Abstract
The variety of physiological functions controlled by dopamine in the brain and periphery is mediated by the D1, D2, D3, D4 and D5 dopamine GPCRs. Drugs acting on dopamine receptors are significant tools for the management of several neuropsychiatric disorders including schizophrenia, bipolar disorder, depression and Parkinson's disease. Recent investigations of dopamine receptor signalling have shown that dopamine receptors, apart from their canonical action on cAMP-mediated signalling, can regulate a myriad of cellular responses to fine-tune the expression of dopamine-associated behaviours and functions. Such signalling mechanisms may involve alternate G protein coupling or non-G protein mechanisms involving ion channels, receptor tyrosine kinases or proteins such as β-arrestins that are classically involved in GPCR desensitization. Another level of complexity is the growing appreciation of the physiological roles played by dopamine receptor heteromers. Applications of new in vivo techniques have significantly furthered the understanding of the physiological functions played by dopamine receptors. Here we provide an update of the current knowledge regarding the complex biology, signalling, physiology and pharmacology of dopamine receptors.
Table of Links
TARGETS | ||
---|---|---|
GPCRsa | Enzymesb | Transportersc |
5-HT receptors | adenylyl cyclases | DAT |
Adenosine A1 receptor | Akt | Catalytic receptorsd |
Adenosine A2A receptor | calpain | ErbB (epidermal growth factor) receptor family |
α1B-Adrenoceptor | CaMKII | ErbB-1 |
β1-Adrenoceptor | CDK5 | IGFR1 |
Dopamine D1 receptor | ERK | PDGFRβ |
Dopamine D2 receptor | Epac1 | RTKs |
Dopamine D3 receptor | Epac2 | TrkB |
Dopamine D4 receptor | GRK2 | Ion channelse |
Dopamine D5 receptor | GSK3 | GIRKs |
Ghrelin receptor | GSK3α | Kir2 channels |
Muscarinic M4 receptor | GSK3β | IP3 receptor |
TA1 receptor | MAPK | Ligand-gated ion channelsf |
PDK1 | GluA1 | |
PKA | GluN2B | |
PKC | Ionotropic glutamate receptors | |
PLC | ||
PLCβ |
LIGANDS | ||
---|---|---|
amphetamine | fluoxetine | quetiapine |
apomorphine | GABA | quinpirole |
aripiprazole | haloperidol | risperidone |
asenapine | huntingtin | SCH23390 |
BDNF | IGF | SKF 38393 |
blonanserin | IGF-1 | SKF 81297 |
brexpiprazole | iloperidone | thioridazine |
bromocriptine | insulin | UNC0006 |
cAMP | L-DOPA | UNC9975 |
cariprazine | lithium | UNC9994 |
chlorpromazine | lurasidone | xanomeline |
clozapine | NMDA | ziprasidone |
cocaine | olanzapine | |
dopamine | perphenazine |
This table lists protein targets and ligands, which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and the Concise Guide to PHARMACOLOGY 2013/14 (a,b,c,d,e,fAlexander et al., 2013a,b,c,d,e,f).
Introduction
Five subtypes of dopamine receptors (D1, D2, D3, D4 and D5 receptors, encoded in humans by genes DRD1, DRD2, DRD3, DRD4 and DRD5, respectively) are known to mediate essentially all of the physiological functions of dopamine. These functions include, but are not limited to, the following: voluntary movement, reward, sleep regulation, feeding, affect, attention, cognitive function, olfaction, vision, hormonal regulation, sympathetic regulation and penile erection. Dopamine receptors are also known to influence the immune system as well as cardiovascular, renal and gastrointestinal functions. As members of the GPCR superfamily, dopamine receptors have a canonical seven-transmembrane structure and can signal through both G protein-dependent and -independent mechanisms. Based on coupling to either Gαs,olf proteins or Gαi/o proteins to stimulate or inhibit the production of the second messenger cAMP, respectively, dopamine receptors are classified as D1-class receptors (D1 and D5) or D2-class receptors (D2, D3 and D4) (Kebabian, 1978; Spano et al., 1978). The alternative splicing of D2 results in the generation of two major D2 dopamine receptor variants that differ in the presence of an additional 29 amino acids on the third intracellular loop with distinct physiological, signalling and pharmacological properties, and are classified as D2S (D2-short) and D2L (D2-long). Dopamine receptors are well-established targets in the clinical pharmacology of numerous disorders and conditions such as schizophrenia, Parkinson's disease, bipolar disorder, depression, restless leg syndrome, hyperprolactinaemia, pituitary tumours, hypertension, gastroparesis, nausea and erectile dysfunction. The basic principles of dopamine receptor structure, signalling, function and pharmacology are covered in detail in several excellent reviews (Niznik and Van Tol, 1992; Sibley and Monsma, 1992; Sokoloff et al., 1992; Civelli et al., 1993; Missale et al., 1998; Vallone et al., 2000; Carlsson, 2001; Seeman, 2006). Recently, we have provided a comprehensive overview of the field in Pharmacological Reviews (Beaulieu and Gainetdinov, 2011). However, although the basic information regarding the structural, genetic and biochemical properties of dopamine receptors has remained essentially unchanged in the last 4 years, a significant amount of new information has emerged on dopamine receptor signalling, functional relevance and pharmacology that requires an update of the status of current knowledge. Here we will focus on newly emerging topics and trends in understanding dopamine receptor biology as well as topics that were not covered or only partially discussed in our previous review (Beaulieu and Gainetdinov, 2011).
Mechanisms of dopamine receptor signalling
The prevailing convention was that dopamine receptors were considered to signal exclusively through G protein-dependent cellular processes. The D1-class receptors (D1 and D5 receptors) are primarily coupled to Gαs/olf proteins and stimulate the activity of AC and the production of the second messenger cAMP (Figure 1). In contrast, the D2 class receptors (D2S, D2L, D3 and D4 receptors) are associated with Gαi/o proteins to inhibit the production of cAMP (Kebabian, 1978; Spano et al., 1978) (Figure 2).
Figure 1.
Schematic diagram representing the signalling cascades activated by the D1 dopamine receptor (D1R). D5R, D5 dopamine receptor; D1R:D2R, D1–D2 receptor heteromer.
Figure 2.
Schematic diagram representing the signalling cascades activated by the D2 dopamine receptor (D2R). BMAL1, aryl hydrocarbon receptor nuclear translocator-like protein; Clock, circadian locomotor output cycles kaput gene; Cry2, cryptochrome 2; KLC2, kinesin light chain 2; Rev/Erbα, nuclear receptor subfamily 1, group D, member 1.
Modulation of cAMP synthesis by dopamine receptors results in the regulation of PKA and potentially of other exchange proteins activated by cAMP (Epac1 and Epac2) (Svenningsson et al., 2004; Beaulieu and Gainetdinov, 2011). Among PKA substrates, the multifunctional dopamine and cAMP-regulated phosphoprotein (DARPP-32/PPP1R1B) has been extensively studied over the last 30 years. When phosphorylated on Thr34 by PKA, DARPP-32 is a negative regulator of protein phosphatase 1 (PP1). In contrast, phosphorylation of DARPP-32 on Thr75 by cyclin-dependent kinase 5 (CDK5), in response to sustained D1 receptor activation, results in PKA inhibition (Figures 1 and 2). The roles of PKA and DARPP-32 in dopamine receptor signalling are well characterized, and strong evidence supports their contribution to the physiological functions of dopamine receptors (Svenningsson et al., 2004; Girault, 2012).
cAMP-mediated signalling and mRNA translation
An interesting development in the characterization of cAMP-mediated dopamine receptor signalling involves its recently appreciated contribution to the regulation of mRNA translation mechanisms. Either D1 receptor activation or D2 receptor blockade by haloperidol has been shown to promote the phosphorylation of the ribosomal protein S6 (rpS6) on Ser235/236 and Ser240/244 (Santini et al., 2009; Valjent et al., 2011). Phosphorylation of rpS6 on these and adjacent residues results in enhanced CAP-dependent mRNA translation (Roux et al., 2007; Hutchinson et al., 2011). Increased phosphorylation of rpS6 via D1 receptors would involve activation of PKA, subsequent inhibition of PP1 by DARPP-32 and activation of the mammalian target of rapamycin (mTOR) complex 1 (Santini et al., 2009; 2012; Bonito-Oliva et al., 2013). In medium spiny neurons expressing D2 receptors, activation of PKA and DARPP-32 by the adenosine A2A receptors also plays a role (Valjent et al., 2001; 2011; Santini et al., 2009), whereas activation of ERK signalling by a DARPP-32-dependent mechanisms are thought to be involved in D1 receptor-expressing medium spiny neurons (Santini et al., 2012). Interestingly, D1 receptor stimulation also promotes rpS6 phosphorylation in the dentate gyrus, albeit through a different, mTOR-independent, pathway involving ERK activation (Gangarossa and Valjent, 2012). Understanding the overall importance of dopamine receptor-mediated regulation of rpS6 on mRNA translation and behaviour is still in its infancy. However, preliminary evidence supports its involvement in the development of L–DOPA-induced dyskinesia (Santini et al., 2009; 2012; Subramaniam et al., 2012) and cocaine sensitization, seeking and relapse behaviours (Wu et al., 2011).
cAMP-independent dopamine receptor signalling
In addition to the regulation of cAMP, several studies have revealed that dopamine receptors can exert some of their biological effects through alternative signalling pathways (Beaulieu et al., 2004; 2005; Hasbi et al., 2009). For instance, there are indications that both D1 and D2 receptors can transactivate the brain-derived neurotrophic factor (BDNF) receptor in neurons (Swift et al., 2011). These two dopamine receptors can also regulate calcium channels through a direct protein–protein interaction in vivo (Kisilevsky and Zamponi, 2008; Kisilevsky et al., 2008). Direct interaction of D1 and D2 receptors and Na+-K+-ATPase has also been demonstrated (Hazelwood et al., 2008; Blom et al., 2012). Under certain circumstances, dopamine receptors can also regulate IP3-mediated signalling (Medvedev et al., 2013; Perreault et al., 2014), and there is evidence for alternative coupling of D1-class receptors to Gαq (Figure 1).
The D2-class D2 and D3 receptors have been shown to signal through both G protein-dependent and G protein-independent mechanisms (Beaulieu and Gainetdinov, 2011). G protein-dependent mechanisms for D2 dopamine receptors are represented by the well-known Gαi/o subunit-mediated cAMP-PKA-DARPP32 cascade (Svenningsson et al., 2004) and the Gβγ-mediated activation of PLC, leading to increased cytoplasmic calcium and downstream signalling events (Hernandez-Lopez et al., 2000; Beaulieu and Gainetdinov, 2011). Furthermore, Gβγ-mediated mechanisms are involved in the regulation of activity of the L- and N-type calcium channels (Yan et al., 1997) as well as G protein coupled inwardly rectifying potassium channels (GIRKs) (Kuzhikandathil et al., 1998; Beaulieu and Gainetdinov, 2011). Recent evidence indicates that all of these G protein-mediated signalling cascades converge on, among other targets, phosphorylation of two subunits of ionotropic glutamate receptors, GluA1 and GluN2B, which are critically involved in glutamatergic transmission (Jenkins and Traynelis, 2012; Dell'anno et al., 2013; Hobson et al., 2013; Jia et al., 2013; Song et al., 2013; Flores-Barrera et al., 2014; Jenkins et al., 2014; Murphy et al., 2014) (Figure 2).
Finally, there is strong evidence that D2 dopamine receptors can signal in vivo by activating cAMP-independent mechanisms involving the multifunctional adaptor protein β-arrestin 2 (βArr2) (Beaulieu et al., 2004; 2005; 2008b,,; Urs et al., 2012) (Figure 2). In the remaining parts of this subsection, we will provide an overview of recent evidence underscoring the importance of cAMP-independent mechanisms in dopamine receptor function.
Coupling of dopamine receptors to Gαq
Several lines of evidence support the regulation of PLC and calcium signalling by dopamine receptors. As early as 1989, Felder et al. reported that the D1 receptor agonist SKF 82526 stimulates PLC activity independently of cAMP in renal tubular membranes (Felder et al., 1989). Activation of PLC leads to the production of inositol trisphosphate (IP3) and DAG. This results in the activation of PKC by DAG and an increased mobilization of intracellular calcium in response to IP3 (Berridge, 2009). The increase of intracellular calcium in the cytoplasm leads to the activation of calcium-dependent PKC variants as well as calcium-regulated enzymes, such as the calcium/calmodulin-dependent PK II (CaMKII) and the protein phosphatase calcineurin/protein phosphatase 2B (PP2B).
The most common way for a GPCR to regulate PLC activity is by coupling to Gαq. Putative D1–D2 receptor heterodimers have been suggested to regulate DAG and IP3 signalling by activating Gαq/11 in transfected cells as well as in striatal membrane preparations (Lee et al., 2004; Rashid et al., 2007b). The physiological relevance of D1–D2 receptor heterodimers is supported by the co-expression of D1 and D2 receptors in small populations of medium spiny neurons of the nucleus accumbens in the mouse (Rashid et al., 2007b) and in other regions of the basal ganglia (Perreault et al., 2010). Notably, analysis of BAC transgenic mice that express fluorescent gene-reporter proteins driven by D1 and D2 receptor promoters showed that the majority of D1 receptor-positive pyramidal neurons in the prefrontal cortex also express low levels of D2 receptors (Zhang et al., 2010). In addition to co-expression studies, FRET studies conducted with fluorescent proteins in transfected cells and treatments of tissue sections with labelled antibodies have produced results that suggest the formation of receptor heterodimers (Rashid et al., 2007b; Perreault et al., 2013).
Despite accumulating evidence, the involvement of D1–D2 receptor heterodimers in the regulation of PLC-mediated signalling in vivo remains poorly understood. It should be noted that recent studies have questioned the selectivity (Chun et al., 2013) and PLC activity (Lee et al., 2014) of the putative D1–D2 receptor heteromer agonist SKF 83959 that was used to characterize the role of D1–D2 receptor heterodimers in the regulation of PLC in vivo (Rashid et al., 2007b). One important aspect of D1–D2 heterodimer signalling in cells is the requirement of co-activation of both the D1 and D2 receptor moiety to activate Gαq/11. Furthermore, the formation of the D1–D2 receptor heterodimers would prevent coupling of either receptors to Gαs/olf or Gαi/o (Perreault et al., 2014). This theory, however, is in contrast with several in vivo observations supporting the regulation of PLC by D1-class receptors without the need for D2 receptor involvement. It was recently reported that acute systemic administration of cocaine, amphetamine, apomorphine or the D1-class receptor agonist SKF 81297 to wild-type mice increases striatal IP3 synthesis (Medvedev et al., 2013). Co-treatments with selective antagonists as well as the use of D1 and D2 receptor knockout (KO) animals revealed that the production of IP3 in response to these pharmacological treatments requires D1, but not D2 receptor activation. Importantly, PLCβ inhibition suppressed spontaneous locomotor hyperactivity in hyperdopaminergic mice lacking the dopamine transporter (DAT) and antagonized the effects of amphetamine, cocaine, SKF 81297 and apomorphine on forward locomotion. Furthermore, the restoration of locomotion by L-DOPA in dopamine-depleted mice (Sotnikova et al., 2005) is also reduced by inhibition of PLCβ resulting in mostly vertical activity following these treatments (Medvedev et al., 2013). These data strongly support a contribution of PLC in mediating the effects of dopamine on forward locomotion. However, further investigation is necessary to decipher the relative contribution of different modes of PLC regulation on the various aspects of dopamine-related behaviours.
At the same time, expression of D1 receptors in transfected HEK293 cells does not affect intracellular calcium signalling. However, expression of D5 receptors in the same cells induces extensive calcium mobilization after stimulation (So et al., 2009), and the D1-class receptor agonist SKF 38393 activates PLC-mediated signalling in D1 receptor KO mice (Friedman et al., 1997). Furthermore, this same agonist, as well as dopamine and SKF 83959, failed to increase IP3 levels in brain slices prepared from mice lacking D5 receptors (Sahu et al., 2009). A similar lack of responsiveness of PLC-mediated signalling to SKF 83959 was also reported following systemic administration of this compound to D5 receptor KO mice (Sahu et al., 2009), suggesting that activation of D5 receptors is sufficient to activate Gαq/11 in response to selected doses of certain D1-class receptor agonists.
Thus, several independent studies support the regulation of PLC-mediated signalling through dopamine receptors, however, these studies are in disagreement with regard to the detailed mechanism of this regulation. Current evidence does not allow us to rule out the contributions of D1 receptors, D5 receptors or D1–D2 receptor heterodimers in this phenomenon (Figure 1). Discrepancies between the results of different research groups raise the possibility that several mechanisms may be involved, perhaps in different neuronal populations. It is also conceivable that different D1-class receptor agonists may be functionally selective for PLC-mediated mechanisms when activating D1 receptors, D5 receptors or D1–D2 receptor heterodimers.
Beyond the question of its detailed mechanism, activation of PLC-mediated signalling by dopamine also raises the question of possible crosstalk between this modality of signalling and cAMP-mediated mechanisms. Among several possibilities, activation of PKC and CaMKII through calcium signalling could affect glutamate receptors concomitantly with PKA (Figure 1). Different mechanisms involving either positive or negative regulation of CDK5 by calcium may also be an important nexus for crosstalk. For instance, PKC has previously been shown to prevent the phosphorylation of DARPP-32 and other substrates by CDK5 (Sahin et al., 2008). Because the global activity of DARPP-32 is modulated by an equilibrium between its phosphorylation by CDK5 and PKA (Bibb et al., 1999) it is possible that Gαq/11-mediated dopaminergic signalling may reduce the phosphorylation of DARPP-32 by CDK5 and potentiate PKA-mediated signalling (Figure 1). In contrast, cleavage of the CDK5 co-activator p35 by the calcium-regulated protease calpain (Lee et al., 2000; Beaulieu and Julien, 2003) may result in CDK5 hyperactivity and an inhibition of PKA signalling. Furthermore, changes in calcium concentration may also affect the activity of PP2B (calcineurin), which is involved in the dephosphorylation of DARPP-32 at Thr34 (Halpain et al., 1990). Overall, the full understanding of the regulation of PLC activity by dopamine remains incomplete yet holds promise for exciting future investigations.
Coupling of D2-class receptors to βArr2, Akt and glycogen synthase kinase (GSK3)
G protein-independent D2 receptor signalling is represented by βArr2-mediated mechanisms. Arrestins are a family of four molecular adaptor proteins that were originally characterized for their role in mediating GPCR desensitization and internalization (Lohse et al., 1990; Ferguson et al., 1996). In addition to these functions, the two ubiquitous arrestins, βArr1 and βArr2, have also been shown to act as molecular scaffolds for signalling molecules such as kinases and phosphatases (Luttrell et al., 2001; Beaulieu et al., 2005).
Several lines of evidence have pointed towards the contribution of a βArr-mediated mechanism in the regulation of the serine/threonine kinases Akt and GSK3 by dopamine. Akt is involved in several cellular processes such as glucose metabolism, gene transcription, cell proliferation, migration and neurotrophin action through the stimulation of receptor tyrosine kinases (RTKs) (Cross et al., 1995; Alessi et al., 1996; Scheid and Woodgett, 2001). Activation of RTKs and some GPCRs regulates PI3K, which converts phosphatidylinositol-2-phosphate (PIP2) to phosphatidylinositol-3-phosphate (PIP3) (Martelli et al., 2010). This newly formed PIP3 interacts with the pleckstrin homology domain of Akt, inducing the recruitment of Akt to the plasma membrane. This, in turn, results in the phosphorylation of Akt at the Thr308 and Ser473 residues by two phosphatidylinositol-dependent kinases, PDK1 and PDK2/rictor-mTOR respectively (Scheid and Woodgett, 2001; Jacinto et al., 2006). Once activated, Akt phosphorylates several substrates including GSK3 (Rossig et al., 2002). Mammalian cells express two isoforms of GSK3, GSK3α and GSK3β, which are constitutively active and can phosphorylate several cellular substrates (Woodgett, 1990; Kaidanovich-Belin and Woodgett, 2011). Phosphorylation by Akt inhibits both isoforms of GSK3 in response to growth factors and hormones, including insulin, IGF, and BDNF (Yamada et al., 2002; Altar et al., 2008). Specifically, Akt phosphorylates Ser21 on GSK3α and Ser9 on GSK3β, which are located on their respective N-terminal domains (Stambolic and Woodgett, 1994; Frame and Cohen, 2001).
Experiments using dopamine receptor agonists/antagonists, dopamine depletion and hyperdopaminergic DAT-KO mice have provided converging evidence for the negative regulation of Akt, resulting in the activation of both GSK3 isoforms by D2-class receptors in mammals and other vertebrates (Beaulieu et al., 2004; Bychkov et al., 2007; Chen et al., 2007; Souza et al., 2011). Consequently, D2-class receptor antagonists induce Akt activation and subsequent GSK3 inhibition (Beaulieu et al., 2004; Emamian et al., 2004). Additional investigations conducted using mice lacking various dopamine receptors have shown that a loss of D2, but not D1 receptors prevents the inactivation of striatal Akt by drugs acting on dopamine neurotransmission (Beaulieu et al., 2007b). In contrast, D3 receptor-deficient mice exhibit a reduction of Akt phosphorylation in response to dopaminergic drugs. This suggests that D2 receptors are critical for the inhibition of Akt by dopamine, whereas the D3 receptors appear to potentiate the D2 receptor-mediated dopamine response (Beaulieu et al., 2007b).
The role of βArr2 in mediating the regulation of Akt and GSK3 by D2 receptors is supported by direct in vivo biochemical observations in pharmacological and genetic models of enhanced dopaminergic neurotransmission (Beaulieu et al., 2004; 2005). Amphetamine and apomorphine have been shown to inhibit the phosphorylation and activation of Akt in the striatum of wild-type mice, whereas these two drugs failed to inhibit Akt in βArr2-KO mice. Furthermore, regulation of Akt and GSK3 signalling observed in mice with genetically increased dopaminergic tone caused by a lack of DAT, was absent in double mutant mice deficient for both DAT and βArr2, suggesting an important role of this scaffolding protein in Akt regulation by dopamine (Beaulieu et al., 2005). Further characterization of the molecular mechanisms underlying the regulation of Akt by D2 receptors, following receptor stimulation has shown that βArr2 is involved in the formation of a protein complex composed of Akt, βArr2 and protein phosphatase 2A (PP2A) (Beaulieu et al., 2005). Formation of this complex allows PP2A to dephosphorylate and inactivate Akt, resulting in the activation of GSK3 (Beaulieu et al., 2004; 2005).
It is worth mentioning that the formation of the Akt : βArr2 : PP2A signalling complex in response to D2 receptor activation represents a mechanism through which dopamine can trigger the inactivation of PI3K/Akt signalling in a regulated fashion. Importantly, the Akt : βArr2 : PP2A signalling complex dissociates in response to lithium, thus providing a probable explanation for the early behavioural observations of the antagonistic effect of lithium on dopaminergic behaviours as well as a reasonable mechanism for the activation of Akt by lithium (Beaulieu and Caron, 2008a; O'Brien et al., 2011; Pan et al., 2011). The details of the mechanism(s) by which lithium triggers this dissociation are not yet fully understood. Current evidence suggests that lithium may affect the stability of this complex by acting on several of its components, possibly in a synergistic fashion. First, lithium has been shown to interfere with the interaction of Akt1 and βArr2 (Beaulieu et al., 2008b). Direct investigation of the Akt–βArr2 interaction using recombinant proteins have demonstrated that this interaction is dependent upon the presence of magnesium ions and that excess magnesium can prevent the dissociation of Akt and βArr2 upon treatment with a therapeutic dose of lithium (1 mM). Second, GSK3β has also been shown to interact with βArr2. Recent evidence obtained from transgenic mice overexpressing Xenopus GSK3β in neurons indicate that activated GSK3 can act as a feed-forward mechanism for its own activation (Figure 2) by stabilizing the Akt : βArr2 : PP2A signalling complex (O'Brien et al., 2011). According to this model, direct inhibition of GSK3 by lithium would thus constitute a mechanism that can promote the disassembly of the Akt : βArr2 : PP2A.
The effect of βArr2-mediated Akt/GSK3 signalling on dopaminergic behaviours is supported by several experimental observations in vivo. βArr2-KO mice have been shown to display spontaneous locomotor hypoactivity, reduced apomorphine-induced climbing and amphetamine-induced hyperlocomotion (Gainetdinov et al., 2004; Beaulieu et al., 2005). These mice also have a reduced responsiveness to the dopamine-dependent locomotor effects of morphine (Bohn et al., 2003). In addition, novelty-driven locomotor hyperactivity, a phenotype that is typical of hyperdopaminergic DAT-KO mice, is less pronounced in double mutant mice lacking both βArr2 and DAT (Beaulieu et al., 2005). Administration of lithium exerts multiple actions on behaviours in DAT-KO and normal mice, including suppression of spontaneous locomotor activity, but this was not observed in βArr2-KO mice (Beaulieu et al., 2004; 2005). In line with these data, mice lacking Akt1 demonstrate an enhanced sensitivity to amphetamine with regard to the disruption of sensorimotor gating in the pre-pulse inhibition (PPI) test, which is used to model psychosis in rodents (Emamian et al., 2004). As described above, Akt1 is inhibited following the stimulation of D2 receptors, thus the increased behavioural effect of amphetamine in Akt1-KO mice is likely to result from the involvement of Akt in dopaminergic behavioural responses.
Genetic suppression of GSK3 activity also inhibits locomotor hyperactivity related to excessive dopaminergic tone in amphetamine-treated mice (Beaulieu et al., 2004). Similarly, several GSK3 inhibitors as well as GSK3β haploinsufficiency can block amphetamine-induced hyperactivity (Beaulieu et al., 2004; Gould et al., 2004; Kalinichev and Dawson, 2011). In contrast, mice overexpressing GSK3β show pronounced locomotor hyperactivity (Prickaerts et al., 2006), and transgenic mice expressing a GSK3β mutant that lacks an inhibitory phosphorylation site (thus is constitutively active) demonstrate increased novelty-driven and amphetamine-induced hyperactivity (Polter et al., 2010).
More recent evidence obtained using strains of cell type-specific conditional GSK3β-KO mice have generated a more nuanced portrait of the contribution of GSK3β in the regulation of dopaminergic behaviour. Ablation of GSK3β expression specifically in D1 or D2 receptor-expressing striatal neurons (Urs et al., 2012) confirmed the selective contribution of GSK3β to the acute action of amphetamine on locomotion in D2 but not D1 receptor-expressing neurons. The antagonistic action of the D2 receptor partial agonist aripiprazole and lithium on amphetamine-induced locomotion is also curbed in mice lacking GSK3β in D2 receptor-expressing neurons. In contrast, haloperidol-induced catalepsy is not affected by diminished GSK3β expression in either D1 or D2 receptor-expressing striatal neurons, whereas the disruptive effects of amphetamine on sensory motor gating is abolished by in either D1 or D2 receptor-expressing neuron-selective GSK3β gene inactivation. Taken together, these observations confirm the role of βArr2-mediated regulation of GSK3β in D2 receptor-expressing neurons in the effects of amphetamine, lithium and aripiprazole on locomotion. The fact that haloperidol-induced catalepsy remains intact in mice lacking GSK3β suggests the involvement of at least two separate signalling pathways mediating the effects of antipsychotics and strengthens the rationale for the development of biased D2 receptor antagonists to selectively target these pathways in schizophrenia (Beaulieu et al., 2007a; Beaulieu, 2012). Further confirmation of these hypotheses should come from repeating these experiments in mice lacking βArr2 in specific neuronal populations.
Selective ablation of GSK3β post-natally in forebrain pyramidal neurons revealed other functions of GSK3β in the regulation of dopamine-associated behaviours (Latapy et al., 2012). The locomotor effects of amphetamine are marginally increased in these mice, which suggests a minor role of cortical neurons in the modulation of amphetamine action and further indicates that the opposing effect of GSK3β inhibition on amphetamine-induced locomotion is mediated by GSK3β in subcortical structures. Additionally, these mice display reduced anxiety and enhanced social interactions. Investigation of the possible contribution of GSK3β in behavioural responses to social defeat stress (Wilkinson et al., 2011; Latapy et al., 2012) using either conditional forebrain KO mice, GSK3β haplo-insufficient mice or mice expressing a dominant negative GSK3 in the nucleus accumbens also revealed a role for subcortical GSK3β inhibition in mediating resilience to this form of stress. This emphasizes the need to further examine the contribution of GSK3-mediated dopamine receptor signalling in coping behaviours.
Beyond its potential involvement in the action of lithium, βArr2-mediated D2 receptor signalling can also contribute to effects of antipsychotics. Characterization of the effects of different antipsychotics using BRET in transfected HEK293 cells revealed that first-generation antipsychotics (chlorpromazine, haloperidol), as well as second- (clozapine, quetiapine, olanzapine, risperidone, ziprasidone) and third- (aripiprazole) generation antipsychotics potently antagonize quinpirole-induced βArr2 recruitment to D2 receptors (Masri et al., 2008). In contrast, strong differences existed in the potency of these drugs in preventing inhibition of cAMP synthesis by D2 receptors. Of interest, D2 receptor partial agonist aripiprazole displayed partial D2 receptor agonist activity for cAMP-mediated signalling in the absence of quinpirole while functioning as an antagonist for cAMP when quinpirole was applied concomitantly. Because second- and third-generation antipsychotics are characterized by fewer extrapyramidal side effects, this study led to the hypothesis that identification of functionally selective D2 receptor antagonists that specifically prevent βArr2 recruitment to D2 receptors may pave the way for the development of new antipsychotics that would have fewer side effects while retaining their therapeutic activity.
This hypothesis led to the development of new aripiprazole derivative compounds: UNC9975, UNC0006 and UNC9994, which display antipsychotic-like activity in rodents (Allen et al., 2011). In the absence of a full agonist, these three compounds have the distinction of acting as partial D2 agonist for βArr2 recruitment without affecting cAMP. It should be noted, however, that these compounds may not be fully functionally selective as Allen et al. also reported that they can act as neutral antagonists for cAMP-mediated D2 signalling. It is also noteworthy that aripiprazole behaves as a partial agonist for βArr2 recruitment when applied alone on cells (Allen et al., 2011) while acting as an antagonist of βArr2 recruitment when simultaneously applied with quinpirole (Masri et al., 2008). It is thus possible that the UNC compounds may display different pharmacological properties when applied alone in vitro and in the context of an active dopamine tone in vivo where they might antagonize both cAMP and βArr2 mediated D2 receptor signalling through a combination of neutral antagonism and partial agonism.
Overall, βArr2-mediated D2 receptor signalling provides interesting avenues for the development of new drugs targeting dopamine neurotransmission. However, it is unclear at the moment whether this type of intervention will be more suited for clinical interventions in schizophrenia or bipolar disorder. Indeed, this form of signalling is directly targeted by lithium (Beaulieu et al., 2004; 2008b,), a drug that has very limited efficacy for the treatment of schizophrenia.
Protein phosphatase metallo-dependent (PPM/PP2C) and Gαi/o mediated regulation of huntingtin (HTT) protein phosphorylation by D2 receptors
Recent investigation has revealed a role of D2 receptors in the regulation of the phosphorylation of the HTT protein on Ser421 (Marion et al., 2014). It is known that phosphorylation of HTT on this residue by Akt in response to IGF-1 leads to reduction of the formation of nuclear inclusions and HTT toxicity (Humbert et al., 2002; Rangone et al., 2004). Intriguingly, D2 receptor stimulation reduces the phosphorylation of HTT on this residue in heterologous cells and in the mouse striatum (Marion et al., 2014). The molecular mechanism of this regulation appears not to involve the regulation of Akt by D2 receptors. Instead, the regulation of HTT phosphorylation by D2 receptors involves the activation of Gαi/o and the formation of a protein complex between HTT and D2 receptors. Indeed, treatment of transfected cells with the Gαi/o inhibitor, Pertussis toxin, prevented the dephosphorylation of HTT in response to D2 receptor stimulation. Furthermore, the study revealed the formation of a protein complex comprising D2 receptors, HTT and two members of the PPM/PP2C family. The first of these phosphatases, PPM1A, was shown to interact directly with HTT in vivo whereas the second phosphatase, PPM1B as well as HTT interact directly with the D2 receptors. While it is not clear at the moment if PPM1A and B both participate in HTT dephosphorylation and the contribution of cAMP-mediated mechanisms has remained unexplored, the potential involvement of this mechanism in the regulation of HTT toxicity certainly warrants further investigations.
Transactivation of RTK by dopamine receptors
RTKs are a major family of cell surface receptors involved in many functions in neuronal and non-neuronal cell types (Lemmon and Schlessinger, 2010). Members of this family include, among others, the BDNF receptor neurotrophic tyrosine kinase, receptor, type 2 (TrkB), EGF/neuregulin family receptors (ErbB family) and receptors for insulin and insulin-like growth factor 1 (IGFR1). Activation of RTKs by their cognate ligands enhances receptor dimer formation, internalization, and recruitment of monomeric receptors to the cell surface. RTK activation generally results in a concomitant rapid activation of several signalling pathways, including PI3K/Akt, Ras/MAPK and PLC-mediated signalling (Figure 2).
In addition to direct activation by their ligands, RTKs can also be transactivated by GPCRs (Eguchi et al., 1998; Maudsley et al., 2000; Rajagopal et al., 2004). However, the molecular mechanisms of this transactivation are not clearly understood. Independent investigations conducted in different systems have underscored the possible contribution of both G protein- and arrestin-mediated mechanisms involving either direct activation of RTK by intracellular processes or autocrine/paracrine RTK activation following ligand shedding in response to GPCR activation.
Dopamine receptors have been shown to transactivate RTKs in different experimental systems (Figure 3). The D4 receptor was shown to transactivate the platelet-derived growth factor β (PDGFβ) receptor, and D2 receptors were able to transactivate IGF receptors in heterologous cell systems (Chi et al., 2010; Mannoury la Cour et al., 2011). Furthermore, both D1 and D2 receptors can transactivate ErbB-1 in transfected CHO-K1 cells (Swift et al., 2011) and primary neuron cultures (Iwakura et al., 2011; Yoon and Baik, 2013). Finally, D1, D2 and potentially D1–D2 receptor heteromers have been shown to transactivate the BDNF receptor in cultured striatal neurons (Iwakura et al., 2008; Swift et al., 2011; Barbeau et al., 2013). Systemic administration of the D1-class receptor agonist SKF 38393 also increased TrkB activation at 3 and 6 h following drug injection in 4-day-old rats (Iwakura et al., 2008). The treatment of rats with the D1 receptor antagonist SCH23390 reduced striatal TrkB activation, suggesting that transactivation of TrkB by D1 receptors occurs in response to normal endogenous dopamine tone.
Figure 3.
Mechanisms and signalling events involved in the transactivation of RTK by dopamine receptors. Raf, proto-oncogene serine/threonine-PK; Ras, rat sarcoma family of small GTPases.
The mechanisms by which dopamine receptors transactivate RTKs are not fully understood. Quantitative pharmacological characterization of ErbB-1 receptors by various GPCRs has shown that these phenomena are not restricted by the coupling of the GPCR to a different G protein (Swift et al., 2011). Increased release of the RTK ligand BDNF does not appear to be essential for the transactivation of TrkB by D1 receptors (Iwakura et al., 2008). However, release of EGF appears to play a role in the transactivation of ErbB-1 by D2 receptors in cultured neurons (Iwakura et al., 2011; Yoon and Baik, 2013). Interestingly, stimulation of either frontal cortex D5 receptors (Perreault et al., 2013) or striatal D1–D2 receptors (Hasbi et al., 2009) has also been reported to increase BDNF levels, therefore potentially leading to increased TrkB activation in response to dopamine receptor stimulation.
Transactivation of RTKs by dopamine receptors can have a major effect on our understanding of dopamine receptor signalling in vivo. RTKs are coupled to several signalling mechanisms that can elicit cellular responses, which are beyond the direct effect of G protein or arrestin-mediated cellular responses. For instance, although D2 receptor stimulation leads to a βArr2-dependent inactivation of Akt and concomitant activation of GSK3 in vivo, the opposite has been reported to occur in heterologous cell systems and, in some cases, cultured neurons (Brami-Cherrier et al., 2005; Beaulieu, 2012). It has recently been shown that activation of Akt by recombinant D2 receptors in transfected cells can be attributed to the transactivation of IGFR and concomitant activation of PI3K-mediated signalling by this RTK (Mannoury la Cour et al., 2011). It is also possible that some instances of MAPK and PLC-mediated signalling in response to dopamine receptor activation may also result from RTK transactivation (Figure 3). The apparent involvement of RTK ligand release in the transactivation of some RTKs by dopamine receptors also raises the possibility that activation of dopamine receptors in dopaminoceptive neurons may elicit paracrine signalling responses to dopamine in either non-dopaminoceptive neurons or non-neuronal cells, possibly leading to indirect regional effects of dopamine receptor stimulation. This type of regional responses may be important considering the potential role of TrkB, and probably other RTKs, in regulating drug-induced reward (Lobo et al., 2013).
Dopamine receptor oligomerization
Historically, GPCRs are believed to function as monomeric units, but now there is mounting evidence indicating that several GPCRs can exist in oligomeric forms (Perreault et al., 2014). Regarding dopamine receptors, they can form both homomers and heteromers with several receptors, including other GPCRs and ionotropic glutamate receptors (Guo et al., 2008; Van Craenenbroeck et al., 2011; Perreault et al., 2014). Some of these interactions may be regulated via mechanisms likely orchestrated by AC and cAMP (Woods and Jackson, 2013). A study by the Javitch group suggested that D2 receptor homodimers and a G protein exist as the minimal single functional unit, which is maximally activated by the binding of an agonist to only one protomer and is either negatively or positively modulated by the ligand to the other protomer of an agonist or an inverse agonist respectively (Han et al., 2009). This allosteric modulation between the two protomers of the complex is mediated through intermolecular interactions by the direct association among receptors and not by downstream effects. The development of RET-based techniques has been of fundamental importance in the discovery and characterization of many homomers and heteromers and is now considered to be the preferred biophysical method in describing complex formations (Milligan, 2004; Pfleger and Eidne, 2006; Marullo and Bouvier, 2007; Salahpour et al., 2012). Both BRET and FRET rely on the principle of a non-radiative energy transfer between a donor protein and a fluorescent acceptor. In case of FRET, the donor is also a fluorescent protein (e.g., CFP), whereas in BRET, the donor is an enzyme (Renilla luciferase) that produces bioluminescence upon the degradation of a substrate (coelenterazine h or derivatives) (Pfleger and Eidne, 2006). Because the energy transfer is only possible when the donor and the acceptor are closer than 10 nm, when two proteins fused to a donor and an acceptor produce a BRET or FRET signal, it is an indication of a physical contact (Pfleger and Eidne, 2006; Marullo and Bouvier, 2007; Lohse et al., 2012). However, a simple BRET or FRET signal is insufficient to distinguish true heterodimerization from a random collision; thus many experimental approaches have been adopted to characterize a putative heterodimer, such as saturation curves, competition assays and others (Marullo and Bouvier, 2007; Salahpour and Masri, 2007). Although these approaches have been extremely useful to study various complexes in cellular systems, these methods cannot simply be applied directly in native tissue, although there are some examples of successful applications in vivo using FRET with selective fluorescent ligands (Albizu et al., 2010) or antibodies (Perreault et al., 2010). After the in vitro description of the heterodimer and the characterization of its functional features, it is more common to prove the existence of the complex in native tissue using indirect evidence, such as identifying the ‘biochemical fingerprint’ and reproducing the specific characteristics of the complex and/or analysing physiological or behavioural responses to co-activation of the receptors (Ferre and Franco, 2010). In this section, we limit our discussion only to the heterodimers formed by dopamine receptors that have at least partial validation in studies in native tissue and/or in vivo.
D1–D2 receptor heterodimer
As mentioned earlier, D1 and D2 receptors can form a heterodimer complex that has been shown to exist in a heterologous system and in primary striatal neurons as well as in vivo in the rodent brain (Perreault et al., 2013; 2014). Several techniques have been used to characterize this complex, ranging from classic biochemical approaches, including co-immunoprecipitation of the two proteins (Lee et al., 2004), to more accurate techniques such as quantitative FRET (Dziedzicka-Wasylewska et al., 2006; Hasbi et al., 2009; Perreault et al., 2010). The expression and cellular localization of D1–D2 receptor heterodimers has been characterized not only in cells, but also in vivo, primarily in rat striatum. Studies from BAC transgenic mice demonstrate that the majority of the D1- and D2 receptor-expressing neurons are segregated in two different populations, although a small percentage of neurons express both receptors, ranging from a 6% in caudate putamen to a 15–30% in nucleus accumbens (Bertran-Gonzalez et al., 2008; Perreault et al., 2010). These MSNs expressing D1 and D2 receptors are interesting in that they express both dynorphin and enkephalin; thus, one might consider them to be a third, distinct subset on MSNs. Among these, not all of the neurons show constitutive D1–D2 receptor heterodimer formation. Although a small proportion of caudate putamen MSNs revealed a D1–D2 receptor complex, in most of the neurons (90%) expressing both D1 and D2 receptors in the nucleus accumbens, these receptors are present as heterodimers (Perreault et al., 2010). As mentioned earlier, this occurs under basal conditions, and it has been shown that several stimuli and pathological conditions could alter the state and the proportion of D1–D2 heterodimers (Dziedzicka-Wasylewska et al., 2006; Perreault et al., 2010). A peculiar aspect of this heterodimer is that it has a unique pharmacology that is distinct from that of its single protomer (Figure 1). Activation of the D1–D2 receptor complex induces the recruitment of the Gαq/11 protein, leading to the release of calcium from the internal stores (Rashid et al., 2007a,b; Hasbi et al., 2009). It appears that D1 and D2 receptors are both necessary for this pathway, thus the application of dopamine or a combination of two selective D1 and D2 receptor agonists are able to increase intracellular calcium, whereas treatment with either a D1 or D2 receptor antagonist can abolish this effect (Hasbi et al., 2009).
A selective D1–D2 receptor heteromer agonist (SKF 83959) has also been described: (Rashid et al., 2007b). It should be noted, however, that a recent report argues the selectivity of this compound by showing that this effect is dependent on the level of Gαq/11 expression in cells and there could be a contribution from the Gβγ subunits and GRK2 on the calcium increase induced by the D1–D2 receptor heterodimer (Chun et al., 2013), while another report questioned in general the ability of SKF 83959 to influence PLC (Lee et al., 2014). It has been suggested that this heteromer may play a role in brain disorders such as addiction (Perreault et al., 2010), schizophrenia (Dziedzicka-Wasylewska et al., 2008) and major depression (Pei et al., 2010), although additional evidence is needed to support these hypotheses.
D1-D3 receptor heterodimers
D1 and D3 receptors are co-expressed in the majority of the substance P – expressing GABAergic medium spiny neurons, suggesting that there could be functional crosstalk between these two receptors. Two independent studies have demonstrated that D1 and D3 receptors can form a constitutive heterodimer (Fiorentini et al., 2008; Marcellino et al., 2008). Using BRET and FRET techniques in transfected cells, it has been shown that D1 and D3 receptors can physically interact with no change in complex formation upon agonist treatment. Moreover, using co-immunoprecipitation, it was possible to isolate the D1–D3 complex from striatal membranes, confirming the existence of this heteromer in the brain. This cooperativity between D1 and D3 receptors is also evident in behavioural experiments. It is known that activation of D1 receptors stimulates locomotor activity, whereas the role of D3 receptors is less clear. In reserpinized mice (a model to isolate postsynaptic effects), D3 receptor agonists can potentiate the stimulatory effects of D1, but not D2 receptor agonists. Furthermore, this potentiation can be counteracted with a D3 receptor antagonist and is not present in D3 receptor KO mice (Marcellino et al., 2008). It has been suggested that the functional synergy of the D1–D3 dimer could be important for processes related to drug addiction and L–DOPA-induced dyskinesia in Parkinson's disease. It will be of interest to further characterize the physiological relevance of the D1–D3 heterodimer in other dopamine-related functions and pathologies.
D2–D4 receptor heterodimers
Both the long and the short D2 receptor isoforms can associate with D4 receptors, (Borroto-Escuela et al., 2011b; Gonzalez et al., 2012b). Using BRET, co-immunoprecipitation and proximity ligation assay, it has been shown in cell culture that D2L receptors can exist in a heterodimeric form with the major variants of the D4 receptors: D4.2, D4.4 and D4.7, with D4.7 being the least effective in forming the complex (Borroto-Escuela et al., 2011b). Ferrè and colleagues showed with BRET that D2S receptors can also associate in a heterodimer complex with the two variants D4.2 and D4.4, but not with the variant D4.7 (Gonzalez et al., 2012b). This study revealed a biochemical fingerprint for this heteromer that could potentiate D4 receptor activation of MAPK. In the mouse striatum, although the single activation of either D2 or D4 receptors had no effect on MAPK, co-administration of D2 and D4 receptor agonists induced a strong ERK phosphorylation response (Gonzalez et al., 2012b). This synergistic activity was lost in knock-in mice carrying the D4.7 variant, demonstrating the lack of mutual functional interaction between these two receptors. Moreover, the same synergistic effect was observed as regard to the ability of D4 receptors to modulate glutamate release in the striatum (Gonzalez et al., 2012b).
Dopamine receptor/NMDA receptor heteromer
Many studies have reported D1 receptor-mediated modulation of NMDA activity, primarily through a G protein-dependent mechanism involving the cAMP/PKA pathway and proteins such as DARPP-32 (Blank et al., 1997). However, there is evidence indicating that NMDA receptors and D1 receptors could also interact physically. A first study demonstrated the existence of this complex in hippocampal extracts by co-immunoprecipitation (Lee et al., 2002). It has been described that there are two sites of interaction in the carboxy-terminal domain of D1 receptor: one interacts with the GluN1 (NR1) subunit and the other with the GluN2A (NR2A) subunit. Each of these sites is responsible for a specific functional characteristic of the complex. These data were confirmed by another group in BRET studies that showed D1 receptors and GluN1 forming a constitutive heterodimer in cells (Fiorentini et al., 2003). Interestingly, the D1–GluN1 complex is formed early in the ER and is translocated to the membrane only upon association with the GluN2B (NR2B) subunit (Fiorentini et al., 2003). When at the cell membrane, the D1 receptors involved in this complex lose the ability to desensitize and internalize upon stimulation, suggesting a dual role of this receptor in native tissue depending on microdomain localization and aggregation with NMDA receptors, as previously reported (Dumartin et al., 1998). The activation of D1 receptors leads to a decrease of NMDA receptor functionality, as measured by NMDA-mediated currents in HEK cells and hippocampal neurons. This loss of activity is likely due to a decrease of NMDA receptor expression to the plasma membrane mediated by GluN2A subunit (Lee et al., 2002). D1 receptor agonists could reduce the cytotoxicity induced by an overactivation of the NMDA receptors. This effect appears to be mediated through a PI3K mechanism dependent on the GluN1 subunit instead of the reduction of the calcium influx (Lee et al., 2002). In another study (Nai et al., 2010), D1 receptor activation in hippocampal slices increased the NMDA-dependent long-term potentiation, as previously reported (Huang and Kandel, 1995). This effect could be abolished by disrupting the heteromer with specific interfering peptides. This interaction can have important functional consequence on cognition, as disruption of the D1–NMDA complex led to working memory impairment (Nai et al., 2010).
Another reported interaction with NMDA receptors is between D2 receptors and GluN2B (Liu et al., 2006). It has been demonstrated that in both the dorsal striatum and nucleus accumbens, D2 receptors and GluN2B are clustered in the PSD and form a complex that is more prevalent upon systemic cocaine treatment. Cocaine is also able to produce a selective decrease in the phosphorylation of Ser1303 on the GluN2B subunit. This decrease is D2 receptor-dependent, blocked by antagonist treatment, enhanced by agonist treatment and mediated by CaMKII. Cocaine treatment induces a D2 receptor-dependent decrease in CaMKII activity, and its association with GluN2B results in the decrease of phosphorylation of Ser1303. Functionally, cocaine application to striatal neurons causes a decrease in NMDA-mediated currents, an effect that can be abolished by heteromer disruption using a selective peptide that prevents the binding between the third intracellular loop of D2 receptors and the carboxy-terminal tail of GluN2B. Furthermore, disruption of the D2-GluN2B heteromer prevents phosphorylation of GluN2B and reduces cocaine-stimulated locomotor activity.
Adenosine receptor/dopamine receptor complexes
Many studies have shown that adenosine and dopamine exert opposing effects in the basal ganglia, with adenosine receptor agonists generally suppressing motor response and antagonists inducing motor activation (Ferre et al., 1997). Because the adenosine A1 receptor is expressed mainly in striato-nigral neurons, which also express dopamine D1 receptors, and the adenosine A2A receptor is expressed predominantly in striato-pallidal neurons, which express D2 receptors, it is suggested that A1-D1 and A2A-D2 heteromers might exist and have discrete localization in the basal ganglia (Fuxe et al., 2010). Thus, heterodimer formation has been reported and extensively characterized for the A1–D1 complex (Gines et al., 2000; Toda et al., 2003) and the A2A–D2 complex (Hillion et al., 2002; Canals et al., 2003) using different biophysical approaches. In both complexes, both A1 and A2A receptor activation can antagonize the cAMP responses upon either D1 or D2 receptor stimulation in transfected cells (Ferre et al., 1991; Gines et al., 2000). For these reasons, it has been proposed that these complexes could be of importance as novel approach to treat some dopamine-related diseases such as Parkinson's disease and schizophrenia. Particularly, A2A receptor antagonists, which can act as potentiators of the dopamine response, could be applied as adjunct therapy with D2 receptor agonists and L-DOPA for the locomotor impairments present in Parkinson's disease patients (Fuxe et al., 2007). In fact, A2A receptor antagonists have been shown to improve motor deficits in several animal models of Parkinson's disease as well as demonstrate anti-parkinsonian properties in clinical trials (Fuxe et al., 2008) and reduce L–DOPA-induced dyskinesia (Chase et al., 2003). At the same time, A2A receptor agonists showed an antipsychotic-like profile in different schizophrenia models, both in rodents (Rimondini et al., 1997) and non-human primates (Andersen et al., 2002). The mechanism proposed on the basis of these effects is the ability of A2A receptor agonists to reduce D2 receptor agonist binding and G protein coupling, particularly in the nucleus accumbens. Because the A2A–D2 heteromer can internalize as one unit, these agonists can also reduce D2 receptor availability at the plasma membrane (Hillion et al., 2002).
Other dopamine receptor heterodimers
Another example how dopamine receptors can alter their signalling pathways comes from a study on the D2 -ghrelin receptor heterodimer (Kern et al., 2012). The ghrelin receptor is a GPCR expressed in different regions of the brain that can be activated by the stomach peptide ghrelin (Kojima et al., 1999). In the hypothalamus, the ghrelin receptor and D2 receptors are co-expressed, and using time-resolved FRET experiments, it has been shown that these two receptors form a heterodimer in native tissue (Kern et al., 2012). Moreover, when co-expressed with the ghrelin receptor, D2 receptors are able to elicit a calcium response dependent on PLC activation, IP3 receptors and intracellular calcium stores. Interestingly, Gαq is not responsible for this increase, but rather the Gβγ subunit, derived from Gαi activation, mediates this effect likely through the direct association with GRK2 and direct stimulation of PLC (Inglese et al., 1994; Koch et al., 1994). This heterodimer formation appears to have an important role in appetite, as the suppression of food intake induced by a dopamine D2 receptor agonist was prevented by a ghrelin antagonist and was absent in ghrelin-KO mice, demonstrating the importance of this heterodimer in physiology.
One interesting example how dimerization can alter the physiological functions of each protomer is the case of the D4-β1 adrenoceptor and D4-α1B adrenoceptor heteromers (Gonzalez et al., 2012a). D4 receptors are highly expressed in the pineal gland, and their expression follows a circadian rhythm, being high at the beginning of the light cycle and low at the end (Bai et al., 2008). The role of the pineal gland is to translate light inputs from the retina by producing and secreting melatonin, a product of serotonin. In this brain region, β1 and α1B adrenoceptors are the primary receptors that control this mechanism. In transfected cells, Gonzalez and colleagues proved the existence of these two heterodimers using BRET, and they also used a proximity ligation assay to demonstrate their presence in vivo in the pineal gland. Similar to the circadian nature of D4 receptors, these complexes were found only at the beginning of the light cycle (Gonzalez et al., 2012a). A distinctive biochemical property of these heterodimers was the modulation of ERK and Akt activity. When co-expressed in the same cells or when naturally present in native tissue, treatment with a D4 receptor agonist was able to reduce ERK and Akt phosphorylation induced by β1 and α1B adrenergic receptors. Similarly, D4 receptor antagonists prevent MAPK and Akt activation induced by β1 or α1B adrenoceptors, and adrenoceptor antagonists could block D4 receptor stimulation of these pathways, showing a cross-antagonist property of this heterodimer. This functional interaction between D4, β1 and α1B receptors was also reflected by the production and release of serotonin and melatonin, with a circadian regulation mediated by D4 receptors and its modulation of β1 and α1B adrenoceptor activity (Gonzalez et al., 2012a).
One GPCR that has emerged as a novel modulator of the dopamine system is the trace amine-associated receptor 1 (TA1, known also as TAAR1) (Borowsky et al., 2001; Sotnikova et al., 2009; Espinoza et al., 2011). The TA1 receptor is expressed in several areas innervated by dopaminergic terminals such as the basal ganglia and frontal cortex, as well as in regions containing monoaminergic nuclei, including dopaminergic neurons in the ventral tegmental area (Lindemann et al., 2008). Several studies have shown that the TA1 receptor can modulate dopaminergic activities such as D2 receptor function and the firing rate of dopaminergic neurons (Lindemann et al., 2008; Bradaia et al., 2009; Espinoza et al., 2011). Moreover, TA1 receptor agonists have been shown to influence effects of a wide range of dopaminergic agents and related behaviours such as amphetamine-induced hyperactivity (Revel et al., 2011) and cocaine self-administration (Revel et al., 2012), and are effective as antipsychotic compounds in several models of schizophrenia against positive, negative and cognitive symptoms (Revel et al., 2013). One mechanism through which TA1 receptors can influence the dopamine system is by forming a heterodimer with D2 receptors. Using BRET, it has been demonstrated that TA1 receptors can form a constitutive heterodimer with D2, but not with D1 receptors (Espinoza et al., 2011; 2013), and that this heteromer can be disrupted by the D2 receptor antagonist haloperidol. This functional interaction exerts its effect in modulating the cAMP pathway and haloperidol could potentiate TA1 receptor-mediated cAMP signalling in cells. Furthermore, haloperidol's effects, such as c-Fos induction in the striatum and catalepsy, were reduced in TA1 receptor-KO mice indicating prominent interaction between D2 and TA1 receptors at the levels of postsynaptic structures (Espinoza et al., 2011). The D2–TA1 receptor interaction can also play an important role in presynaptic regulation of dopaminergic transmission by modulating D2 receptor autoreceptor functions via TA1 receptors (Leo et al., 2014).
Other developments and emerging trends
Newly identified functions mediated by dopamine receptors
Recent studies that often employed newly developed approaches and tools have revealed novel functions mediated by dopamine receptors and clarified the contribution of specific subtypes to previously established functions. Several observations regarding the functional role of dopamine receptors have been made using transgenic techniques. In addition to gene KO studies, which have been described in previous reviews (Sibley, 1999; Holmes et al., 2004; Beaulieu and Gainetdinov, 2011), mice overexpressing D2 and D3 receptors have been recently characterized. Particularly, mice selectively overexpressing D2 receptors in the striatum have persistent abnormalities in prefrontal cortex function and deficits in working memory and behavioural flexibility (Kellendonk et al., 2006), as well as motivation (Simpson et al., 2012) and timing precision (Ward et al., 2009) deficits that are often found of schizophrenia models. These mutants also demonstrated altered dendritic morphology of medium spiny neurons via involvement of Kir2 channels (Cazorla et al., 2012), and a deficit in inhibitory GABA-mediated transmission and dopamine sensitivity in the prefrontal cortex (Li et al., 2011b). Taken together, these studies indicated an important role for the striatal processes in the pathogenesis of the cognitive symptoms of schizophrenia. Based on this model, overstimulation or excessive D2 receptor activity in the striatum leads to altered functioning of prefrontal cortex neurons via several mechanisms, culminating in deficiencies in executive function and working memory – key components of schizophrenia-related deficits (Simpson et al., 2010). In contrast, mice overexpressing D3 receptors in the striatum have less pronounced, but still significant phenotype. They do not demonstrate cognitive deficits, but show disrupted motivation, suggesting that targeting D3 receptors may have effect on motivational symptoms, which are not improved by the currently available antipsychotics in schizophrenic patients (Simpson et al., 2014).
In another study, a role of subpopulation of D1 receptor-expressing cholinergic neurons in putative antipsychotic action of the M4 muscarinic receptor agonist xanomeline was demonstrated (Dencker et al., 2011). In mutant mice lacking the M4 muscarinic receptors only in D1 dopamine receptor-expressing cells, the antipsychotic-like effects of xanomeline were completely abolished suggesting that M4 muscarinic receptors co-localized with D1 receptors are involved.
Several recent observations have indicated that the direct modulation of the DAT function by D2 receptors might occur (Chen et al., 2013) and this interaction has physiological or pathological relevance (Bowton et al., 2010; Owens et al., 2012); however, no alterations in DAT function were found in D2 autoreceptor KO mice (Bello et al., 2011). Based on studies involving mice that constitutively express only the short isoform D2S or lacking both isoforms, it has been proposed that D2L is the major postsynaptic isoform expressed in medium spiny GABA neurons while D2S is predominantly expressed on presynaptic terminals and involved in autoreceptor function (Lindgren et al., 2003). However, recent study with virus-mediated receptor restoration in D2 receptor KO mice has indicated that both these alternatively spliced forms of the D2 receptors are equally capable of acting as postsynaptic receptors and autoreceptors (Neve et al., 2013). Interestingly, while the role of presynaptic D2 receptors in autoreceptor regulation of dopamine neuron firing rate, as well as dopamine synthesis and release is well established (Bello et al., 2011), a recent study involving cell-specific KO mice has shown a significant contribution of postsynaptic D2 receptors in the local feedback regulation of dopaminergic transmission in the dorsal striatum as well (Anzalone et al., 2012). As D3 receptors are also shown to contribute to the regulation of dopamine release and are expressed both at the presynaptic terminals and postsynaptic structures (Gainetdinov et al., 1996; Joseph et al., 2002; Gross et al., 2013), it would be of interest to explore if postsynaptic D3 receptors could be involved in the regulation of presynaptic transmission via similar local feedback mechanisms.
Application of optogenetic techniques has allowed for more precise anatomical and cellular dissection of the role of specific dopamine receptors in dopamine-related functions. An important advance in understanding the complex interplay of basal ganglia output by direct and indirect pathway projection neurons in regulating movement has been achieved (Kravitz et al., 2010; Freeze et al., 2013). Application of optogenetic and pharmacological approaches in transgenic mice has highlighted an important role of striatal D2 receptors in the concerted balance of the striatal output system and structural plasticity (Cazorla et al., 2014). Distinct roles for striatal neurons in the direct and indirect pathways in reinforcement have been shown by demonstrating that optogenetic stimulation of D1 receptor-expressing neurons induce persistent reinforcement whereas stimulation of D2 receptor-expressing neurons induce transient punishment (Kravitz et al., 2012). The role of medial prefrontal D1 receptor-expressing neurons in the control of food intake (Land et al., 2014) and temporal control (Narayanan et al., 2012) has also been demonstrated. Recent evidence indicates that optogenetic inhibition of D1 but not D2 receptor-expressing medium spiny neurons in the nucleus accumbens alters cocaine-mediated regulation of the T-lymphoma invasion and metastasis 1 (Tiam1) protein, which has been implicated in structural and synaptic plasticity (Chandra et al., 2013). Furthermore, using a combination of optogenetic and chemogenetic approaches based on designer receptors exclusively activated by designer drugs (DREADD) technology (Lee et al., 2013), it has been shown that strengthening the accumbal indirect pathway via D2 receptors promotes resilience to compulsive cocaine use (Bock et al., 2013).
A role of D2 receptor-related signalling in suppressing human osteoclastogenesis has been recently identified (Hanami et al., 2013), which may provide a plausible mechanism for the skeletal effects of antipsychotics observed in children and adolescents (Calarge et al., 2013). A complex role of D1 and D2 receptors has been demonstrated for epileptogenesis (Bozzi and Borrelli, 2013), and cohlear functions (Maison et al., 2012). Evidence gained from humans and mice also indicates that D4 receptors may contribute to longevity (Grady et al., 2013). Studies involving D4 receptor KO mice and selective D4 receptor agonists and antagonists in animal models of drug-seeking and drug-taking behaviours have suggested that treatments based on antagonism of the D4 receptor may be effective approaches for the management of psychostimulant and nicotine abuse (Di Ciano et al., 2014). That presynaptic D2 receptors might be involved in cocaine- and nicotine-induced structural plasticity in mesencephalic dopaminergic neurons via mechanism involving ERK and Akt signalling is also intriguing (Collo et al., 2012; 2013). A novel mechanism for D1 receptor-mediated regulation of ERK signalling involving the tyrosine phosphatase Shp-2, that is required for ERK activation by tyrosine kinase receptors, has been recently shown (Fiorentini et al., 2011), and this mechanism appears to be involved in L–DOPA-induced dyskinesia in the experimental model of Parkinson's disease (Fiorentini et al., 2013).
Recent evidence also indicated that dopamine receptors are involved in immune system regulation and processes related to inflammation and autoimmune reactions. It has been shown that astrocytic D2 receptors modulate innate immunity through αB-crystallin, which results in suppression of neuroinflammation (Shao et al., 2013). A role of D2 receptors in renal inflammation has been demonstrated as well (Zhang et al., 2012). It has been also observed that stimulation of D5 receptors expressed on dendritic cells potentiates Th17-mediated immunity, thus indicating that D5 receptors are able to modulate the development of an autoimmune response in vivo (Prado et al., 2012).
Although the role of retinal dopamine and dopamine receptors in the regulation of vision and related processes is well established, recent studies have uncovered previously unappreciated mechanisms. Studies involving KO mice lacking specific dopamine receptors have shown that in addition to the role of dopamine in suppressing rod-driven signals in bright light, it also enhances the same signals under dim illumination via D1 receptor-dependent sensitization of rod bipolar cells by GABA (Herrmann et al., 2011). A role of both D1 and D4 receptors in various dimensions of light-adapted vision has been demonstrated as well (Jackson et al., 2012). In a mouse model of type 1 diabetes, which develops early visual dysfunction because of dopamine deficiencies, acute treatment with either D1 or D4 receptor agonists improved overall retinal and visual function (Aung et al., 2014). Retinal dopamine plays a critical role in the development of myopia predominantly via the D2 receptor, but there is recent evidence suggesting that the balance of D2 and D1 receptor activation is important (Feldkaemper and Schaeffel, 2013). Evidence from KO mice suggests that not only retinal, but also central dopamine could play a role in the regulation of retinal function via differential involvement of D1 and D2 receptors (Lavoie et al., 2014a). Furthermore, D2 receptor-regulated GSK3 signalling also appears to contribute to electroretinogram anomalies observed in subjects at high genetic risk for schizophrenia and bipolar disorder, suggesting that electroretinograms can serve as a biomarker for central dopamine abnormalities related to psychiatric disorders (Lavoie et al., 2014b).
It is well known that dopamine acting through D2 receptors controls lactotroph proliferation and prolactin levels; however, the role of the D2S and D2L receptor isoforms was unknown. Recent investigations have shown that the presence of either the D2S or D2L isoforms in vivo prevents hyperprolactinaemia, the development of lactotroph hyperplasia, and tumourigenesis, all of which are observed when both isoforms are deleted in mice (Radl et al., 2013). However, the protective function of a single D2 receptor isoform is overridden when single isoform-KO mice are challenged by chronic estrogen treatments, suggesting that signalling from both isoforms is necessary in conditions that simulate pathological states (Radl et al., 2013).
Selective disruption of D2 receptors in pituitary lactotrophes in conditional mutant mice results in an increase in body weight and adiposity (Perez Millan et al., 2014), indicating that D2 receptors might be involved in the metabolic side effects of antipsychotic drug treatment such as obesity, cardiovascular complications and increased incidence of type 2 diabetes (Reynolds and Kirk, 2010). Interestingly, the D2 receptor agonist bromocriptine has been recently approved for clinical use for the management of type 2 diabetes mellitus, particularly in forms associated with cardiovascular deficits (Grunberger, 2013).
The general role of D2 receptors and dopamine in endocrine tumours is supported by reports that D2 receptor agonists are effective in controlling hormone secretion and cell proliferation in experimental studies (Gatto and Hofland, 2011). Furthermore, dopamine agonists have been found to be efficacious in a subgroup of patients with pituitary adenomas and a few reported cases of carcinoids (Gatto and Hofland, 2011). It has also been shown that dopamine, acting via D2 receptors, blocks stress-mediated ovarian carcinoma growth (Moreno-Smith et al., 2011) in part through the antiangiogenic effect of dopamine and in part through D1 receptors that stimulates vessel stabilization by increasing pericyte recruitment to tumour endothelial cells (Moreno-Smith et al., 2013). Interestingly, recent unbiased screening attempts to identify novel anti-cancer therapies yielded dopaminergic compounds such as the D2 receptor antagonist thioridazine, which selectively targets cancer stem cells by inducing differentiation to overcome neoplastic self-renewal (Sachlos et al., 2012), and perphenazine, which was found to be effective in T-cell acute leukaemia by inducing PP2A-mediated apoptosis (Gutierrez et al., 2014).
Influence on glutamate and GABA transmission
One recurrent topic that has received significant attention recently is related to signalling mechanisms involved in the convergence of dopamine receptor-mediated signalling with glutamatergic and GABA neurotransmission. It is well known that, as a slow neurotransmitter, dopamine exerts its actions through the modulation of the effects of fast neurotransmitters such as glutamate and GABA. This multi-transmitter interaction is critical for many vital functions mediated by dopamine as well as related disorders such as schizophrenia. As presented in Figures 1 and 2, both D1 and D2 receptors have multiple potential signalling mechanisms that may be involved in this modulation. Dopamine receptors can alter the phosphorylation of critical subunits of glutamate AMPA receptors (GluA1) and NMDA receptors (GluN2B), via the Gα subunit-mediated cAMP-PKA-DARPP32 signalling cascade (Dell'anno et al., 2013; Hobson et al., 2013; Song et al., 2013; Flores-Barrera et al., 2014; Murphy et al., 2014), as well as Gαq and Gβγ subunit-mediated PLC signalling and subsequent Ca2+-dependent events (Jenkins and Traynelis, 2012; Jenkins et al., 2014). Activation of the G protein-independent Akt/GSK3 signalling cascade by D2 receptors could lead to significant alterations in glutamatergic signalling mediated by NMDA and AMPA receptors (Li et al., 2009; Li and Gao, 2011a) as well as changes in kinesin-mediated AMPA receptor trafficking (Du et al., 2010). Similar mechanisms can also affect GABA transmission (Li et al., 2012). As discussed earlier, the direct interaction of dopamine receptors with GluN1 and GluN2B subunits of NMDA receptors has been demonstrated (Fiorentini et al., 2003; Liu et al., 2006). Several mechanisms of direct and indirect interaction of D3 receptors with glutamate neurotransmission have also been documented (Sokoloff et al., 2013).
Further understanding of the mechanisms of the intricate interaction of dopamine receptor-mediated signalling events with glutamate and GABA signalling could manifest into exciting new molecular targets for novel pharmacological approaches to dopamine-related disorders.
Search for biased ligands of dopamine receptors
The growing realization of the complexity of signalling mediated by GPCRs in general, and dopamine receptors in particular, has provided a theoretical framework for the development of pathway-specific biased ligand pharmacology of D2 receptors (Beaulieu et al., 2007a; Beaulieu and Gainetdinov, 2011). In addition to the studies described earlier (Masri et al., 2008; Allen et al., 2011), a structure–activity analysis of pathway-specific biased agonism at D2 receptors based on aripiprazole derivatives (Chen et al., 2012) and search for novel potential antipsychotic cariprazine derivatives (Shonberg et al., 2013) and other D2 receptor targeting compounds (Hiller et al., 2013) have been performed. These studies have recently resulted in identification of several functionally selective D2 receptor agonists that are biased towards G protein- versus βArr2-dependent signalling (Free et al., 2014; Möller et al., 2014). Future characterization of the biochemical and behavioural effects of these biased compounds could eventually result in the development of improved antipsychotic drugs with reduced propensity of undesirable side effects. It should be noted that although the majority of current studies are focused on the development of biased ligands targeting either G protein-dependent or βArr2-dependent signalling mechanisms, it might be expected that biased ligands that, for example, will specifically target one, but not another modality of G protein-independent signalling could be identified in future (Figures 1 and 2).
Allosteric modulators of dopamine receptors
Another exciting direction in the pharmacology of GPCRs that is gaining attention recently is related to the development of allosteric modulators (Nickols and Conn, 2014). Although classic drug discovery approaches targeting GPCRs have traditionally focused on developing ligands for orthosteric sites, which bind endogenous ligands, more recent efforts have been aimed at modulating receptor function via allosteric modulators, which target a site distinct from the orthosteric site. It is expected that the development of such positive or negative allosteric modulators or ‘bitopic’ ligands that interact with both the allosteric and the orthosteric sites could eventually result in increased drug selectivity for therapeutic action and potentially decreased adverse side effects. Thus, a specific allosteric modulator of D2 receptors, the peptidomimetic 3(R)-[(2(S)-pyrrolidinylcarbonyl) amino]-2-oxo-1-pyrrolidineacetamide (PAOPA), has been recently characterized as binding to a site on the D2 receptor that is distinct from the endogenous ligand binding site (Tan et al., 2013). PAOPA can influence several signalling and cellular events characteristic of D2 receptor activation (Basu et al., 2013), and in preclinical models, can attenuate schizophrenia-like behavioural manifestations without causing significant motor abnormalities common with many current antipsychotics (Tan et al., 2013).
Based on the crystal structure of D3 receptors (Chien et al., 2010) a virtual screen targeting allosteric sites of this receptor has been recently performed (Lane et al., 2013). This screening resulted in the identification of a number of allosteric ligands for D3 receptors, including chemically diverse compounds with a variety of functional activity profiles and high affinities and ligand efficiencies. It is believed that identification of the allosteric structural features of D3 receptors that are essential to selectivity and efficacy could be critical for the identification of highly selective D3 receptor ligands, which are notorious for their difficulty in development (Newman et al., 2012). Allosteric modulation of certain ligands can also occur at the level of receptor heterodimers as demonstrated by the ability of a newly identified dopamine agonist to negatively modulate adenosine A2A receptor binding properties by interacting with the A2A–D2 receptor heteromer (Trincavelli et al., 2012). Interestingly, A2A receptor agonist-induced modulation of D2 receptor agonist-induced βArr2 recruitment has been described, suggesting the involvement of a possible A2A–D2–βArr2 complex in this allosteric modulation (Borroto-Escuela et al., 2011a).
New dopamine receptor-based drugs
In addition to the clinically approved compounds targeting dopamine receptors that were discussed in our recent review (Beaulieu and Gainetdinov, 2011), several new compounds have either been approved for clinical use or are in the last stages of clinical trials. Essentially all of these compounds are antipsychotics targeting D2 receptors. It should be noted that no clinically approved antipsychotic drugs exist to date that are not D2 receptor blockers. Among the newer antipsychotics recently approved for the treatment of schizophrenia, the most conspicuous are iloperidone, asenapine, lurasidone and blonanserin, considered to be atypical antipsychotics (George et al., 2013). These compounds at least partially target D2 receptors with additional influence on other receptors, particularly 5-HT2A receptors. Although they exert clear antipsychotic activity, essentially all of them produce metabolic side effects and hyperprolactinaemia, which require appropriate monitoring (Wang et al., 2014). In addition to the olanzapine-fluoxetine combination, quetiapine, lurasidone is now FDA-approved for the acute treatment of bipolar depression (McIntyre et al., 2013). Among several emerging antipsychotic compounds, cariprazine has attracted the most attention because this drug has partial agonist activity at D2 and D3 receptors, with a six- to eightfold higher affinity for the human D3 receptor over the D2 receptor. Cariprazine is at latest stages of development, and an application has been submitted to the FDA for approval as a treatment for schizophrenia, bipolar mania and depression (Veselinovic et al., 2013). Another interesting compound is brexpiprazole, a novel drug candidate in clinical development for psychiatric disorders with high affinity for D2 dopamine, 5-HT2A and adrenoceptors (Maeda et al., 2014). Brexpiprazole was showing significant antipsychotic-like properties in several preclinical models suggesting that brexpiprazole is a 5-HT-dopamine activity modulator with a unique pharmacology that might provide novel treatment option for psychiatric disorders.
D2 receptor antibodies in autoimmune encephalitis
One exciting direction in the current neuropsychiatry is the emergence of novel class of disorders related to autoantibodies against critical synaptic proteins such as NMDA receptors and D2 receptors (Lancaster and Dalmau, 2012). Remarkably, some patients with these autoantibodies have symptoms virtually indistinguishable from schizophrenia or other neuropsychiatric or movement disorders. Antibodies to surface D2 receptors were found in patients with autoimmune movement and psychiatric disorders (Dale et al., 2012). D2 receptor autoantibodies were found also in patients with Sydenham chorea (Cox et al., 2013), and recent study reported that herpes simplex encephalitis relapse with chorea was associated with autoantibodies against D2 receptors and the NMDA receptors (Mohammad et al., 2014). This recently emerging field is growing monthly, with more patients identified, and it is expected that the autoimmune theory of neuropsychiatric and movement disorders will have significant support in the future.
Conclusions
The studies described here show the significant progress made in understanding dopamine receptor functions, the complexity of their signalling mechanisms and potential new applications of dopamine receptor-based pharmacological strategies. Using a variety of the most up-to-date approaches, multidimensional analysis of dopamine receptor biology will eventually provide an opportunity for the precise targeting of desired components of post-receptor intracellular processes either via receptor-related mechanisms or post-receptor signalling cascades, thereby providing an exciting opportunity to target pathological processes with minimal propensity of developing side effects. Such approaches involving ‘biased agonism’, allosteric-based targeting of receptors and heteromers and downstream intracellular signalling events could eventually result in emergence of a new generation of dopamine receptor-based therapies for a variety of dopamine-related disorders.
Acknowledgments
This work was supported by funding from the Canadian Institutes of Health Research (CIHR) (Grant NSA 93798) and a discovery grant from the Natural Sciences and Engineering Research Council of Canada (NSERC) awarded to J. M. B. This work was also supported by research funding awarded to R. R. G. from the F. Hoffmann – La Roche, Ltd. (Basel, Switzerland) and an internal St. Petersburg State University (St. Petersburg, Russia) grant #1.38.201.2014. J. M. B. holds a Canada research Chair in Molecular Psychiatry.
Glossary
- βArr2
β-arrestin 2
- BDNF
brain-derived neurotrophic factor
- CaMKII
Ca2+/calmodulin-dependent PK II
- CDK5
cyclin-dependent kinase 5
- DAT
dopamine transporter
- DARPP-32
dopamine and cAMP-regulated phosphoprotein, 32 kDa
- GluA1
glutamate receptor, ionotropic, AMPA 1 subunit
- GluN2B
glutamate receptor, ionotropic, NMDA 2B subunit
- GIRKs
G protein coupled inwardly rectifying potassium channels
- GSK3
glycogen synthase kinase
- HTT
huntingtin
- KO
knockout
- IP3
inositol trisphosphate
- PDK
phosphatidylinositol-dependent kinase
- PP1
protein phosphatase 1
- PP2A
protein phosphatase 2A
- rpS6
ribosomal protein S6
- RTK
receptor tyrosine kinases
- TCS1/2
tuberous sclerosis proteins 1 and 2
- TrkB
neurotrophic tyrosine kinase, receptor, type 2
Author contributions
All authors contributed equally to this work.
Conflict of interest
The authors declare no conflict of interest in writing this paper.
References
- Albizu L, Cottet M, Kralikova M, Stoev S, Seyer R, Brabet I, et al. Time-resolved FRET between GPCR ligands reveals oligomers in native tissues. Nat Chem Biol. 2010;6:587–594. doi: 10.1038/nchembio.396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alessi DR, Andjelkovic M, Caudwell B, Cron P, Morrice N, Cohen P, et al. Mechanism of activation of protein kinase B by insulin and IGF-1. EMBO J. 1996;15:6541–6551. [PMC free article] [PubMed] [Google Scholar]
- Alexander SP, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: G protein-coupled receptors. Br J Pharmacol. 2013a;170:1459–1581. doi: 10.1111/bph.12445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The concise guide to PHARMACOLOGY 2013/14: enzymes. Br J Pharmacol. 2013b;170:1797–1867. doi: 10.1111/bph.12451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: Transporters. Br J Pharmacol. 2013c;170:1706–1796. doi: 10.1111/bph.12450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The concise guide to PHARMACOLOGY 2013/14: catalytic receptors. Br J Pharmacol. 2013d;170:1676–1705. doi: 10.1111/bph.12449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Catterall WA, et al. The concise guide to PHARMACOLOGY 2013/14: ion channels. Br J Pharmacol. 2013e;170:1607–1651. doi: 10.1111/bph.12447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The concise guide to PHARMACOLOGY 2013/14: ligand-gated ion channels. Br J Pharmacol. 2013f;170:1582–1606. doi: 10.1111/bph.12446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Allen JA, Yost JM, Setola V, Chen X, Sassano MF, Chen M, et al. Discovery of beta-arrestin-biased dopamine D2 ligands for probing signal transduction pathways essential for antipsychotic efficacy. Proc Natl Acad Sci U S A. 2011;108:18488–18493. doi: 10.1073/pnas.1104807108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Altar CA, Hunt RA, Jurata LW, Webster MJ, Derby E, Gallagher P, et al. Insulin, IGF-1, and muscarinic agonists modulate schizophrenia-associated genes in human neuroblastoma cells. Biol Psychiatry. 2008;64:1077–1087. doi: 10.1016/j.biopsych.2008.08.031. [DOI] [PubMed] [Google Scholar]
- Andersen MB, Fuxe K, Werge T, Gerlach J. The adenosine A2A receptor agonist CGS 21680 exhibits antipsychotic-like activity in Cebus apella monkeys. Behav Pharmacol. 2002;13:639–644. doi: 10.1097/01.fbp.0000047148.28986.67. [DOI] [PubMed] [Google Scholar]
- Anzalone A, Lizardi-Ortiz JE, Ramos M, De Mei C, Hopf FW, Iaccarino C, et al. Dual control of dopamine synthesis and release by presynaptic and postsynaptic dopamine D2 receptors. J Neurosci. 2012;32:9023–9034. doi: 10.1523/JNEUROSCI.0918-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aung MH, Park HN, Han MK, Obertone TS, Abey J, Aseem F, et al. Dopamine deficiency contributes to early visual dysfunction in a rodent model of type 1 diabetes. J Neurosci. 2014;34:726–736. doi: 10.1523/JNEUROSCI.3483-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bai L, Zimmer S, Rickes O, Rohleder N, Holthues H, Engel L, et al. Daily oscillation of gene expression in the retina is phase-advanced with respect to the pineal gland. Brain Res. 2008;1203:89–96. doi: 10.1016/j.brainres.2008.01.073. [DOI] [PubMed] [Google Scholar]
- Barbeau A, Godin AG, Swift JL, De Koninck Y, Wiseman PW, Beaulieu JM. Quantification of receptor tyrosine kinase activation and transactivation by G–protein-coupled receptors using spatial intensity distribution analysis (SpIDA) Methods Enzymol. 2013;522:109–131. doi: 10.1016/B978-0-12-407865-9.00007-8. [DOI] [PubMed] [Google Scholar]
- Basu D, Tian Y, Bhandari J, Jiang JR, Hui P, Johnson RL, et al. Effects of the dopamine D2 allosteric modulator, PAOPA, on the expression of GRK2, arrestin-3, ERK1/2, and on receptor internalization. PLoS ONE. 2013;8:e70736. doi: 10.1371/journal.pone.0070736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beaulieu JM. A role for Akt and glycogen synthase kinase-3 as integrators of dopamine and serotonin neurotransmission in mental health. J Psychiatry Neurosci. 2012;37:7–16. doi: 10.1503/jpn.110011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beaulieu JM, Caron MG. Looking at lithium: molecular moods and complex behaviour. Mol Interv. 2008a;8:230–241. doi: 10.1124/mi.8.5.8. [DOI] [PubMed] [Google Scholar]
- Beaulieu JM, Gainetdinov RR. The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol Rev. 2011;63:182–217. doi: 10.1124/pr.110.002642. [DOI] [PubMed] [Google Scholar]
- Beaulieu JM, Julien JP. Peripherin-mediated death of motor neurons rescued by overexpression of neurofilament NF-H proteins. J Neurochem. 2003;85:248–256. doi: 10.1046/j.1471-4159.2003.01653.x. [DOI] [PubMed] [Google Scholar]
- Beaulieu JM, Sotnikova TD, Yao WD, Kockeritz L, Woodgett JR, Gainetdinov RR, et al. Lithium antagonizes dopamine-dependent behaviors mediated by an AKT/glycogen synthase kinase 3 signaling cascade. Proc Natl Acad Sci U S A. 2004;101:5099–5104. doi: 10.1073/pnas.0307921101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beaulieu JM, Sotnikova TD, Marion S, Lefkowitz RJ, Gainetdinov RR, Caron MG. An Akt/beta-arrestin 2/PP2A signaling complex mediates dopaminergic neurotransmission and behavior. Cell. 2005;122:261–273. doi: 10.1016/j.cell.2005.05.012. [DOI] [PubMed] [Google Scholar]
- Beaulieu JM, Gainetdinov RR, Caron MG. The Akt-GSK-3 signaling cascade in the actions of dopamine. Trends Pharmacol Sci. 2007a;28:166–172. doi: 10.1016/j.tips.2007.02.006. [DOI] [PubMed] [Google Scholar]
- Beaulieu JM, Tirotta E, Sotnikova TD, Masri B, Salahpour A, Gainetdinov RR, et al. Regulation of Akt signaling by D2 and D3 dopamine receptors in vivo. J Neurosci. 2007b;27:881–885. doi: 10.1523/JNEUROSCI.5074-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beaulieu JM, Marion S, Rodriguiz RM, Medvedev IO, Sotnikova TD, Ghisi V, et al. A beta-arrestin 2 signaling complex mediates lithium action on behavior. Cell. 2008b;132:125–136. doi: 10.1016/j.cell.2007.11.041. [DOI] [PubMed] [Google Scholar]
- Bello EP, Mateo Y, Gelman DM, Noain D, Shin JH, Low MJ, et al. Cocaine supersensitivity and enhanced motivation for reward in mice lacking dopamine D2 autoreceptors. Nat Neurosci. 2011;14:1033–1038. doi: 10.1038/nn.2862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berridge MJ. Inositol trisphosphate and calcium signalling mechanisms. Biochim Biophys Acta. 2009;1793:933–940. doi: 10.1016/j.bbamcr.2008.10.005. [DOI] [PubMed] [Google Scholar]
- Bertran-Gonzalez J, Bosch C, Maroteaux M, Matamales M, Herve D, Valjent E, et al. Opposing patterns of signaling activation in dopamine D1 and D2 receptor-expressing striatal neurons in response to cocaine and haloperidol. J Neurosci. 2008;28:5671–5685. doi: 10.1523/JNEUROSCI.1039-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bibb JA, Snyder GL, Nishi A, Yan Z, Meijer L, Fienberg AA, et al. Phosphorylation of DARPP-32 by Cdk5 modulates dopamine signalling in neurons. Nature. 1999;402:669–671. doi: 10.1038/45251. [DOI] [PubMed] [Google Scholar]
- Blank T, Nijholt I, Teichert U, Kugler H, Behrsing H, Fienberg A, et al. The phosphoprotein DARPP-32 mediates cAMP-dependent potentiation of striatal N-methyl-D-aspartate responses. Proc Natl Acad Sci U S A. 1997;94:14859–14864. doi: 10.1073/pnas.94.26.14859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blom H, Ronnlund D, Scott L, Spicarova Z, Rantanen V, Widengren J, et al. Nearest neighbor analysis of dopamine D1 receptors and Na(+)-K(+)-ATPases in dendritic spines dissected by STED microscopy. Microsc Res Tech. 2012;75:220–228. doi: 10.1002/jemt.21046. [DOI] [PubMed] [Google Scholar]
- Bock R, Shin JH, Kaplan AR, Dobi A, Markey E, Kramer PF, et al. Strengthening the accumbal indirect pathway promotes resilience to compulsive cocaine use. Nat Neurosci. 2013;16:632–638. doi: 10.1038/nn.3369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bohn LM, Gainetdinov RR, Sotnikova TD, Medvedev IO, Lefkowitz RJ, Dykstra LA, et al. Enhanced rewarding properties of morphine, but not cocaine, in beta(arrestin)-2 knock-out mice. J Neurosci. 2003;23:10265–10273. doi: 10.1523/JNEUROSCI.23-32-10265.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonito-Oliva A, Pallottino S, Bertran-Gonzalez J, Girault JA, Valjent E, Fisone G. Haloperidol promotes mTORC1-dependent phosphorylation of ribosomal protein S6 via dopamine- and cAMP-regulated phosphoprotein of 32 kDa and inhibition of protein phosphatase-1. Neuropharmacology. 2013;72:197–203. doi: 10.1016/j.neuropharm.2013.04.043. [DOI] [PubMed] [Google Scholar]
- Borowsky B, Adham N, Jones KA, Raddatz R, Artymyshyn R, Ogozalek KL, et al. Trace amines: identification of a family of mammalian G protein-coupled receptors. Proc Natl Acad Sci U S A. 2001;98:8966–8971. doi: 10.1073/pnas.151105198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Borroto-Escuela DO, Romero-Fernandez W, Tarakanov AO, Ciruela F, Agnati LF, Fuxe K. On the existence of a possible A2A–D2–beta-arrestin2 complex: A2A agonist modulation of D2 agonist-induced beta-arrestin2 recruitment. J Mol Biol. 2011a;406:687–699. doi: 10.1016/j.jmb.2011.01.022. [DOI] [PubMed] [Google Scholar]
- Borroto-Escuela DO, Van Craenenbroeck K, Romero-Fernandez W, Guidolin D, Woods AS, Rivera A, et al. Dopamine D2 and D4 receptor heteromerization and its allosteric receptor–receptor interactions. Biochem Biophys Res Commun. 2011b;404:928–934. doi: 10.1016/j.bbrc.2010.12.083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bowton E, Saunders C, Erreger K, Sakrikar D, Matthies HJ, Sen N, et al. Dysregulation of dopamine transporters via dopamine D2 autoreceptors triggers anomalous dopamine efflux associated with attention-deficit hyperactivity disorder. J Neurosci. 2010;30:6048–6057. doi: 10.1523/JNEUROSCI.5094-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bozzi Y, Borrelli E. The role of dopamine signaling in epileptogenesis. Front Cell Neurosci. 2013;7:157. doi: 10.3389/fncel.2013.00157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bradaia A, Trube G, Stalder H, Norcross RD, Ozmen L, Wettstein JG, et al. The selective antagonist EPPTB reveals TAAR1-mediated regulatory mechanisms in dopaminergic neurons of the mesolimbic system. Proc Natl Acad Sci U S A. 2009;106:20081–20086. doi: 10.1073/pnas.0906522106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brami-Cherrier K, Valjent E, Herve D, Darragh J, Corvol JC, Pages C, et al. Parsing molecular and behavioral effects of cocaine in mitogen- and stress-activated protein kinase-1-deficient mice. J Neurosci. 2005;25:11444–11454. doi: 10.1523/JNEUROSCI.1711-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bychkov E, Ahmed MR, Dalby KN, Gurevich EV. Dopamine depletion and subsequent treatment with L-DOPA, but not the long-lived dopamine agonist pergolide, enhances activity of the Akt pathway in the rat striatum. J Neurochem. 2007;102:699–711. doi: 10.1111/j.1471-4159.2007.04586.x. [DOI] [PubMed] [Google Scholar]
- Calarge CA, Ivins SD, Motyl KJ, Shibli-Rahhal AA, Bliziotes MM, Schlechte JA. Possible mechanisms for the skeletal effects of antipsychotics in children and adolescents. Ther Adv Psychopharmacol. 2013;3:278–293. doi: 10.1177/2045125313487548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Canals M, Marcellino D, Fanelli F, Ciruela F, de Benedetti P, Goldberg SR, et al. Adenosine A2A-dopamine D2 receptor-receptor heteromerization: qualitative and quantitative assessment by fluorescence and bioluminescence energy transfer. J Biol Chem. 2003;278:46741–46749. doi: 10.1074/jbc.M306451200. [DOI] [PubMed] [Google Scholar]
- Carlsson A. A paradigm shift in brain research. Science. 2001;294:1021–1024. doi: 10.1126/science.1066969. [DOI] [PubMed] [Google Scholar]
- Cazorla M, Shegda M, Ramesh B, Harrison NL, Kellendonk C. Striatal D2 receptors regulate dendritic morphology of medium spiny neurons via Kir2 channels. J Neurosci. 2012;32:2398–2409. doi: 10.1523/JNEUROSCI.6056-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cazorla M, de Carvalho FD, Chohan MO, Shegda M, Chuhma N, Rayport S, et al. Dopamine D2 receptors regulate the anatomical and functional balance of basal ganglia circuitry. Neuron. 2014;81:153–164. doi: 10.1016/j.neuron.2013.10.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chandra R, Lenz JD, Gancarz AM, Chaudhury D, Schroeder GL, Han MH, et al. Optogenetic inhibition of D1R containing nucleus accumbens neurons alters cocaine-mediated regulation of Tiam1. Front Mol Neurosci. 2013;6:13. doi: 10.3389/fnmol.2013.00013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chase TN, Bibbiani F, Bara-Jimenez W, Dimitrova T, Oh-Lee JD. Translating A2A antagonist KW6002 from animal models to parkinsonian patients. Neurology. 2003;61:S107–S111. doi: 10.1212/01.wnl.0000095223.08711.48. [DOI] [PubMed] [Google Scholar]
- Chen PC, Lao CL, Chen JC. Dual alteration of limbic dopamine D1 receptor-mediated signalling and the Akt/GSK3 pathway in dopamine D3 receptor mutants during the development of methamphetamine sensitization. J Neurochem. 2007;100:225–241. doi: 10.1111/j.1471-4159.2006.04203.x. [DOI] [PubMed] [Google Scholar]
- Chen R, Daining CP, Sun H, Fraser R, Stokes SL, Leitges M, et al. Protein kinase Cbeta is a modulator of the dopamine D2 autoreceptor-activated trafficking of the dopamine transporter. J Neurochem. 2013;125:663–672. doi: 10.1111/jnc.12229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen X, Sassano MF, Zheng L, Setola V, Chen M, Bai X, et al. Structure-functional selectivity relationship studies of beta-arrestin-biased dopamine D(2) receptor agonists. J Med Chem. 2012;55:7141–7153. doi: 10.1021/jm300603y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chi SS, Vetiska SM, Gill RS, Hsiung MS, Liu F, Van Tol HH. Transactivation of PDGFRbeta by dopamine D4 receptor does not require PDGFRbeta dimerization. Mol Brain. 2010;3:22. doi: 10.1186/1756-6606-3-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chien EY, Liu W, Zhao Q, Katritch V, Han GW, Hanson MA, et al. Structure of the human dopamine D3 receptor in complex with a D2/D3 selective antagonist. Science. 2010;330:1091–1095. doi: 10.1126/science.1197410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chun LS, Free RB, Doyle TB, Huang XP, Rankin ML, Sibley DR. D1–D2 dopamine receptor synergy promotes calcium signaling via multiple mechanisms. Mol Pharmacol. 2013;84:190–200. doi: 10.1124/mol.113.085175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Civelli O, Bunzow JR, Grandy DK. Molecular diversity of the dopamine receptors. Annu Rev Pharmacol Toxicol. 1993;33:281–307. doi: 10.1146/annurev.pa.33.040193.001433. [DOI] [PubMed] [Google Scholar]
- Collo G, Bono F, Cavalleri L, Plebani L, Merlo Pich E, Millan MJ, et al. Pre-synaptic dopamine D(3) receptor mediates cocaine-induced structural plasticity in mesencephalic dopaminergic neurons via ERK and Akt pathways. J Neurochem. 2012;120:765–778. doi: 10.1111/j.1471-4159.2011.07618.x. [DOI] [PubMed] [Google Scholar]
- Collo G, Bono F, Cavalleri L, Plebani L, Mitola S, Merlo Pich E, et al. Nicotine-induced structural plasticity in mesencephalic dopaminergic neurons is mediated by dopamine D3 receptors and Akt-mTORC1 signaling. Mol Pharmacol. 2013;83:1176–1189. doi: 10.1124/mol.113.084863. [DOI] [PubMed] [Google Scholar]
- Cox CJ, Sharma M, Leckman JF, Zuccolo J, Zuccolo A, Kovoor A, et al. Brain human monoclonal autoantibody from sydenham chorea targets dopaminergic neurons in transgenic mice and signals dopamine D2 receptor: implications in human disease. J Immunol. 2013;191:5524–5541. doi: 10.4049/jimmunol.1102592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cross DA, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature. 1995;378:785–789. doi: 10.1038/378785a0. [DOI] [PubMed] [Google Scholar]
- Dale RC, Merheb V, Pillai S, Wang D, Cantrill L, Murphy TK, et al. Antibodies to surface dopamine-2 receptor in autoimmune movement and psychiatric disorders. Brain. 2012;135:3453–3468. doi: 10.1093/brain/aws256. [DOI] [PubMed] [Google Scholar]
- Dell'anno MT, Pallottino S, Fisone G. mGlu5R promotes glutamate AMPA receptor phosphorylation via activation of PKA/DARPP-32 signaling in striatopallidal medium spiny neurons. Neuropharmacology. 2013;66:179–186. doi: 10.1016/j.neuropharm.2012.03.025. [DOI] [PubMed] [Google Scholar]
- Dencker D, Wortwein G, Weikop P, Jeon J, Thomsen M, Sager TN, et al. Involvement of a subpopulation of neuronal M4 muscarinic acetylcholine receptors in the antipsychotic-like effects of the M1/M4 preferring muscarinic receptor agonist xanomeline. J Neurosci. 2011;31:5905–5908. doi: 10.1523/JNEUROSCI.0370-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Ciano P, Grandy DK, Le Foll B. Dopamine D4 receptors in psychostimulant addiction. Adv Pharmacol. 2014;69:301–321. doi: 10.1016/B978-0-12-420118-7.00008-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Du J, Wei Y, Liu L, Wang Y, Khairova R, Blumenthal R, et al. A kinesin signaling complex mediates the ability of GSK-3beta to affect mood-associated behaviors. Proc Natl Acad Sci U S A. 2010;107:11573–11578. doi: 10.1073/pnas.0913138107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dumartin B, Caille I, Gonon F, Bloch B. Internalization of D1 dopamine receptor in striatal neurons in vivo as evidence of activation by dopamine agonists. J Neurosci. 1998;18:1650–1661. doi: 10.1523/JNEUROSCI.18-05-01650.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dziedzicka-Wasylewska M, Faron-Gorecka A, Andrecka J, Polit A, Kusmider M, Wasylewski Z. Fluorescence studies reveal heterodimerization of dopamine D1 and D2 receptors in the plasma membrane. Biochemistry. 2006;45:8751–8759. doi: 10.1021/bi060702m. [DOI] [PubMed] [Google Scholar]
- Dziedzicka-Wasylewska M, Faron-Gorecka A, Gorecki A, Kusemider M. Mechanism of action of clozapine in the context of dopamine D1–D2 receptor hetero-dimerization – a working hypothesis. Pharmacol Rep. 2008;60:581–587. [PubMed] [Google Scholar]
- Eguchi S, Numaguchi K, Iwasaki H, Matsumoto T, Yamakawa T, Utsunomiya H, et al. Calcium-dependent epidermal growth factor receptor transactivation mediates the angiotensin II-induced mitogen-activated protein kinase activation in vascular smooth muscle cells. J Biol Chem. 1998;273:8890–8896. doi: 10.1074/jbc.273.15.8890. [DOI] [PubMed] [Google Scholar]
- Emamian ES, Hall D, Birnbaum MJ, Karayiorgou M, Gogos JA. Convergent evidence for impaired AKT1-GSK3beta signaling in schizophrenia. Nat Genet. 2004;36:131–137. doi: 10.1038/ng1296. [DOI] [PubMed] [Google Scholar]
- Espinoza S, Salahpour A, Masri B, Sotnikova TD, Messa M, Barak LS, et al. Functional interaction between trace amine-associated receptor 1 and dopamine D2 receptor. Mol Pharmacol. 2011;80:416–425. doi: 10.1124/mol.111.073304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Espinoza S, Masri B, Salahpour A, Gainetdinov RR. BRET approaches to characterize dopamine and TAAR1 receptor pharmacology and signaling. Methods Mol Biol. 2013;964:107–122. doi: 10.1007/978-1-62703-251-3_8. [DOI] [PubMed] [Google Scholar]
- Felder CC, Jose PA, Axelrod J. The dopamine-1 agonist, SKF 82526, stimulates phospholipase-C activity independent of adenylate cyclase. J Pharmacol Exp Ther. 1989;248:171–175. [PubMed] [Google Scholar]
- Feldkaemper M, Schaeffel F. An updated view on the role of dopamine in myopia. Exp Eye Res. 2013;114:106–119. doi: 10.1016/j.exer.2013.02.007. [DOI] [PubMed] [Google Scholar]
- Ferguson SS, Barak LS, Zhang J, Caron MG. G–protein-coupled receptor regulation: role of G–protein-coupled receptor kinases and arrestins. Can J Physiol Pharmacol. 1996;74:1095–1110. doi: 10.1139/cjpp-74-10-1095. [DOI] [PubMed] [Google Scholar]
- Ferre S, Franco R. Oligomerization of G–protein-coupled receptors: a reality. Curr Opin Pharmacol. 2010;10:1–5. doi: 10.1016/j.coph.2009.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferre S, von Euler G, Johansson B, Fredholm BB, Fuxe K. Stimulation of high-affinity adenosine A2 receptors decreases the affinity of dopamine D2 receptors in rat striatal membranes. Proc Natl Acad Sci U S A. 1991;88:7238–7241. doi: 10.1073/pnas.88.16.7238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferre S, Fredholm BB, Morelli M, Popoli P, Fuxe K. Adenosine-dopamine receptor–receptor interactions as an integrative mechanism in the basal ganglia. Trends Neurosci. 1997;20:482–487. doi: 10.1016/s0166-2236(97)01096-5. [DOI] [PubMed] [Google Scholar]
- Fiorentini C, Gardoni F, Spano P, Di Luca M, Missale C. Regulation of dopamine D1 receptor trafficking and desensitization by oligomerization with glutamate N-methyl-D-aspartate receptors. J Biol Chem. 2003;278:20196–20202. doi: 10.1074/jbc.M213140200. [DOI] [PubMed] [Google Scholar]
- Fiorentini C, Busi C, Gorruso E, Gotti C, Spano P, Missale C. Reciprocal regulation of dopamine D1 and D3 receptor function and trafficking by heterodimerization. Mol Pharmacol. 2008;74:59–69. doi: 10.1124/mol.107.043885. [DOI] [PubMed] [Google Scholar]
- Fiorentini C, Mattanza C, Collo G, Savoia P, Spano P, Missale C. The tyrosine phosphatase Shp-2 interacts with the dopamine D(1) receptor and triggers D(1) -mediated Erk signaling in striatal neurons. J Neurochem. 2011;117:253–263. doi: 10.1111/j.1471-4159.2011.07196.x. [DOI] [PubMed] [Google Scholar]
- Fiorentini C, Savoia P, Savoldi D, Barbon A, Missale C. Persistent activation of the D1R/Shp-2/Erk1/2 pathway in L-DOPA-induced dyskinesia in the 6-hydroxy-dopamine rat model of Parkinson's disease. Neurobiol Dis. 2013;54:339–348. doi: 10.1016/j.nbd.2013.01.005. [DOI] [PubMed] [Google Scholar]
- Flores-Barrera E, Thomases DR, Heng LJ, Cass DK, Caballero A, Tseng KY. Late adolescent expression of GluN2B transmission in the prefrontal cortex is input-specific and requires postsynaptic protein kinase A and D1 dopamine receptor signaling. Biol Psychiatry. 2014;75:508–516. doi: 10.1016/j.biopsych.2013.07.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frame S, Cohen P. GSK3 takes centre stage more than 20 years after its discovery. Biochem J. 2001;359:1–16. doi: 10.1042/0264-6021:3590001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Free RB, Chun LS, Moritz AE, Miller BN, Doyle TB, Conroy JL, et al. Discovery and characterization of a G protein-biased agonist that inhibits β-arrestin recruitment to the D2 dopamine receptor. Mol Pharmacol. 2014;86:96–105. doi: 10.1124/mol.113.090563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Freeze BS, Kravitz AV, Hammack N, Berke JD, Kreitzer AC. Control of basal ganglia output by direct and indirect pathway projection neurons. J Neurosci. 2013;33:18531–18539. doi: 10.1523/JNEUROSCI.1278-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Friedman E, Jin LQ, Cai GP, Hollon TR, Drago J, Sibley DR, et al. D1-like dopaminergic activation of phosphoinositide hydrolysis is independent of D1A dopamine receptors: evidence from D1A knockout mice. Mol Pharmacol. 1997;51:6–11. doi: 10.1124/mol.51.1.6. [DOI] [PubMed] [Google Scholar]
- Fuxe K, Marcellino D, Genedani S, Agnati L. Adenosine A(2A) receptors, dopamine D(2) receptors and their interactions in Parkinson's disease. Mov Disord. 2007;22:1990–2017. doi: 10.1002/mds.21440. [DOI] [PubMed] [Google Scholar]
- Fuxe K, Marcellino D, Rivera A, Diaz-Cabiale Z, Filip M, Gago B, et al. Receptor–receptor interactions within receptor mosaics. Impact on neuropsychopharmacology. Brain Res Rev. 2008;58:415–452. doi: 10.1016/j.brainresrev.2007.11.007. [DOI] [PubMed] [Google Scholar]
- Fuxe K, Marcellino D, Leo G, Agnati LF. Molecular integration via allosteric interactions in receptor heteromers. A working hypothesis. Curr Opin Pharmacol. 2010;10:14–22. doi: 10.1016/j.coph.2009.10.010. [DOI] [PubMed] [Google Scholar]
- Gainetdinov RR, Sotnikova TD, Grekhova TV, Rayevsky KS. In vivo evidence for preferential role of dopamine D3 receptor in the presynaptic regulation of dopamine release but not synthesis. Eur J Pharmacol. 1996;308:261–269. doi: 10.1016/0014-2999(96)00300-7. [DOI] [PubMed] [Google Scholar]
- Gainetdinov RR, Premont RT, Bohn LM, Lefkowitz RJ, Caron MG. Desensitization of G protein-coupled receptors and neuronal functions. Annu Rev Neurosci. 2004;27:107–144. doi: 10.1146/annurev.neuro.27.070203.144206. [DOI] [PubMed] [Google Scholar]
- Gangarossa G, Valjent E. Regulation of the ERK pathway in the dentate gyrus by in vivo dopamine D1 receptor stimulation requires glutamatergic transmission. Neuropharmacology. 2012;63:1107–1117. doi: 10.1016/j.neuropharm.2012.06.062. [DOI] [PubMed] [Google Scholar]
- Gatto F, Hofland LJ. The role of somatostatin and dopamine D2 receptors in endocrine tumors. Endocr Relat Cancer. 2011;18:R233–R251. doi: 10.1530/ERC-10-0334. [DOI] [PubMed] [Google Scholar]
- George M, Amrutheshwar R, Rajkumar RP, Kattimani S, Dkhar SA. Newer antipsychotics and upcoming molecules for schizophrenia. Eur J Clin Pharmacol. 2013;69:1497–1509. doi: 10.1007/s00228-013-1498-4. [DOI] [PubMed] [Google Scholar]
- Gines S, Hillion J, Torvinen M, Le Crom S, Casado V, Canela EI, et al. Dopamine D1 and adenosine A1 receptors form functionally interacting heteromeric complexes. Proc Natl Acad Sci U S A. 2000;97:8606–8611. doi: 10.1073/pnas.150241097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Girault JA. Signaling in striatal neurons: the phosphoproteins of reward, addiction, and dyskinesia. Prog Mol Biol Transl Sci. 2012;106:33–62. doi: 10.1016/B978-0-12-396456-4.00006-7. [DOI] [PubMed] [Google Scholar]
- Gonzalez S, Moreno-Delgado D, Moreno E, Perez-Capote K, Franco R, Mallol J, et al. Circadian-related heteromerization of adrenergic and dopamine D(4) receptors modulates melatonin synthesis and release in the pineal gland. PLoS Biol. 2012a;10:e1001347. doi: 10.1371/journal.pbio.1001347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gonzalez S, Rangel-Barajas C, Peper M, Lorenzo R, Moreno E, Ciruela F, et al. Dopamine D4 receptor, but not the ADHD-associated D4.7 variant, forms functional heteromers with the dopamine D2S receptor in the brain. Mol Psychiatry. 2012b;17:650–662. doi: 10.1038/mp.2011.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gould TD, Einat H, Bhat R, Manji HK. AR-A014418, a selective GSK-3 inhibitor, produces antidepressant-like effects in the forced swim test. Int J Neuropsychopharmacol. 2004;7:387–390. doi: 10.1017/S1461145704004535. [DOI] [PubMed] [Google Scholar]
- Grady DL, Thanos PK, Corrada MM, Barnett JC, Jr, Ciobanu V, Shustarovich D, et al. DRD4 genotype predicts longevity in mouse and human. J Neurosci. 2013;33:286–291. doi: 10.1523/JNEUROSCI.3515-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gross G, Wicke K, Drescher KU. Dopamine D3 receptor antagonism– still a therapeutic option for the treatment of schizophrenia. Naunyn Schmiedebergs Arch Pharmacol. 2013;386:155–166. doi: 10.1007/s00210-012-0806-3. [DOI] [PubMed] [Google Scholar]
- Grunberger G. Novel therapies for the management of type 2 diabetes mellitus: part 1. pramlintide and bromocriptine-QR. J Diabetes. 2013;5:110–117. doi: 10.1111/1753-0407.12034. [DOI] [PubMed] [Google Scholar]
- Guo W, Urizar E, Kralikova M, Mobarec JC, Shi L, Filizola M, et al. Dopamine D2 receptors form higher order oligomers at physiological expression levels. EMBO J. 2008;27:2293–2304. doi: 10.1038/emboj.2008.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gutierrez A, Pan L, Groen RW, Baleydier F, Kentsis A, Marineau J, et al. Phenothiazines induce PP2A-mediated apoptosis in T cell acute lymphoblastic leukemia. J Clin Invest. 2014;124:644–655. doi: 10.1172/JCI65093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Halpain S, Girault JA, Greengard P. Activation of NMDA receptors induces dephosphorylation of DARPP-32 in rat striatal slices. Nature. 1990;343:369–372. doi: 10.1038/343369a0. [DOI] [PubMed] [Google Scholar]
- Han Y, Moreira IS, Urizar E, Weinstein H, Javitch JA. Allosteric communication between protomers of dopamine class A GPCR dimers modulates activation. Nat Chem Biol. 2009;5:688–695. doi: 10.1038/nchembio.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hanami K, Nakano K, Saito K, Okada Y, Yamaoka K, Kubo S, et al. Dopamine D2-like receptor signaling suppresses human osteoclastogenesis. Bone. 2013;56:1–8. doi: 10.1016/j.bone.2013.04.019. [DOI] [PubMed] [Google Scholar]
- Hasbi A, Fan T, Alijaniaram M, Nguyen T, Perreault ML, O'Dowd BF, et al. Calcium signaling cascade links dopamine D1–D2 receptor heteromer to striatal BDNF production and neuronal growth. Proc Natl Acad Sci U S A. 2009;106:21377–21382. doi: 10.1073/pnas.0903676106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hazelwood LA, Free RB, Cabrera DM, Skinbjerg M, Sibley DR. Reciprocal modulation of function between the D1 and D2 dopamine receptors and the Na+,K+-ATPase. J Biol Chem. 2008;283:36441–36453. doi: 10.1074/jbc.M805520200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hernandez-Lopez S, Tkatch T, Perez-Garci E, Galarraga E, Bargas J, Hamm H, et al. D2 dopamine receptors in striatal medium spiny neurons reduce L-type Ca2+ currents and excitability via a novel PLC[beta]1-IP3-calcineurin-signaling cascade. J Neurosci. 2000;20:8987–8995. doi: 10.1523/JNEUROSCI.20-24-08987.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herrmann R, Heflin SJ, Hammond T, Lee B, Wang J, Gainetdinov RR, et al. Rod vision is controlled by dopamine-dependent sensitization of rod bipolar cells by GABA. Neuron. 2011;72:101–110. doi: 10.1016/j.neuron.2011.07.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hiller C, Kling RC, Heinemann FW, Meyer K, Hubner H, Gmeiner P. Functionally selective dopamine D2/D3 receptor agonists comprising an enyne moiety. J Med Chem. 2013;56:5130–5141. doi: 10.1021/jm400520c. [DOI] [PubMed] [Google Scholar]
- Hillion J, Canals M, Torvinen M, Casado V, Scott R, Terasmaa A, et al. Coaggregation, cointernalization, and codesensitization of adenosine A2A receptors and dopamine D2 receptors. J Biol Chem. 2002;277:18091–18097. doi: 10.1074/jbc.M107731200. [DOI] [PubMed] [Google Scholar]
- Hobson BD, O'Neill CE, Levis SC, Monteggia LM, Neve RL, Self DW, et al. Adenosine A1 and dopamine d1 receptor regulation of AMPA receptor phosphorylation and cocaine-seeking behavior. Neuropsychopharmacology. 2013;38:1974–1983. doi: 10.1038/npp.2013.96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holmes A, Lachowicz JE, Sibley DR. Phenotypic analysis of dopamine receptor knockout mice; recent insights into the functional specificity of dopamine receptor subtypes. Neuropharmacology. 2004;47:1117–1134. doi: 10.1016/j.neuropharm.2004.07.034. [DOI] [PubMed] [Google Scholar]
- Huang YY, Kandel ER. D1/D5 receptor agonists induce a protein synthesis-dependent late potentiation in the CA1 region of the hippocampus. Proc Natl Acad Sci U S A. 1995;92:2446–2450. doi: 10.1073/pnas.92.7.2446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Humbert S, Bryson EA, Cordelières FP, Connors NC, Datta SR, Finkbeiner S, et al. The IGF-1/Akt pathway is neuroprotective in Huntington's disease and involves Huntingtin phosphorylation by Akt. Dev Cell. 2002;2:831–837. doi: 10.1016/s1534-5807(02)00188-0. [DOI] [PubMed] [Google Scholar]
- Hutchinson JA, Shanware NP, Chang H, Tibbetts RS. Regulation of ribosomal protein S6 phosphorylation by casein kinase 1 and protein phosphatase 1. J Biol Chem. 2011;286:8688–8696. doi: 10.1074/jbc.M110.141754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Inglese J, Luttrell LM, Iniguez-Lluhi JA, Touhara K, Koch WJ, Lefkowitz RJ. Functionally active targeting domain of the beta-adrenergic receptor kinase: an inhibitor of G beta gamma-mediated stimulation of type II adenylyl cyclase. Proc Natl Acad Sci U S A. 1994;91:3637–3641. doi: 10.1073/pnas.91.9.3637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iwakura Y, Nawa H, Sora I, Chao MV. Dopamine D1 receptor-induced signaling through TrkB receptors in striatal neurons. J Biol Chem. 2008;283:15799–15806. doi: 10.1074/jbc.M801553200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iwakura Y, Zheng Y, Sibilia M, Abe Y, Piao YS, Yokomaku D, et al. Qualitative and quantitative re-evaluation of epidermal growth factor-ErbB1 action on developing midbrain dopaminergic neurons in vivo and in vitro: target-derived neurotrophic signaling (part 1) J Neurochem. 2011;118:45–56. doi: 10.1111/j.1471-4159.2011.07287.x. [DOI] [PubMed] [Google Scholar]
- Jacinto E, Facchinetti V, Liu D, Soto N, Wei S, Jung SY, et al. SIN1/MIP1 maintains rictor–mTOR complex integrity and regulates Akt phosphorylation and substrate specificity. Cell. 2006;127:125–137. doi: 10.1016/j.cell.2006.08.033. [DOI] [PubMed] [Google Scholar]
- Jackson CR, Ruan GX, Aseem F, Abey J, Gamble K, Stanwood G, et al. Retinal dopamine mediates multiple dimensions of light-adapted vision. J Neurosci. 2012;32:9359–9368. doi: 10.1523/JNEUROSCI.0711-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jenkins MA, Traynelis SF. PKC phosphorylates GluA1-Ser831 to enhance AMPA receptor conductance. Channels (Austin) 2012;6:60–64. doi: 10.4161/chan.18648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jenkins MA, Wells G, Bachman J, Snyder JP, Jenkins A, Huganir RL, et al. Regulation of GluA1 AMPA receptor function by protein kinase C at serine-818 and threonine-840. Mol Pharmacol. 2014;85:618–629. doi: 10.1124/mol.113.091488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jia JM, Zhao J, Hu Z, Lindberg D, Li Z. Age-dependent regulation of synaptic connections by dopamine D2 receptors. Nat Neurosci. 2013;16:1627–1636. doi: 10.1038/nn.3542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joseph JD, Wang YM, Miles PR, Budygin EA, Picetti R, Gainetdinov RR, et al. Dopamine autoreceptor regulation of release and uptake in mouse brain slices in the absence of D(3) receptors. Neuroscience. 2002;112:39–49. doi: 10.1016/s0306-4522(02)00067-2. [DOI] [PubMed] [Google Scholar]
- Kaidanovich-Belin O, Woodgett JR. GSK-3: functional insights from cell biology and animal models. Front Mol Neurosci. 2011;4:40. doi: 10.3389/fnmol.2011.00040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalinichev M, Dawson LA. Evidence for antimanic efficacy of glycogen synthase kinase-3 (GSK3) inhibitors in a strain-specific model of acute mania. Int J Neuropsychopharmacol. 2011;14:1051–1067. doi: 10.1017/S1461145710001495. [DOI] [PubMed] [Google Scholar]
- Kebabian JW. Multiple classes of dopamine receptors in mammalian central nervous system: the involvement of dopamine-sensitive adenylyl cyclase. Life Sci. 1978;23:479–483. doi: 10.1016/0024-3205(78)90157-1. [DOI] [PubMed] [Google Scholar]
- Kellendonk C, Simpson EH, Polan HJ, Malleret G, Vronskaya S, Winiger V, et al. Transient and selective overexpression of dopamine D2 receptors in the striatum causes persistent abnormalities in prefrontal cortex functioning. Neuron. 2006;49:603–615. doi: 10.1016/j.neuron.2006.01.023. [DOI] [PubMed] [Google Scholar]
- Kern A, Albarran-Zeckler R, Walsh HE, Smith RG. Apo-ghrelin receptor forms heteromers with DRD2 in hypothalamic neurons and is essential for anorexigenic effects of DRD2 agonism. Neuron. 2012;73:317–332. doi: 10.1016/j.neuron.2011.10.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kisilevsky AE, Zamponi GW. D2 dopamine receptors interact directly with N-type calcium channels and regulate channel surface expression levels. Channels (Austin) 2008;2:269–277. doi: 10.4161/chan.2.4.6402. [DOI] [PubMed] [Google Scholar]
- Kisilevsky AE, Mulligan SJ, Altier C, Iftinca MC, Varela D, Tai C, et al. D1 receptors physically interact with N-type calcium channels to regulate channel distribution and dendritic calcium entry. Neuron. 2008;58:557–570. doi: 10.1016/j.neuron.2008.03.002. [DOI] [PubMed] [Google Scholar]
- Koch WJ, Hawes BE, Inglese J, Luttrell LM, Lefkowitz RJ. Cellular expression of the carboxyl terminus of a G protein-coupled receptor kinase attenuates G beta gamma-mediated signaling. J Biol Chem. 1994;269:6193–6197. [PubMed] [Google Scholar]
- Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature. 1999;402:656–660. doi: 10.1038/45230. [DOI] [PubMed] [Google Scholar]
- Kravitz AV, Freeze BS, Parker PR, Kay K, Thwin MT, Deisseroth K, et al. Regulation of parkinsonian motor behaviours by optogenetic control of basal ganglia circuitry. Nature. 2010;466:622–626. doi: 10.1038/nature09159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kravitz AV, Tye LD, Kreitzer AC. Distinct roles for direct and indirect pathway striatal neurons in reinforcement. Nat Neurosci. 2012;15:816–818. doi: 10.1038/nn.3100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuzhikandathil EV, Yu W, Oxford GS. Human dopamine D3 and D2L receptors couple to inward rectifier potassium channels in mammalian cell lines. Mol Cell Neurosci. 1998;12:390–402. doi: 10.1006/mcne.1998.0722. [DOI] [PubMed] [Google Scholar]
- Lancaster E, Dalmau J. Neuronal autoantigens–pathogenesis, associated disorders and antibody testing. Nat Rev Neurol. 2012;8:380–390. doi: 10.1038/nrneurol.2012.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Land BB, Narayanan NS, Liu RJ, Gianessi CA, Brayton CE, Grimaldi DM, et al. Medial prefrontal D1 dopamine neurons control food intake. Nat Neurosci. 2014;17:248–253. doi: 10.1038/nn.3625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lane JR, Chubukov P, Liu W, Canals M, Cherezov V, Abagyan R, et al. Structure-based ligand discovery targeting orthosteric and allosteric pockets of dopamine receptors. Mol Pharmacol. 2013;84:794–807. doi: 10.1124/mol.113.088054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Latapy C, Rioux V, Guitton MJ, Beaulieu JM. Selective deletion of forebrain glycogen synthase kinase 3beta reveals a central role in serotonin-sensitive anxiety and social behaviour. Philos Trans R Soc Lond B Biol Sci. 2012;367:2460–2474. doi: 10.1098/rstb.2012.0094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lavoie J, Illiano P, Sotnikova TD, Gainetdinov RR, Beaulieu JM, Hebert M. The electroretinogram as a biomarker of central dopamine and serotonin: potential relevance to psychiatric disorders. Biol Psychiatry. 2014a;75:479–486. doi: 10.1016/j.biopsych.2012.11.024. [DOI] [PubMed] [Google Scholar]
- Lavoie J, Hébert M, Beaulieu JM. Glycogen synthase kinase-3 overexpression replicates electroretinogram anomalies of offspring at high genetic risk for schizophrenia and bipolar disorder. Biol Psychiatry. 2014b;76:93–100. doi: 10.1016/j.biopsych.2013.08.035. [DOI] [PubMed] [Google Scholar]
- Lee FJ, Xue S, Pei L, Vukusic B, Chery N, Wang Y, et al. Dual regulation of NMDA receptor functions by direct protein–protein interactions with the dopamine D1 receptor. Cell. 2002;111:219–230. doi: 10.1016/s0092-8674(02)00962-5. [DOI] [PubMed] [Google Scholar]
- Lee HM, Giguere PM, Roth BL. DREADDs: novel tools for drug discovery and development. Drug Discov Today. 2013;19:469–473. doi: 10.1016/j.drudis.2013.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee M, O'Regan S, Moreau JL, Johnson AL, Johnston LH, Goding CR. Regulation of the Pcl7-Pho85 cyclin–CDK complex by Pho81. Mol Microbiol. 2000;38:411–422. doi: 10.1046/j.1365-2958.2000.02140.x. [DOI] [PubMed] [Google Scholar]
- Lee SM, Kant A, Blake D, Murthy V, Boyd K, Wyrick SJ, et al. SKF-83959 is not a highly-biased functionally selective D dopamine receptor ligand with activity at phospholipase C. Neuropharmacology. 2014;86:145–154. doi: 10.1016/j.neuropharm.2014.05.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee SP, So CH, Rashid AJ, Varghese G, Cheng R, Lanca AJ, et al. Dopamine D1 and D2 receptor co-activation generates a novel phospholipase C-mediated calcium signal. J Biol Chem. 2004;279:35671–35678. doi: 10.1074/jbc.M401923200. [DOI] [PubMed] [Google Scholar]
- Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell. 2010;141:1117–1134. doi: 10.1016/j.cell.2010.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leo D, Mus L, Espinoza S, Hoener MC, Sotnikova TD, Gainetdinov RR. Taar1-mediated modulation of presynaptic dopaminergic neurotransmission: role of D2 dopamine autoreceptors. Neuropharmacology. 2014;81:283–291. doi: 10.1016/j.neuropharm.2014.02.007. [DOI] [PubMed] [Google Scholar]
- Li YC, Gao WJ. GSK-3beta activity and hyperdopamine-dependent behaviors. Neurosci Biobehav Rev. 2011a;35:645–654. doi: 10.1016/j.neubiorev.2010.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li YC, Xi D, Roman J, Huang YQ, Gao WJ. Activation of glycogen synthase kinase-3 beta is required for hyperdopamine and D2 receptor-mediated inhibition of synaptic NMDA receptor function in the rat prefrontal cortex. J Neurosci. 2009;29:15551–15563. doi: 10.1523/JNEUROSCI.3336-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li YC, Kellendonk C, Simpson EH, Kandel ER, Gao WJ. D2 receptor overexpression in the striatum leads to a deficit in inhibitory transmission and dopamine sensitivity in mouse prefrontal cortex. Proc Natl Acad Sci U S A. 2011b;108:12107–12112. doi: 10.1073/pnas.1109718108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li YC, Wang MJ, Gao WJ. Hyperdopaminergic modulation of inhibitory transmission is dependent on GSK-3beta signaling-mediated trafficking of GABAA receptors. J Neurochem. 2012;122:308–320. doi: 10.1111/j.1471-4159.2012.07790.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lindemann L, Meyer CA, Jeanneau K, Bradaia A, Ozmen L, Bluethmann H, et al. Trace amine-associated receptor 1 modulates dopaminergic activity. J Pharmacol Exp Ther. 2008;324:948–956. doi: 10.1124/jpet.107.132647. [DOI] [PubMed] [Google Scholar]
- Lindgren N, Usiello A, Goiny M, Haycock J, Erbs E, Greengard P, et al. Distinct roles of dopamine D2L and D2S receptor isoforms in the regulation of protein phosphorylation at presynaptic and postsynaptic sites. Proc Natl Acad Sci U S A. 2003;100:4305–4309. doi: 10.1073/pnas.0730708100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu XY, Chu XP, Mao LM, Wang M, Lan HX, Li MH, et al. Modulation of D2R–NR2B interactions in response to cocaine. Neuron. 2006;52:897–909. doi: 10.1016/j.neuron.2006.10.011. [DOI] [PubMed] [Google Scholar]
- Lobo MK, Zaman S, Damez-Werno DM, Koo JW, Bagot RC, DiNieri JA, et al. DeltaFosB induction in striatal medium spiny neuron subtypes in response to chronic pharmacological, emotional, and optogenetic stimuli. J Neurosci. 2013;33:18381–18395. doi: 10.1523/JNEUROSCI.1875-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lohse MJ, Benovic JL, Codina J, Caron MG, Lefkowitz RJ. Beta-arrestin: a protein that regulates beta-adrenergic receptor function. Science. 1990;248:1547–1550. doi: 10.1126/science.2163110. [DOI] [PubMed] [Google Scholar]
- Lohse MJ, Nuber S, Hoffmann C. Fluorescence/bioluminescence resonance energy transfer techniques to study G–protein-coupled receptor activation and signaling. Pharmacol Rev. 2012;64:299–336. doi: 10.1124/pr.110.004309. [DOI] [PubMed] [Google Scholar]
- Luttrell LM, Roudabush FL, Choy EW, Miller WE, Field ME, Pierce KL, et al. Activation and targeting of extracellular signal-regulated kinases by beta-arrestin scaffolds. Proc Natl Acad Sci U S A. 2001;98:2449–2454. doi: 10.1073/pnas.041604898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maeda K, Lerdrup L, Sugino H, Akazawa H, Amada N, McQuade RD, et al. Brexpiprazole II: antipsychotic-like profile and pro-cognitive effects of a novel serotonin-dopamineactivity modulator. J Pharmacol Exp Ther. 2014;350:605–614. doi: 10.1124/jpet.114.213819. [DOI] [PubMed] [Google Scholar]
- Maison SF, Liu XP, Eatock RA, Sibley DR, Grandy DK, Liberman MC. Dopaminergic signaling in the cochlea: receptor expression patterns and deletion phenotypes. J Neurosci. 2012;32:344–355. doi: 10.1523/JNEUROSCI.4720-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mannoury la Cour C, Salles MJ, Pasteau V, Millan MJ. Signaling pathways leading to phosphorylation of Akt and GSK-3beta by activation of cloned human and rat cerebral D(2)and D(3) receptors. Mol Pharmacol. 2011;79:91–105. doi: 10.1124/mol.110.065409. [DOI] [PubMed] [Google Scholar]
- Marcellino D, Ferre S, Casado V, Cortes A, Le Foll B, Mazzola C, et al. Identification of dopamine D1-D3 receptor heteromers. Indications for a role of synergistic D1–D3 receptor interactions in the striatum. J Biol Chem. 2008;283:26016–26025. doi: 10.1074/jbc.M710349200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marion S, Urs NM, Peterson SM, Sotnikova TD, Beaulieu JM, Gainetdinov RR, et al. Dopamine D2 receptor relies upon PPM/PP2C protein phosphatases to dephosphorylate huntingtin protein. J Biol Chem. 2014;289:11715–11724. doi: 10.1074/jbc.M113.544312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martelli AM, Chiarini F, Evangelisti C, Grimaldi C, Ognibene A, Manzoli L, et al. The phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin signaling network and the control of normal myelopoiesis. Histol Histopathol. 2010;25:669–680. doi: 10.14670/HH-25.669. [DOI] [PubMed] [Google Scholar]
- Marullo S, Bouvier M. Resonance energy transfer approaches in molecular pharmacology and beyond. Trends Pharmacol Sci. 2007;28:362–365. doi: 10.1016/j.tips.2007.06.007. [DOI] [PubMed] [Google Scholar]
- Masri B, Salahpour A, Didriksen M, Ghisi V, Beaulieu JM, Gainetdinov RR, et al. Antagonism of dopamine D2 receptor/beta-arrestin 2 interaction is a common property of clinically effective antipsychotics. Proc Natl Acad Sci U S A. 2008;105:13656–13661. doi: 10.1073/pnas.0803522105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maudsley S, Pierce KL, Zamah AM, Miller WE, Ahn S, Daaka Y, et al. The beta(2)-adrenergic receptor mediates extracellular signal-regulated kinase activation via assembly of a multi-receptor complex with the epidermal growth factor receptor. J Biol Chem. 2000;275:9572–9580. doi: 10.1074/jbc.275.13.9572. [DOI] [PubMed] [Google Scholar]
- McIntyre RS, Cha DS, Kim RD, Mansur RB. A review of FDA-approved treatment options in bipolar depression. CNS Spectr. 2013;18(Suppl. 1):4–20. doi: 10.1017/S1092852913000746. [DOI] [PubMed] [Google Scholar]
- Medvedev IO, Ramsey AJ, Masoud ST, Bermejo MK, Urs N, Sotnikova TD, et al. D1 dopamine receptor coupling to PLCbeta regulates forward locomotion in mice. J Neurosci. 2013;33:18125–18133. doi: 10.1523/JNEUROSCI.2382-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Milligan G. Applications of bioluminescence- and fluorescence resonance energy transfer to drug discovery at G protein-coupled receptors. Eur J Pharm Sci. 2004;21:397–405. doi: 10.1016/j.ejps.2003.11.010. [DOI] [PubMed] [Google Scholar]
- Missale C, Nash SR, Robinson SW, Jaber M, Caron MG. Dopamine receptors: from structure to function. Physiol Rev. 1998;78:189–225. doi: 10.1152/physrev.1998.78.1.189. [DOI] [PubMed] [Google Scholar]
- Mohammad SS, Sinclair K, Pillai S, Merheb V, Aumann TD, Gill D, et al. Herpes simplex encephalitis relapse with chorea is associated with autoantibodies to N-methyl-D-aspartate receptor or dopamine-2 receptor. Mov Disord. 2014;29:117–122. doi: 10.1002/mds.25623. [DOI] [PubMed] [Google Scholar]
- Moreno-Smith M, Lu C, Shahzad MM, Pena GN, Allen JK, Stone RL, et al. Dopamine blocks stress-mediated ovarian carcinoma growth. Clin Cancer Res. 2011;17:3649–3659. doi: 10.1158/1078-0432.CCR-10-2441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moreno-Smith M, Lee SJ, Lu C, Nagaraja AS, He G, Rupaimoole R, et al. Biologic effects of dopamine on tumor vasculature in ovarian carcinoma. Neoplasia. 2013;15:502–510. doi: 10.1593/neo.121412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Möller D, Kling RC, Skultety M, Leuner K, Hübner H, Gmeiner P. Functionally selective dopamine d2, d3 receptor partial agonists. J Med Chem. 2014;57:4861–4875. doi: 10.1021/jm5004039. [DOI] [PubMed] [Google Scholar]
- Murphy JA, Stein IS, Lau CG, Peixoto RT, Aman TK, Kaneko N, et al. Phosphorylation of Ser1166 on GluN2B by PKA is critical to synaptic NMDA receptor function and Ca2+signaling in spines. J Neurosci. 2014;34:869–879. doi: 10.1523/JNEUROSCI.4538-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nai Q, Li S, Wang SH, Liu J, Lee FJ, Frankland PW, et al. Uncoupling the D1-N-methyl-D-aspartate (NMDA) receptor complex promotes NMDA-dependent long-term potentiation and working memory. Biol Psychiatry. 2010;67:246–254. doi: 10.1016/j.biopsych.2009.08.011. [DOI] [PubMed] [Google Scholar]
- Narayanan NS, Land BB, Solder JE, Deisseroth K, DiLeone RJ. Prefrontal D1 dopamine signaling is required for temporal control. Proc Natl Acad Sci U S A. 2012;109:20726–20731. doi: 10.1073/pnas.1211258109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neve KA, Ford CP, Buck DC, Grandy DK, Neve RL, Phillips TJ. Normalizing dopamine D2 receptor-mediated responses in D2 null mutant mice by virus-mediated receptor restoration: comparing D2 and D2. Neuroscience. 2013;248C:479–487. doi: 10.1016/j.neuroscience.2013.06.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Newman AH, Beuming T, Banala AK, Donthamsetti P, Pongetti K, LaBounty A, et al. Molecular determinants of selectivity and efficacy at the dopamine D3 receptor. J Med Chem. 2012;55:6689–6699. doi: 10.1021/jm300482h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nickols HH, Conn PJ. Development of allosteric modulators of GPCRs for treatment of CNS disorders. Neurobiol Dis. 2014;61:55–71. doi: 10.1016/j.nbd.2013.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niznik HB, Van Tol HH. Dopamine receptor genes: new tools for molecular psychiatry. J Psychiatry Neurosci. 1992;17:158–180. [PMC free article] [PubMed] [Google Scholar]
- O'Brien WT, Huang J, Buccafusca R, Garskof J, Valvezan AJ, Berry GT, et al. Glycogen synthase kinase-3 is essential for beta-arrestin-2 complex formation and lithium-sensitive behaviors in mice. J Clin Invest. 2011;121:3756–3762. doi: 10.1172/JCI45194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Owens WA, Williams JM, Saunders C, Avison MJ, Galli A, Daws LC. Rescue of dopamine transporter function in hypoinsulinemic rats by a D2 receptor-ERK-dependent mechanism. J Neurosci. 2012;32:2637–2647. doi: 10.1523/JNEUROSCI.3759-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan JQ, Lewis MC, Ketterman JK, Clore EL, Riley M, Richards KR, et al. AKT kinase activity is required for lithium to modulate mood-related behaviors in mice. Neuropsychopharmacology. 2011;36:1397–1411. doi: 10.1038/npp.2011.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SP, Buneman OP, et al. The IUPHAR/BPS Guide to PHARMACOLOGY: an expert-driven knowledgebase of drug targets and their ligands. Nucleic Acids Res. 2014;42(Database Issue):D1098–D1106. doi: 10.1093/nar/gkt1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pei L, Li S, Wang M, Diwan M, Anisman H, Fletcher PJ, et al. Uncoupling the dopamine D1–D2 receptor complex exerts antidepressant-like effects. Nat Med. 2010;16:1393–1395. doi: 10.1038/nm.2263. [DOI] [PubMed] [Google Scholar]
- Perez Millan MI, Luque GM, Ramirez MC, Noain D, Ornstein AM, Rubinstein M, et al. Selective disruption of dopamine d2 receptors in pituitary lactotropes increases body weight and adiposity in female mice. Endocrinology. 2014;155:829–839. doi: 10.1210/en.2013-1707. [DOI] [PubMed] [Google Scholar]
- Perreault ML, Hasbi A, Alijaniaram M, Fan T, Varghese G, Fletcher PJ, et al. The dopamine D1–D2 receptor heteromer localizes in dynorphin/enkephalin neurons: increased high affinity state following amphetamine and in schizophrenia. J Biol Chem. 2010;285:36625–36634. doi: 10.1074/jbc.M110.159954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perreault ML, Jones-Tabah J, O'Dowd BF, George SR. A physiological role for the dopamine D5 receptor as a regulator of BDNF and Akt signalling in rodent prefrontal cortex. Int J Neuropsychopharmacol. 2013;16:477–483. doi: 10.1017/S1461145712000685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perreault ML, Hasbi A, O'Dowd BF, George SR. Heteromeric dopamine receptor signaling complexes: emerging neurobiology and disease relevance. Neuropsychopharmacology. 2014;39:156–168. doi: 10.1038/npp.2013.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pfleger KD, Eidne KA. Illuminating insights into protein–protein interactions using bioluminescence resonance energy transfer (BRET) Nat Methods. 2006;3:165–174. doi: 10.1038/nmeth841. [DOI] [PubMed] [Google Scholar]
- Polter A, Beurel E, Yang S, Garner R, Song L, Miller CA, et al. Deficiency in the inhibitory serine-phosphorylation of glycogen synthase kinase-3 increases sensitivity to mood disturbances. Neuropsychopharmacology. 2010;35:1761–1774. doi: 10.1038/npp.2010.43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prado C, Contreras F, Gonzalez H, Diaz P, Elgueta D, Barrientos M, et al. Stimulation of dopamine receptor D5 expressed on dendritic cells potentiates Th17-mediated immunity. J Immunol. 2012;188:3062–3070. doi: 10.4049/jimmunol.1103096. [DOI] [PubMed] [Google Scholar]
- Prickaerts J, Moechars D, Cryns K, Lenaerts I, van Craenendonck H, Goris I, et al. Transgenic mice overexpressing glycogen synthase kinase 3beta: a putative model of hyperactivity and mania. J Neurosci. 2006;26:9022–9029. doi: 10.1523/JNEUROSCI.5216-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Radl D, De Mei C, Chen E, Lee H, Borrelli E. Each individual isoform of the dopamine D2 receptor protects from lactotroph hyperplasia. Mol Endocrinol. 2013;27:953–965. doi: 10.1210/me.2013-1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rajagopal R, Chen ZY, Lee FS, Chao MV. Transactivation of Trk neurotrophin receptors by G–protein-coupled receptor ligands occurs on intracellular membranes. J Neurosci. 2004;24:6650–6658. doi: 10.1523/JNEUROSCI.0010-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rangone H, Poizat G, Troncoso J, Ross CA, MacDonald ME, Saudou F, et al. The serum- and glucocorticoid-induced kinase SGK inhibits mutant huntingtin-induced toxicity by phosphorylating serine 421 of huntingtin. Eur J Neurosci. 2004;19:273–279. doi: 10.1111/j.0953-816x.2003.03131.x. [DOI] [PubMed] [Google Scholar]
- Rashid AJ, O'Dowd BF, Verma V, George SR. Neuronal Gq/11-coupled dopamine receptors: an uncharted role for dopamine. Trends Pharmacol Sci. 2007a;28:551–555. doi: 10.1016/j.tips.2007.10.001. [DOI] [PubMed] [Google Scholar]
- Rashid AJ, So CH, Kong MM, Furtak T, El-Ghundi M, Cheng R, et al. D1–D2 dopamine receptor heterooligomers with unique pharmacology are coupled to rapid activation of Gq/11 in the striatum. Proc Natl Acad Sci U S A. 2007b;104:654–659. doi: 10.1073/pnas.0604049104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Revel FG, Moreau JL, Gainetdinov RR, Bradaia A, Sotnikova TD, Mory R, et al. TAAR1 activation modulates monoaminergic neurotransmission, preventing hyperdopaminergic and hypoglutamatergic activity. Proc Natl Acad Sci U S A. 2011;108:8485–8490. doi: 10.1073/pnas.1103029108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Revel FG, Moreau JL, Gainetdinov RR, Ferragud A, Velazquez-Sanchez C, Sotnikova TD, et al. Trace amine-associated receptor 1 partial agonism reveals novel paradigm for neuropsychiatric therapeutics. Biol Psychiatry. 2012;72:934–942. doi: 10.1016/j.biopsych.2012.05.014. [DOI] [PubMed] [Google Scholar]
- Revel FG, Moreau JL, Pouzet B, Mory R, Bradaia A, Buchy D, et al. A new perspective for schizophrenia: TAAR1 agonists reveal antipsychotic- and antidepressant-like activity, improve cognition and control body weight. Mol Psychiatry. 2013;18:543–556. doi: 10.1038/mp.2012.57. [DOI] [PubMed] [Google Scholar]
- Reynolds GP, Kirk SL. Metabolic side effects of antipsychotic drug treatment– pharmacological mechanisms. Pharmacol Ther. 2010;125:169–179. doi: 10.1016/j.pharmthera.2009.10.010. [DOI] [PubMed] [Google Scholar]
- Rimondini R, Ferre S, Ogren SO, Fuxe K. Adenosine A2A agonists: a potential new type of atypical antipsychotic. Neuropsychopharmacology. 1997;17:82–91. doi: 10.1016/S0893-133X(97)00033-X. [DOI] [PubMed] [Google Scholar]
- Rossig L, Badorff C, Holzmann Y, Zeiher AM, Dimmeler S. Glycogen synthase kinase-3 couples AKT-dependent signaling to the regulation of p21Cip1 degradation. J Biol Chem. 2002;277:9684–9689. doi: 10.1074/jbc.M106157200. [DOI] [PubMed] [Google Scholar]
- Roux PP, Shahbazian D, Vu H, Holz MK, Cohen MS, Taunton J, et al. RAS/ERK signaling promotes site-specific ribosomal protein S6 phosphorylation via RSK and stimulates cap-dependent translation. J Biol Chem. 2007;282:14056–14064. doi: 10.1074/jbc.M700906200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sachlos E, Risueno RM, Laronde S, Shapovalova Z, Lee JH, Russell J, et al. Identification of drugs including a dopamine receptor antagonist that selectively target cancer stem cells. Cell. 2012;149:1284–1297. doi: 10.1016/j.cell.2012.03.049. [DOI] [PubMed] [Google Scholar]
- Sahin B, Hawasli AH, Greene RW, Molkentin JD, Bibb JA. Negative regulation of cyclin-dependent kinase 5 targets by protein kinase C. Eur J Pharmacol. 2008;581:270–275. doi: 10.1016/j.ejphar.2007.11.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sahu A, Tyeryar KR, Vongtau HO, Sibley DR, Undieh AS. D5 dopamine receptors are required for dopaminergic activation of phospholipase C. Mol Pharmacol. 2009;75:447–453. doi: 10.1124/mol.108.053017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salahpour A, Masri B. Experimental challenge to a ‘rigorous’ BRET analysis of GPCR oligomerization. Nat Methods. 2007;4:599–600. doi: 10.1038/nmeth0807-599. author reply 601. [DOI] [PubMed] [Google Scholar]
- Salahpour A, Espinoza S, Masri B, Lam V, Barak LS, Gainetdinov RR. BRET biosensors to study GPCR biology, pharmacology, and signal transduction. Front Endocrinol (Lausanne) 2012;3:105. doi: 10.3389/fendo.2012.00105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santini E, Heiman M, Greengard P, Valjent E, Fisone G. Inhibition of mTOR signaling in Parkinson's disease prevents L–DOPA-induced dyskinesia. Sci Signal. 2009;2:ra36. doi: 10.1126/scisignal.2000308. [DOI] [PubMed] [Google Scholar]
- Santini E, Feyder M, Gangarossa G, Bateup HS, Greengard P, Fisone G. Dopamine- and cAMP-regulated phosphoprotein of 32-kDa (DARPP-32)-dependent activation of extracellular signal-regulated kinase (ERK) and mammalian target of rapamycin complex 1 (mTORC1) signaling in experimental parkinsonism. J Biol Chem. 2012;287:27806–27812. doi: 10.1074/jbc.M112.388413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scheid MP, Woodgett JR. PKB/AKT: functional insights from genetic models. Nat Rev Mol Cell Biol. 2001;2:760–768. doi: 10.1038/35096067. [DOI] [PubMed] [Google Scholar]
- Seeman P. Targeting the dopamine D2 receptor in schizophrenia. Expert Opin Ther Targets. 2006;10:515–531. doi: 10.1517/14728222.10.4.515. [DOI] [PubMed] [Google Scholar]
- Shao W, Zhang SZ, Tang M, Zhang XH, Zhou Z, Yin YQ, et al. Suppression of neuroinflammation by astrocytic dopamine D2 receptors via alphaB-crystallin. Nature. 2013;494:90–94. doi: 10.1038/nature11748. [DOI] [PubMed] [Google Scholar]
- Shonberg J, Herenbrink CK, Lopez L, Christopoulos A, Scammells PJ, Capuano B, et al. A structure–activity analysis of biased agonism at the dopamine D2 receptor. J Med Chem. 2013;56:9199–9221. doi: 10.1021/jm401318w. [DOI] [PubMed] [Google Scholar]
- Sibley DR. New insights into dopaminergic receptor function using antisense and genetically altered animals. Annu Rev Pharmacol Toxicol. 1999;39:313–341. doi: 10.1146/annurev.pharmtox.39.1.313. [DOI] [PubMed] [Google Scholar]
- Sibley DR, Monsma FJ., Jr Molecular biology of dopamine receptors. Trends Pharmacol Sci. 1992;13:61–69. doi: 10.1016/0165-6147(92)90025-2. [DOI] [PubMed] [Google Scholar]
- Simpson EH, Kellendonk C, Kandel E. A possible role for the striatum in the pathogenesis of the cognitive symptoms of schizophrenia. Neuron. 2010;65:585–596. doi: 10.1016/j.neuron.2010.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simpson EH, Waltz JA, Kellendonk C, Balsam PD. Schizophrenia in translation: dissecting motivation in schizophrenia and rodents. Schizophr Bull. 2012;38:1111–1117. doi: 10.1093/schbul/sbs114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simpson EH, Winiger V, Biezonski DK, Haq I, Kandel ER, Kellendonk C. Selective overexpression of dopamine D3 receptors in the striatum disrupts motivation but not cognition. Biol Psychiatry. 2014;10:823–831. doi: 10.1016/j.biopsych.2013.11.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- So CH, Verma V, Alijaniaram M, Cheng R, Rashid AJ, O'Dowd BF, et al. Calcium signaling by dopamine D5 receptor and D5-D2 receptor hetero-oligomers occurs by a mechanism distinct from that for dopamine D1–D2 receptor hetero-oligomers. Mol Pharmacol. 2009;75:843–854. doi: 10.1124/mol.108.051805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sokoloff P, Andrieux M, Besancon R, Pilon C, Martres MP, Giros B, et al. Pharmacology of human dopamine D3 receptor expressed in a mammalian cell line: comparison with D2 receptor. Eur J Pharmacol. 1992;225:331–337. doi: 10.1016/0922-4106(92)90107-7. [DOI] [PubMed] [Google Scholar]
- Sokoloff P, Leriche L, Diaz J, Louvel J, Pumain R. Direct and indirect interactions of the dopamine D(3) receptor with glutamate pathways: implications for the treatment of schizophrenia. Naunyn Schmiedebergs Arch Pharmacol. 2013;386:107–124. doi: 10.1007/s00210-012-0797-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song RS, Massenburg B, Wenderski W, Jayaraman V, Thompson L, Neves SR. ERK regulation of phosphodiesterase 4 enhances dopamine-stimulated AMPA receptor membrane insertion. Proc Natl Acad Sci U S A. 2013;110:15437–15442. doi: 10.1073/pnas.1311783110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sotnikova TD, Beaulieu JM, Barak LS, Wetsel WC, Caron MG, Gainetdinov RR. Dopamine-independent locomotor actions of amphetamines in a novel acute mouse model of Parkinson disease. PLoS Biol. 2005;3:e271. doi: 10.1371/journal.pbio.0030271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sotnikova TD, Caron MG, Gainetdinov RR. Trace amine-associated receptors as emerging therapeutic targets. Mol Pharmacol. 2009;76:229–235. doi: 10.1124/mol.109.055970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Souza BR, Romano-Silva MA, Tropepe V. Dopamine D2 receptor activity modulates Akt signaling and alters GABAergic neuron development and motor behavior in zebrafish larvae. J Neurosci. 2011;31:5512–5525. doi: 10.1523/JNEUROSCI.5548-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spano PF, Govoni S, Trabucchi M. Studies on the pharmacological properties of dopamine receptors in various areas of the central nervous system. Adv Biochem Psychopharmacol. 1978;19:155–165. [PubMed] [Google Scholar]
- Stambolic V, Woodgett JR. Mitogen inactivation of glycogen synthase kinase-3 beta in intact cells via serine 9 phosphorylation. Biochem J. 1994;303(Pt 3):701–704. doi: 10.1042/bj3030701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Subramaniam S, Napolitano F, Mealer RG, Kim S, Errico F, Barrow R, et al. Rhes, a striatal-enriched small G protein, mediates mTOR signaling and L–DOPA-induced dyskinesia. Nat Neurosci. 2012;15:191–193. doi: 10.1038/nn.2994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Svenningsson P, Nishi A, Fisone G, Girault JA, Nairn AC, Greengard P. DARPP-32: an integrator of neurotransmission. Annu Rev Pharmacol Toxicol. 2004;44:269–296. doi: 10.1146/annurev.pharmtox.44.101802.121415. [DOI] [PubMed] [Google Scholar]
- Swift JL, Godin AG, Dore K, Freland L, Bouchard N, Nimmo C, et al. Quantification of receptor tyrosine kinase transactivation through direct dimerization and surface density measurements in single cells. Proc Natl Acad Sci U S A. 2011;108:7016–7021. doi: 10.1073/pnas.1018280108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tan ML, Basu D, Kwiecien JM, Johnson RL, Mishra RK. Preclinical pharmacokinetic and toxicological evaluation of MIF-1 peptidomimetic, PAOPA: examining the pharmacology of a selective dopamine D2 receptor allosteric modulator for the treatment of schizophrenia. Peptides. 2013;42:89–96. doi: 10.1016/j.peptides.2013.02.004. [DOI] [PubMed] [Google Scholar]
- Toda S, Alguacil LF, Kalivas PW. Repeated cocaine administration changes the function and subcellular distribution of adenosine A1 receptor in the rat nucleus accumbens. J Neurochem. 2003;87:1478–1484. doi: 10.1046/j.1471-4159.2003.02121.x. [DOI] [PubMed] [Google Scholar]
- Trincavelli ML, Daniele S, Orlandini E, Navarro G, Casado V, Giacomelli C, et al. A new D(2) dopamine receptor agonist allosterically modulates A(2A) adenosine receptor signalling by interacting with the A(2A)/D(2) receptor heteromer. Cell Signal. 2012;24:951–960. doi: 10.1016/j.cellsig.2011.12.018. [DOI] [PubMed] [Google Scholar]
- Urs NM, Snyder JC, Jacobsen JP, Peterson SM, Caron MG. Deletion of GSK3beta in D2R-expressing neurons reveals distinct roles for beta-arrestin signaling in antipsychotic and lithium action. Proc Natl Acad Sci U S A. 2012;109:20732–20737. doi: 10.1073/pnas.1215489109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Valjent E, Caboche J, Vanhoutte P. Mitogen-activated protein kinase/extracellular signal-regulated kinase induced gene regulation in brain: a molecular substrate for learning and memory? Mol Neurobiol. 2001;23:83–99. doi: 10.1385/MN:23:2-3:083. [DOI] [PubMed] [Google Scholar]
- Valjent E, Bertran-Gonzalez J, Bowling H, Lopez S, Santini E, Matamales M, et al. Haloperidol regulates the state of phosphorylation of ribosomal protein S6 via activation of PKA and phosphorylation of DARPP-32. Neuropsychopharmacology. 2011;36:2561–2570. doi: 10.1038/npp.2011.144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vallone D, Picetti R, Borrelli E. Structure and function of dopamine receptors. Neurosci Biobehav Rev. 2000;24:125–132. doi: 10.1016/s0149-7634(99)00063-9. [DOI] [PubMed] [Google Scholar]
- Van Craenenbroeck K, Borroto-Escuela DO, Romero-Fernandez W, Skieterska K, Rondou P, Lintermans B, et al. Dopamine D4 receptor oligomerization–contribution to receptor biogenesis. FEBS J. 2011;278:1333–1344. doi: 10.1111/j.1742-4658.2011.08052.x. [DOI] [PubMed] [Google Scholar]
- Veselinovic T, Paulzen M, Grunder G. Cariprazine, a new, orally active dopamine D2/3 receptor partial agonist for the treatment of schizophrenia, bipolar mania and depression. Expert Rev Neurother. 2013;13:1141–1159. doi: 10.1586/14737175.2013.853448. [DOI] [PubMed] [Google Scholar]
- Wang ZM, Xiang YT, An FR, Correll CU, Ungvari GS, Wang CY, et al. Frequency of hyperprolactinemia and its associations with demographic and clinical characteristics and antipsychotic medications in psychiatric inpatients in China. Perspect Psychiatr Care. 2014;50:257–263. doi: 10.1111/ppc.12050. [DOI] [PubMed] [Google Scholar]
- Ward RD, Kellendonk C, Simpson EH, Lipatova O, Drew MR, Fairhurst S, et al. Impaired timing precision produced by striatal D2 receptor overexpression is mediated by cognitive and motivational deficits. Behav Neurosci. 2009;123:720–730. doi: 10.1037/a0016503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilkinson MB, Dias C, Magida J, Mazei-Robison M, Lobo M, Kennedy P, et al. A novel role of the WNT-dishevelled-GSK3beta signaling cascade in the mouse nucleus accumbens in a social defeat model of depression. J Neurosci. 2011;31:9084–9092. doi: 10.1523/JNEUROSCI.0039-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woodgett JR. Molecular cloning and expression of glycogen synthase kinase-3/factor A. EMBO J. 1990;9:2431–2438. doi: 10.1002/j.1460-2075.1990.tb07419.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woods AS, Jackson SN. How adenylate cyclase choreographs the pas de deux of the receptors heteromerization dance. Neuroscience. 2013;238:335–344. doi: 10.1016/j.neuroscience.2013.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu J, McCallum SE, Glick SD, Huang Y. Inhibition of the mammalian target of rapamycin pathway by rapamycin blocks cocaine-induced locomotor sensitization. Neuroscience. 2011;172:104–109. doi: 10.1016/j.neuroscience.2010.10.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamada T, Katagiri H, Asano T, Tsuru M, Inukai K, Ono H, et al. Role of PDK1 in insulin-signaling pathway for glucose metabolism in 3T3-L1 adipocytes. Am J Physiol Endocrinol Metab. 2002;282:E1385–E1394. doi: 10.1152/ajpendo.00486.2001. [DOI] [PubMed] [Google Scholar]
- Yan Z, Song WJ, Surmeier J. D2 dopamine receptors reduce N-type Ca2+ currents in rat neostriatal cholinergic interneurons through a membrane-delimited, protein-kinase-C-insensitive pathway. J Neurophysiol. 1997;77:1003–1015. doi: 10.1152/jn.1997.77.2.1003. [DOI] [PubMed] [Google Scholar]
- Yoon S, Baik JH. Dopamine D2 receptor-mediated epidermal growth factor receptor transactivation through a disintegrin and metalloprotease regulates dopaminergic neuron development via extracellular signal-related kinase activation. J Biol Chem. 2013;288:28435–28446. doi: 10.1074/jbc.M113.461202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y, Cuevas S, Asico LD, Escano C, Yang Y, Pascua AM, et al. Deficient dopamine D2 receptor function causes renal inflammation independently of high blood pressure. PLoS ONE. 2012;7:e38745. doi: 10.1371/journal.pone.0038745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang ZW, Burke MW, Calakos N, Beaulieu JM, Vaucher E. Confocal analysis of cholinergic and dopaminergic inputs onto pyramidal cells in the prefrontal cortex of rodents. Front Neuroanat. 2010;4:21. doi: 10.3389/fnana.2010.00021. [DOI] [PMC free article] [PubMed] [Google Scholar]