A1 or dopamine D1 [103] G. Navarro, S. Ferre, A. Cordomi, E. Moreno, J. Mallol, V. Casado, A. Cortes, H. receptors and accessory proteins, J. Neural Transm. 114 (2007) 93–104. Hoffmann, J. Ortiz, E.I. Canela, C. Lluis, L. Pardo, R. Franco, A.S. Woods, Interactions [76] R.J. Lefkowitz, The superfamily of heptahelical receptors, Nat. Cell Biol. 2 (2000) between intracellular domains as key determinants of the quaternary structure and E133–E136. function of receptor heteromers, J. Biol. Chem. 285 (2010) 27346–27359. [77] P.H. McDonald, R.J. Lefkowitz, Beta-Arrestins: new roles in regulating heptahe- [104] D.O. Borroto-Escuela, W. Romero-Fernandez, A.O. Tarakanov, M. Gomez-Soler, F. lical receptors' functions, Cell. Signal. 13 (2001) 683–689. Corrales, D. Marcellino, M. Narvaez, M. Frankowska, M. Flajolet, N. Heintz, L.F.

[78] K. Fuxe, S. Ferre, S. Genedani, R. Franco, L.F. Agnati, Adenosine receptor– Agnati, F. Ciruela, K. Fuxe, Characterization of the A2AR-D2R interface: focus on dopamine receptor interactions in the basal ganglia and their relevance for brain the role of the C-terminal tail and the transmembrane helices, Biochem. Biophys. function, Physiol. Behav. 92 (2007) 210–217. Res. Commun. (In press) (2010). [79] J.P. Pin, R. Duvoisin, The metabotropic glutamate receptors: structure and [105] P. Popoli, A. Pezzola, M. Torvinen, R. Reggio, A. Pintor, L. Scarchilli, K. Fuxe, S.

functions, Neuropharmacology 34 (1995) 1–26. Ferre, The selective mGlu5 receptor agonist CHPG inhibits quinpirole-induced [80] M. Hollmann, S. Heinemann, Cloned glutamate receptors, Annu. Rev. Neurosci. turning in 6-hydroxydopamine-lesioned rats and modulates the binding

17 (1994) 31–108. characteristics of dopamine D2 receptors in the rat striatum: interactions with [81] J.P. Pin, C. Waeber, L. Prezeau, J. Bockaert, S.F. Heinemann, Alternative splicing adenosine A2A receptors, Neuropsychopharmacology 25 (2001) 505–513. generates metabotropic glutamate receptors inducing different patterns of [106] K. Ossowska, J. Konieczny, S. Wolfarth, J. Wieronska, A. Pilc, Blockade of the calcium release in Xenopus oocytes, Proc. Natl Acad. Sci. USA 89 (1992) metabotropic glutamate receptor subtype 5 (mGluR5) produces antiparkinso- 10331–10335. nian-like effects in rats, Neuropharmacology 41 (2001) 413–420. [82] P. Grandes, J.M. Mateos, D. Ruegg, R. Kuhn, T. Knopfel, Differential cellular [107] A. Kachroo, L.R. Orlando, D.K. Grandy, J.F. Chen, A.B. Young, M.A. Schwarzschild,

localization of three splice variants of the mGluR1 metabotropic glutamate Interactions between metabotropic glutamate 5 and adenosine A2A receptors in receptor in rat cerebellum, NeuroReport 5 (1994) 2249–2252. normal and parkinsonian mice, J. Neurosci. 25 (2005) 10414–10419.

[83] F. Ciruela, M.J. Robbins, A.C. Willis, R.A. McIlhinney, Interactions of the C [108] R. Coccurello, N. Breysse, M. Amalric, Simultaneous blockade of adenosine A2A terminus of metabotropic glutamate receptor type 1alpha with rat brain and metabotropic glutamate mGlu5 receptors increase their efficacy in reversing proteins: evidence for a direct interaction with tubulin, J. Neurochem. 72 Parkinsonian deficits in rats, Neuropsychopharmacology 29 (2004) 1451–1461. (1999) 346–354. [109] Y. Smith, A. Charara, J.E. Hanson, M. Paquet, A.I. Levey, GABA(B) and group I [84] F. Ciruela, R.A. McIlhinney, Metabotropic glutamate receptor type 1alpha and metabotropic glutamate receptors in the striatopallidal complex in primates, J. tubulin assemble into dynamic interacting complexes, J. Neurochem. 76 (2001) Anat. 196 (Pt 4) (2000) 555–576. 750–757. [110] R. Lujan, Z. Nusser, J.D. Roberts, R. Shigemoto, P. Somogyi, Perisynaptic location [85] D. Lu, H. Yan, T. Othman, C.P. Turner, T. Woolf, S.A. Rivkees, Cytoskeletal protein of metabotropic glutamate receptors mGluR1 and mGluR5 on dendrites and 4.1G binds to the third intracellular loop of the A1 adenosine receptor and dendritic spines in the rat hippocampus, Eur. J. Neurosci. 8 (1996) 1488–1500. inhibits receptor action, Biochem. J. 377 (2004) 51–59. [111] J.D. Sweatt, The neuronal MAP kinase cascade: a biochemical signal integration [86] T.V. Dunwiddie, The physiological role of adenosine in the central nervous system subserving synaptic plasticity and memory, J. Neurochem. 76 (2001) 1–10. system, Int. Rev. Neurobiol. 27 (1985) 63–139. [112] S. Ferre, W.T. O'Connor, K. Fuxe, U. Ungerstedt, The striopallidal neuron: a main locus [87] H. Yawo, N. Chuhma, Preferential inhibition of omega-conotoxin-sensitive for adenosine-dopamine interactions in the brain, J. Neurosci. 13 (1993) 5402–5406. presynaptic Ca2+ channels by adenosine autoreceptors, Nature 365 (1993) [113] S. Maudsley, K.L. Pierce, A.M. Zamah, W.E. Miller, S. Ahn, Y. Daaka, R.J. Lefkowitz,

256–258. L.M. Luttrell, The β2- mediates extracellular signal-regulated [88] M. Kimura, N. Saitoh, T. Takahashi, Adenosine A1 receptor-mediated presynaptic kinase activation via assembly of a multi-receptor complex with the epidermal inhibition at the calyx of Held of immature rats, J. Physiol. 553 (2003) 415–426. growth factor receptor, J. Biol. Chem. 275 (2000) 9572–9580. [89] K.A. Moore, R.A. Nicoll, D. Schmitz, Adenosine gates synaptic plasticity at [114] L.M. Luttrell, F.L. Roudabush, E.W. Choy, W.E. Miller, M.E. Field, K.L. Pierce, R.J. hippocampal mossy fiber synapses, Proc. Natl Acad. Sci. USA 100 (2003) Lefkowitz, Activation and targeting of extracellular signal-regulated kinases by 14397–14402. beta-arrestin scaffolds, Proc. Natl Acad. Sci. USA 98 (2001) 2449–2454. 2+ [90] N.J. Toms, P.J. Roberts, Group 1 mGlu receptors elevate [Ca ]i in rat cultured [115] G. Schulte, B.B. Fredholm, Human adenosine A1,A2A,A2B, and A3 receptors 2+ cortical type 2 astrocytes: [Ca ]i synergy with adenosine A1 receptors, expressed in Chinese hamster ovary cells all mediate the phosphorylation of Neuropharmacology 38 (1999) 1511–1517. extracellular-regulated kinase 1/2, Mol. Pharmacol. 58 (2000) 477–482.

[91] S.N. Schiffmann, G. Fisone, R. Moresco, R.A. Cunha, S. Ferre, Adenosine A2A [116] M.J. Marinissen, J.S. Gutkind, G protein-coupled receptors and signaling receptors and basal ganglia physiology, Prog. Neurobiol. 83 (2007) 277–292. networks: emerging paradigms, Trends Pharmacol. Sci. 22 (2001) 368–376. [92] F. Ciruela, J. Burgueno, V. Casado, M. Canals, D. Marcellino, S.R. Goldberg, M. [117] R.A. Hall, R.T. Premont, R.J. Lefkowitz, Heptahelical receptor signaling: beyond Bader, K. Fuxe, L.F. Agnati, C. Lluis, R. Franco, S. Ferre, A.S. Woods, Combining the G protein paradigm, J. Cell Biol. 145 (1999) 927–932. mass spectrometry and pull-down techniques for the study of receptor [118] K. Cho, Z.I. Bashir, Cooperation between mglu receptors: a depressing heteromerization. Direct epitope-epitope electrostatic interactions between mechanism? Trends Neurosci. 25 (2002) 405–411.

adenosine A2A and dopamine D2 receptors, Anal. Chem. 76 (2004) 5354–5363. [119] S. Alagarsamy, M.J. Marino, S.T. Rouse, R.W. Gereau 4th, S.F. Heinemann, P.J. [93] T. Shindou, H. Nonaka, P.J. Richardson, A. Mori, H. Kase, M. Ichimura, Presynaptic Conn, Activation of NMDA receptors reverses desensitization of mGluR5 in

adenosine A2A receptors enhance GABAergic synaptic transmission via a cyclic native and recombinant systems, Nat. Neurosci. 2 (1999) 234–240. AMP dependent mechanism in the rat globus pallidus, Br. J. Pharmacol. 136 [120] I. Song, R.L. Huganir, Regulation of AMPA receptors during synaptic plasticity, (2002) 296–302. Trends Neurosci. 25 (2002) 578–588. [94] P. Svenningsson, M. Lindskog, C. Ledent, M. Parmentier, P. Greengard, B.B. [121] G.M. Thomas, R.L. Huganir, MAPK cascade signalling and synaptic plasticity, Nat. Fredholm, G. Fisone, Regulation of the phosphorylation of the dopamine- and Rev. Neurosci. 5 (2004) 173–183.

cAMP-regulated phosphoprotein of 32 kDa in vivo by dopamine D1, dopamine [122] P. d'Alcantara, C. Ledent, S. Swillens, S.N. Schiffmann, Inactivation of adenosine D2,andadenosineA2A receptors, Proc. Natl Acad. Sci. USA 97 (2000) A2A receptor impairs long term potentiation in the accumbens nucleus without 1856–1860. altering basal synaptic transmission, Neuroscience 107 (2001) 455–464. [95] A. Nishi, F. Liu, S. Matsuyama, M. Hamada, H. Higashi, A.C. Nairn, P. Greengard, [123] P. Gubellini, E. Saulle, D. Centonze, C. Costa, D. Tropepi, G. Bernardi, F. Conquet, P.

Metabotropic mGlu5 receptors regulate adenosine A2A receptor signaling, Proc. Calabresi, Corticostriatal LTP requires combined mGluR1 and mGluR5 activation, Natl Acad. Sci. USA 100 (2003) 1322–1327. Neuropharmacology 44 (2003) 8–16. F. Ciruela et al. / Biochimica et Biophysica Acta 1808 (2011) 1245–1255 1255

[124] N. Breysse, C. Baunez, W. Spooren, F. Gasparini, M. Amalric, Chronic but not [128] C.R. Gerfen, Basal ganglia, in: G. Paxinos (Ed.), the Rat Nervous System, Elsevier acute treatment with a metabotropic glutamate 5 receptor antagonist reverses Academic Press, Amsterdam, 2004, pp. 445–508. the akinetic deficits in a rat model of parkinsonism, J. Neurosci. 22 (2002) [129] N. Cabello, J. Gandia, D.C. Bertarelli, M. Watanabe, C. Lluis, R. Franco, S. Ferre, R.

5669–5678. Lujan, F. Ciruela, Metabotropic glutamate type 5, dopamine D2 and adenosine [125] S. Ferre, G. von Euler, B. Johansson, B.B. Fredholm, K. Fuxe, Stimulation of high- A2A receptors form higher-order oligomers in living cells, J. Neurochem. 109 affinity adenosine A2 receptors decreases the affinity of dopamine D2 receptors (2009) 1497–1507. in rat striatal membranes, Proc. Natl Acad. Sci. USA 88 (1991) 7238–7241. [130] L. Canela, R. Lujan, C. Lluis, J. Burgueno, J. Mallol, E.I. Canela, R. Franco, F. Ciruela, 2+ [126] P. Jenner, , a novel adenosine A2A receptor antagonist, for the The neuronal Ca -binding protein 2 (NECAB2) interacts with the adenosine A2A treatment of Parkinson's disease, Expert Opin. Investig. Drugs 14 (2005) receptor and modulates the cell surface expression and function of the receptor, 729–738. Mol. Cell. Neurosci. 36 (2007) 1–12. [127] O. Lindvall, A. Bjorklund, Anatomy of the dopaminergic neuron systems in the rat [131] D. Boison, J.F. Chen, B.B. Fredholm, Adenosine signaling and function in glial cells, brain, Adv. Biochem. Psychopharmacol. 19 (1978) 1–23. Cell Death Differ. 17 (2010) 1071–1082.