19,413
Views
595
CrossRef citations to date
0
Altmetric
Original

Dopamine Receptor Signaling

, &
Pages 165-205 | Published online: 20 Sep 2004

Abstract

The D1-like (D1, D5) and D2-like (D2, D3, D4) classes of dopamine receptors each has shared signaling properties that contribute to the definition of the receptor class, although some differences among subtypes within a class have been identified. D1-like receptor signaling is mediated chiefly by the heterotrimeric G proteins Gαs and Gαolf, which cause sequential activation of adenylate cyclase, cylic AMP-dependent protein kinase, and the protein phosphatase-1 inhibitor DARPP-32. The increased phosphorylation that results from the combined effects of activating cyclic AMP-dependent protein kinase and inhibiting protein phosphatase 1 regulates the activity of many receptors, enzymes, ion channels, and transcription factors. D1 or a novel D1-like receptor also signals via phospholipase C-dependent and cyclic AMP-independent mobilization of intracellular calcium. D2-like receptor signaling is mediated by the heterotrimeric G proteins Gαi and Gαo. These pertussis toxin-sensitive G proteins regulate some effectors, such as adenylate cyclase, via their Gα subunits, but regulate many more effectors such as ion channels, phospholipases, protein kinases, and receptor tyrosine kinases as a result of the receptor-induced liberation of Gβγ subunits. In addition to interactions between dopamine receptors and G proteins, other protein:protein interactions such as receptor oligomerization or receptor interactions with scaffolding and signal-switching proteins are critical for regulation of dopamine receptor signaling.

Introduction

Five subtypes of mammalian dopamine receptors are grouped into two classes, with the D1-like receptor class composed of the D1 and D5 receptor subtypes, and the D2-like receptor class composed of the D2, D3, and D4 receptor subtypes (Citation[1]). At the molecular level, most signaling properties are shared among all of the subtypes within a class; similarity of signal transduction pathways is one of the criteria by which subtypes are grouped into classes. In this review of dopamine receptor signaling, we avoid repetition by discussing each class as a whole rather than treating each subtype separately when little is known about subtype-specific properties, making specific reference to a subtype primarily when there are data supporting a unique property of that subtype.

All of the dopamine receptors are G protein-coupled receptors (GPCRs), whose signaling is primarily mediated by interaction with and activation of heterotrimeric GTP-binding proteins (G proteins). Members of this superfamily are also called 7-transmembrane receptors because they traverse the cell membrane seven times, or serpentine receptors because of the manner in which they wind back and forth across the membrane.

D1-Like Receptors and G Proteins

As receptors that stimulate adenylate cyclase, the D1-like receptors were assumed to couple to the adenylate cyclase stimulatory G protein Gαs. Because Gαs is ubiquitously expressed, the ability of D1-like receptors to stimulate adenylate cyclase in virtually any cell line (Citation[2]), together with physical and functional coupling of both D1 and D5 receptors to Gαs (Citation[3], Citation[4]), strongly support the notion that Gαs mediates the D1-like receptor signaling in some tissues. In the neostriatum, however, the brain region with the densest dopamine innervation and the highest expression of the D1 receptor, expression of Gαs is very low, whereas Gαolf is abundantly expressed (Citation[5]). The nucleus accumbens and olfactory tubercle also express abundant Gαolf and little Gαs (Citation[6]). Gαolf is the heterotrimeric G protein involved in olfaction, is very closely related to Gαs (88% amino acid homology), and also stimulates adenylate cyclase (Citation[7]). Gαolf null mutant mice exhibit little dopamine-stimulated adenylate cyclase; they also lack other functional and behavioral responses to D1 receptor stimulation, including cocaine or D1 agonist-induced locomotor activation and induction of c-fos expression in the neostriatum or nucleus accumbens, strongly suggesting that Gαolf mediates D1 receptor signaling to adenylate cyclase in these basal ganglia nuclei (Citation[5], Citation[8]).

Less is known about the βγ subunits that combine with Gαs or Gαolf to mediate D1-like receptor activation of adenylate cyclase. In human embryonic kidney (HEK) 293 cells, depletion of endogenous γ7 subunit reduces D1 receptor stimulation of adenylate cyclase, but not D5 receptor stimulation, indicating that γ7 contributes to this pathway for the former subtype (Citation[9]). Depletion of γ7 also decreases the abundance of the β1 subunit, consistent with the formation of a β1γ7 dimer; the presence of Gαs in HEK293 cells suggests that a G protein heterotrimer that mediates D1 receptor activation of adenylate cyclase is Gαsβ1γ7. Because γ7 is abundantly expressed in neostriatal medium spiny neurons (Citation[10]), particularly in neurons that also express D1 receptor mRNA (Citation[9]), neostriatal D1 receptors may signal via a G protein heterotrimer that includes both Gαolf and γ7. In other brain regions, including dopamine target areas that express D1 and/or D5 receptors such as the cerebral cortex and hippocampus where the expression of Gαolf is much lower than that of Gαs (Citation[6]), it seems likely that Gαs mediates D1 and D5 receptor signaling to adenylate cyclase. Coupling of the D1 receptor to other heterotrimeric G proteins, such as Gαo and Gαq, and of the D5 receptor to Gαz, has also been described (Citation[4], Citation[11]). D1 receptor interactions with Gαq are particularly interesting in light of the possibility that D1 or D1-like receptors activate phospholipase C, a phenomenon that will be discussed in greater detail below.

D1-like Dopamine Receptor Signaling

D1-Like Receptor Stimulation of Adenylate Cyclase

The first biochemical evidence for a dopamine receptor was the identification in 1972 of dopamine-stimulated adenylate cyclase activity and cyclic AMP accumulation in the retina (Citation[12]) and in rat neostriatum (Citation[13]). Gαs and, presumably, Gαolf bind primarily to the C2 cytosolic domain of adenylate cyclase, bringing the C1 and C2 domains together in a way that enhances the catalytic efficiency of the enzyme (Citation[14]). Adenylate cyclase catalyzes the conversion of ATP to cyclic AMP, which binds to the regulatory subunits of the protein kinase A (PKA) holoenzyme to disinhibit the catalytic subunits. The selective concentration of the type 5 subtype of adenylate cyclase in the neostriatum suggested that it mediates dopamine receptor signaling (Citation[15]), a hypothesis confirmed with the creation of adenylate cyclase 5 null mutant mice, in which neostriatal D1 receptor-stimulated adenylate cyclase is markedly diminished (Citation[16], Citation[17]). Thus, in the rodent neostriatum the primary pathway for D1-like receptor signaling is likely to be D1 receptor → Gαolf/γ7 → adenylate cyclase 5 → PKA (). PKA phosphorylates a number of proteins involved in signal transduction and regulation of gene expression (Citation[2], Citation[18]).

Figure 1. D1-like receptor signaling pathways. Stimulatory effects are indicated with a solid line ending in an arrowhead, and inhibitory effects with a dashed line ending in a bar. Intervening steps (e.g., MAPKKK → MAPKK → MAPK) are frequently omitted from the figure for simplicity and ion channels are indicated generically, but are identified individually in . AC5, adenylate cyclase type 5; CREB, cyclic AMP response element binding protein; DARPP-32, dopamine-related phosphoprotein, 32 kDa; MAPK, mitogen-activated protein kinase; NHE, Na+/H+ exchanger; PKA, protein kinase A; PKC, protein kinase C; PLC, phospholipase C; PP1 or PP2A, protein phosphatase 1 or 2A.

Figure 1. D1-like receptor signaling pathways. Stimulatory effects are indicated with a solid line ending in an arrowhead, and inhibitory effects with a dashed line ending in a bar. Intervening steps (e.g., MAPKKK → MAPKK → MAPK) are frequently omitted from the figure for simplicity and ion channels are indicated generically, but are identified individually in Fig. 2. AC5, adenylate cyclase type 5; CREB, cyclic AMP response element binding protein; DARPP-32, dopamine-related phosphoprotein, 32 kDa; MAPK, mitogen-activated protein kinase; NHE, Na+/H+ exchanger; PKA, protein kinase A; PKC, protein kinase C; PLC, phospholipase C; PP1 or PP2A, protein phosphatase 1 or 2A.

D1-Like Receptor Regulation of PKA Substrates

PKA Substrates—DARPP32

DARPP-32 (dopamine and cyclic AMP-regulated phosphoprotein, 32 kDa) is a neostriatum-enriched bifunctional signaling protein that inhibits protein phosphatase 1 (PP1) when phosphorylated on Thr34 by PKA or several other kinases (Citation[19], Citation[20]) and inhibits PKA when phosphorylated on Thr75 by cyclin-dependent kinase 5 (Citation[21]). Because Thr75 is dephosphorylated by the PKA-stimulated protein phosphatase-2A (Citation[22]), D1-like receptor signaling is amplified by both positive feedback and feedforward loops. D1 receptor stimulation simultaneously activates PKA by stimulating the production of cyclic AMP and disinhibits PKA by phosphorylation-dependent activation of protein phosphatase-2A and Thr75 dephosphorylation of DARPP-32. At the same time, D1-like receptor activation of PKA not only stimulates PKA-catalyzed phosphorylation of numerous substrates including some described below but also prevents PP1-catalyzed dephosphorylation of many of the same phosphoproteins by phosphorylating DARPP-32 on Thr34. Studies with DARPP-32 null mutant mice have shown that DARPP-32 contributes to acute D1 receptor-mediated responses, both at the cellular and behavioral levels (Citation[20]). Consistent with a signal amplification role for DARPP-32, many deficits associated with genetic deletion of the PP1 inhibitor are observed only at low doses of dopamine receptor agonist (Citation[23], Citation[24], Citation[25]).

PKA Substrates—Ion Channels

D1-like receptor activation of PKA increases the phosphorylation of numerous voltage- and ligand-gated ion channels by various combinations of direct PKA-catalyzed phosphorylation of channel subunits and DARPP-32-mediated inhibition of PP1. For example, there are at least five potential PKA phosphorylation sites in the LI-II region of the pore-forming α-subunit of voltage-gated Na+ channels (Citation[26]), and activation of DARPP-32 also decreases PP1-catalyzed dephosphorylation at Ser573. Enhanced phosphorylation at Ser573 decreases Na+ currents through a decrease in the open probability of the channel (Citation[26], Citation[27], Citation[28], Citation[29], Citation[30]). D1 receptor stimulation of PKA also decreases K+ currents through several types of inwardly rectifying channels, increases L-type, and decreases N and P/Q type Ca2+ channel activity, increases NMDA receptor activity via phosphorylation of the NR1 subunit, enhances AMPA currents, and modulates GABA currents. Because D1-like receptor regulation of ion channels often involves multiple, incompletely defined signaling pathways in addition to PKA, this topic is discussed in greater detail below.

PKA Substrates—CREB

D1 receptor stimulation induces the expression of a number of transcription factors (Citation[31], Citation[32]), at least some of which are dependent on initial activation of the transcription factor cyclic AMP response element-binding protein (CREB) (Citation[31], Citation[33]). D1 receptor activation of CREB involves PKA-dependent CREB phosphorylation of Ser133, which permits binding of CREB to CREB-binding protein and transcriptional activation of genes with cyclic AMP response elements (Citation[34]). D1 receptor stimulation may also increase Ser133-phosphorylated CREB via activation of extracellular signal-regulated kinase (ERK) (Citation[35]). Phosphorylation of CREB is also regulated by Ca2+, so the transcription factor is likely to be an important site of integration for signals from dopamine and N-methyl-D-aspartate (NMDA) glutamate receptors (). CREB-mediated changes in gene expression are important in synaptic plasticity and probably contribute to synaptic rearrangements that underlie the persistence of drug addiction (Citation[36]).

D1-Like Receptor Stimulation of Phospholipase C

One finding that is difficult to reconcile with a model of D1 receptor signaling that includes a central role for a cyclic AMP/PKA cascade is that a null mutation of adenylate cyclase 5 enhances D1 agonist-stimulated locomoter activity even though D1 receptor stimulation of adenylate cyclase activity is almost abolished (Citation[16], Citation[17]). Mice with genetic deletion of adenylate cyclase 5 have altered abundance of several proteins involved in dopamine receptor signaling and also disrupted D2 receptor signaling, making interpretation of behavioral effects complicated, but one possible explanation is that a cyclic AMP-independent signaling pathway mediates D1 receptor locomotor activation, and perhaps other behavioral effects of D1 receptor stimulation. One such alternative pathway for D1-like receptor signaling is phospholipase C-mediated mobilization of intracellular calcium (Citation[37]). There are at least two distinct potential mechanisms for D1-like receptor activation of phospholipase C. Bergson and colleagues demonstrated that heterologously expressed D1 and D5 dopamine receptors, when co-expressed with calcyon, stimulate the release of calcium from intracellular stores following priming of the cells with a Gαq-coupled receptor agonist (Citation[38]). The effect of calcyon is dependent on its binding to the C terminus of the D1 receptor. Endogenous D1-like receptors in neocortical or hippocampal neurons, but not neostriatal neurons, display a similar priming-dependent ability to mobilize calcium (Citation[39]). Whether this involves calcyon-dependent enhancement of coupling to Gαq and direct activation of phospholipase C is unknown, although the lack of effect of cyclic AMP analogues indicates that it is not mediated by PKA.

A second potential cyclic AMP-independent mechanism for D1-like receptor signaling invokes a novel SCH23390-binding receptor that is linked to phospholipase C via Gαq (Citation[40], Citation[41]). The regional distribution and pharmacological profile of D1-like receptor-stimulated phospholipase C differ from both D1 and D5 receptors. Some compounds efficacious for stimulating adenylate cyclase are weak agonists or antagonists at the phospholipase C-coupled receptor, whereas the drug SKF83959 has little efficacy for adenylate cyclase but is one of the most potent and efficacious agonists known to stimulate the phospholipase C-coupled receptor (Citation[42], Citation[43]). D1-like receptor-stimulated phospholipase C is most abundant in the amygdala and hippocampus (Citation[40]); it is of interest that the amygdala has a relatively dense dopaminergic innervation and abundant D1 receptors but little or no D1 receptor-stimulated adenylate cyclase (Citation[44]). D1-like receptors are also functionally and physically coupled to Gαq, with the distribution of Gαq-coupled receptors roughly paralleling that of D1-like receptor-stimulated phospholipase C (Citation[4], Citation[41], Citation[45]). This putative D1-like Gαq-coupled receptor does not react with a D1 receptor antibody (Citation[4], Citation[45]) and is not a product of the drd1 gene because D1-like receptor-stimulated phospholipase C is spared in D1 receptor null mutant mice (Citation[46]); indeed, the existence of a genetically distinct phospholipase C-coupled D1-like receptor has been used to explain the conservation of D1 agonist-stimulated behaviors in D1 null mutant mice (Citation[47]), although the mixed genetic background of the mice also affected the observed phenotype (Citation[48]). A major inconsistency in this otherwise intriguing hypothesis is that [3H]SCH23390 binding sites are undetectable in most brain regions of the D1 null mutant mouse (Citation[49]), including in amygdala where stimulation of phospholipase C is most robust, a finding difficult to reconcile with numerous reports that SCH23390 is a potent antagonist ligand of the Gαq- and phospholipase C-coupled D1-like receptor.

D1-Like Receptor Regulation of Ion Channels

D1-Like Receptor Regulation of K+ Channels

Activation of D1-like or D2-like receptors has opposing effects on K+ currents in most cells. In general, D1-like receptors attenuate these currents via stimulation of the PKA-DARPP-32 signaling cascade, whereas the D2-like receptors enhance them via inhibition of that pathway as well as by release of Gβγ subunits (). We focus here on voltage-gated (IA/IAf, ID/IAs/IKS, IK/IKSS) and inwardly rectifying potassium currents that primarily act to regulate firing rate, stabilize membrane potential, and integrate subthreshold inputs.

Figure 2. Regulation of ion channels by D1-like (top) and D2-like (bottom) dopamine receptors. Stimulatory (solid line with arrowhead) or inhibitory (dashed line with bar) effects of dopamine receptors on GABA receptors (GABA), NMDA glutamate receptors (NMDA), AMPA glutamate receptors (AMPA), L- and N,P,Q-type Ca2+ channels, persistent (P) and transient (T) Na+ channels, G protein-regulated inwardly rectifying K+ channels (GIRK), and voltage-gated K+ channels (VGK+C) are schematically depicted, as well as whether the effect is thought to be mediated by protein kinase A (PKA), protein kinase C (PKC), G protein βγ subunits (Gβγ), or inhibition of the cyclic AMP/PKA pathway (↓PKA).

Figure 2. Regulation of ion channels by D1-like (top) and D2-like (bottom) dopamine receptors. Stimulatory (solid line with arrowhead) or inhibitory (dashed line with bar) effects of dopamine receptors on GABA receptors (GABA), NMDA glutamate receptors (NMDA), AMPA glutamate receptors (AMPA), L- and N,P,Q-type Ca2+ channels, persistent (P) and transient (T) Na+ channels, G protein-regulated inwardly rectifying K+ channels (GIRK), and voltage-gated K+ channels (VGK+C) are schematically depicted, as well as whether the effect is thought to be mediated by protein kinase A (PKA), protein kinase C (PKC), G protein βγ subunits (Gβγ), or inhibition of the cyclic AMP/PKA pathway (↓PKA).

The family of outwardly rectifying voltage-gated K+ channels in neocortical neurons has been separated into three different components based on their inactivation kinetics (IAf, IKS, IKSS). The terminology differs among studies so that it can sometimes be difficult to understand exactly which component is being measured. Here, we have separated the components according to the criteria developed by Foehring and Surmeier (Citation[50]). IAf is the rapidly inactivating component of this current and can also be referred to as the fast A-type current (IA) because it shows fast inactivation kinetics (around 12 msec at −10 mV). IKS, also referred to as ID, IAs, or the slow A-type current, is the slowly inactivating component of this voltage-gated K+ current and has an inactivation time constant of 293 msec at −10 mV. Finally, IKSS, also referred to as IK, is the very slowly inactivating component, with an inactivation time constant between 2 and 20 sec.

In medium spiny neurons of the striatum and nucleus accumbens, the IA type current is important for regulation of firing rate. This current has at least two components that are activated depending on the membrane potential of the cell. The fast component (IAf) is activated at more depolarized membrane potentials and does not appear to be modulated by D1 receptor stimulation. However, at more hyperpolarized membrane potentials, such as during the down state, the slower component of this current (IKS/IAs/ID) is available. In the nucleus accumbens, combined activation of D1 and D2 receptors increases neuronal firing rate by inhibition of ID. The precise signaling mechanism used by each receptor has not been worked out, but this response is dependent on activation of cAMP/PKA and on D2-like receptor-mediated liberation of Gβγ. These pathways converge on the A-type K+ current to attenuate the outward flow of K+ ions (Citation[51]). These authors speculated that D1-Gαs/Gαolf and D2-Gβγ are acting cooperatively to increase adenylate cyclase and PKA activity and produce the observed changes in potassium currents (Citation[52], Citation[53]). This is one of several mechanisms that may contribute to D1/D2 receptor synergism (Citation[54]).

D1-like suppression of K+ currents in the prefrontal cortex has important effects on the excitability of both interneurons and pyramidal cells; elucidating these effects is important for understanding how dopamine will modulate these cortical activity states. D1 stimulation increases evoked GABA release from interneurons via an increase in their excitability. This increase in excitability appears to be due to D1-PKA-stimulated decrease of K+ currents in one class of interneurons, the so-called fast-spiking parvalbumin positive cells (Citation[55]). Dopamine via D1-like receptors suppresses an inwardly rectifying K+ current, a K+-dependent leak current, and the slowly inactivating membrane outward rectification in prefrontal cortex fast-spiking interneurons. Collectively, these D1-mediated modulations of K+ currents act to depolarize and increase the evoked output of a specific subclass of interneurons. Pyramidal cell excitability is regulated by D1 receptor- and PKA-mediated decreases in K+ currents in a similar manner. D1 reduction of the voltage-gated, slowly inactivating K+ current (ID) in pyramidal cells results in a stronger effect of subthreshold inputs because a decrease in IK will lesson opposition to depolarization and latency to first spike (Citation[56], Citation[57]). These D1 effects on pyramidal cells may be mediated through elevation of PKA and increased phosphorylation of DARPP-32. In contrast, the inwardly rectifying K+ channels may be modulated in a slightly different manner from other K+ currents. It appears that D1-like receptor stimulation inhibits voltage-gated K+ channels as a result of direct binding of cyclic AMP to the channel, thus increasing cell excitability (Citation[58], Citation[59]). Overall, in the prefrontal cortex, dopamine acts via D1-like receptors to increase the excitability of both pyramidal neurons and fast-spiking interneurons through inhibition of several K+ currents.

D1-Like Receptor Regulation of Ca2+ Channels

Dopamine modulates high-voltage-activated Ca2+ currents in several types of vertebrate and invertebrate neurons in vitro (Citation[60], Citation[61], Citation[62], Citation[63]). D1 receptor stimulation increases L-type and decreases N, P/Q-type Ca2+ channel conductances in most brain areas (). In cortical cells L-type channels are associated with the somatic region of cells and exert a powerful effect on signal integration. N- and P/Q-type channel activation produces Ca2+ spikes mainly within more distal dendritic regions of somotosensory (Citation[64]) but not prefrontal cortical neurons (Citation[65]).

In striatum, D1-like receptor-mediated decreases in N- and P/Q-type conductances inhibit spike-induced Ca2+ influx, whereas increases in L-type currents lead to depolarization (Citation[62]). This effect on L-type channels becomes even more pronounced in conjunction with the D1-mediated decrease in K+ currents discussed above and the increase in NMDA current. D1 receptor-stimulated increases in L-type conductances, which are very slow to inactivate, might help to stabilize the prolonged depolarizations (Citation[63]).

D1-like receptors decrease N- and P/Q-type Ca2+ channels via activation of PKA and DARPP-32 (Citation[62]). The α1 subunit of these channels contains phosphorylation sites for PKA, but phosphorylation of these sites by PKA activates the channels (Citation[66]). Surmeier et al. (Citation[62]) speculate that the D1-mediated decrease in N- and P/Q-type currents is therefore due not to a direct phosphorylation of the channel by PKA, but rather to a transient increase in PP1-catalyzed dephosphorylation caused by PKA-dependent phosphorylation of PP1-targeting proteins. This effect would be rapidly reversed by comcomitant PKA phosphorylation of DARPP-32, which attenuates PP1 dephosphorylation of the channel (Citation[67]). According to this hypothesis, however, D1-like receptor inhibition of these channels should be prolonged in DARPP-32 null mutant mice; in contrast, D1 agonist-induced inhibition of the channels is decreased in DARPP-32 null mutant mice (Citation[23]).

D1-like receptor effects on Ca2+ channels in the prefrontal cortex are similar to those in the striatum, although the pathways have not been completely worked out, and appear to serve as a gain control mechanism to amplify somatic inputs via increases in L-type Ca2+ currents and attenuate dendritic inputs via decreases in N- and P/Q-type currents (Citation[56], Citation[58]). Direct D1-like receptor stimulation induces a complex modulation of Ca2+ potentials in prefrontal cortex neurons. D1-like agonists suppress full Ca2+ spikes via a Ca2+- and PKC-dependent pathway (Citation[56], Citation[68]). In contrast, subthreshold depolarizations produced by L-type Ca2+ channels (Citation[65]) are augmented transiently (∼7 min) by D1-like receptor agonists (Citation[68]), an effect that is blocked by PKA inhibitors. In this way, D1-like receptor stimulation activates PKA to potentiate subthreshold L-type Ca2+ currents, yet it acts via PKC to suppress large amplitude Ca2+ spikes, thereby tuning Ca2+ currents to have the greatest activation in the voltage range necessary to produce spikes. Coupled with the D1 receptor-mediated increase in INap (see below) and decrease in K+ currents, D1 receptor activation greatly prolongs the output of prefrontal pyramidal neurons.

D1-Like Receptor Regulation of Na+ Channels

Voltage-gated Na+ channels are important for determining threshold for action potential initiation, the duration and frequency of firing, EPSP amplification, and resonance properties of neurons (Citation[69], Citation[70], Citation[71]). D1-like receptor stimulation exerts a powerful influence over cellular activity through manipulation of the phosphorylation state of these channels. D1 effects may differ depending on the specific type of Na+ channel activated and the brain area in which DA is acting.

In the neostriatum and hippocampus, down-regulation of transient Na+ currents by a D1-PKA-DARPP-32 pathway that increases phosphorylation of Ser573 of the Na+ channel α-subunit decreases excitability of cells and increases rheobase current by increasing the action potential threshold to more depolarized levels (Citation[20], Citation[23], Citation[26], Citation[27], Citation[72], Citation[73], Citation[74]). The functional effect of this modulation is to decrease the output of these neurons, but the modulation may be state dependent. Neostriatal cells have rhythmic oscillations in membrane potential (up- and downstates), and it appears that D1 receptor activation creates more selectivity for depolarizing inputs that will evoke an upstate because it opposes the activation of Na+ currents (Citation[27]). As a result, depolarizing inputs must be stronger to evoke a postsynaptic response and thus cells are more selective about which inputs drive upstates. The D1-PKA-DARPP-32 modulation of striatal Na+ channels, therefore, tends to counteract the effectiveness of depolarizing inputs that drive these neurons into upstates.

In neocortical neurons, one type of voltage-gated Na+ channel carries two Na+ currents by virtue of its ability to switch between gating modes (Citation[75], Citation[76], Citation[77]). The fast-gating mode is similar to the transient Na+ current discussed above, whereas the other current is a slowly inactivating or persistent Na+ current (INaP) (Citation[78], Citation[79]). Individual Na+ channels can exhibit this persistent gating mode, but only a small fraction of the channels are in this gating mode at any time.

As in neostriatal neurons, D1-like receptor stimulation of PKA decreases transient Na+ currents in neocortical pyramidal cells (Citation[80]). On the other hand, D1-like receptors may enhance INaP (Citation[56], Citation[81]), decrease it (Citation[82]), or have no effect (Citation[80]). The D1-mediated modulation reported by Gorelova and Yang (Citation[81]) occurs at subthreshold membrane potentials by shifting INaP activation to more negative membrane potentials and slowing its inactivation (Citation[56], Citation[81]). Rather than being mediated by PKA, the D1 receptor effect on INaP in intact prefrontal cortical neurons from brain slices is mediated by PKC (Citation[81]). Maurice et al. (Citation[80]), who observed no effect of D1-like receptor activation on INaP in acutely dissociated prefrontal cortical neurons, proposed that the Nav1.6 α-subunit, which lacks the phosphorylated residue Ser573, contributes disproportionately to the persistent current, whereas the transient current is attributable to the PKA-sensitive Nav1.1/1.2 channel, again supporting the notion that modulation of the persistent current is not through PKA ().

Figure 3. D2-like receptor signaling pathways. Stimulatory effects are indicated with a solid line ending in an arrowhead, and inhibitory effects with a dashed line ending with a bar. Intervening steps (e.g., MAPKKK → MAPKK → MAPK, or the multiple possible steps between Gβγ and RTK) are frequently omitted from the figure for simplicity. See for more specific description of ion channels. AA, arachidonic acid; AC2 or AC5, adenylate cyclase type 2 or 5; CREB, cyclic AMP response element-binding protein; DARPP-32, dopamine- and cyclic AMP-regulated phosphoprotein, 32 kDa; MAPK, mitogen-activated protein kinase; NHE, Na+/H+ exchanger; PA, phosphatidic acid; PC, phosphatidylcholine; PI3K, phosphatidylinositol 3-kinase; PKA, protein kinase A; PKC, protein kinase C; PLA2, phospholipase A2; PLC, phospholipase C; PLD, phospholipase D; PP1 or PP2A, protein phosphatase 1 or 2A; RTK, receptor tyrosine kinase.

Figure 3. D2-like receptor signaling pathways. Stimulatory effects are indicated with a solid line ending in an arrowhead, and inhibitory effects with a dashed line ending with a bar. Intervening steps (e.g., MAPKKK → MAPKK → MAPK, or the multiple possible steps between Gβγ and RTK) are frequently omitted from the figure for simplicity. See Fig. 2 for more specific description of ion channels. AA, arachidonic acid; AC2 or AC5, adenylate cyclase type 2 or 5; CREB, cyclic AMP response element-binding protein; DARPP-32, dopamine- and cyclic AMP-regulated phosphoprotein, 32 kDa; MAPK, mitogen-activated protein kinase; NHE, Na+/H+ exchanger; PA, phosphatidic acid; PC, phosphatidylcholine; PI3K, phosphatidylinositol 3-kinase; PKA, protein kinase A; PKC, protein kinase C; PLA2, phospholipase A2; PLC, phospholipase C; PLD, phospholipase D; PP1 or PP2A, protein phosphatase 1 or 2A; RTK, receptor tyrosine kinase.

D1-Like Receptor Regulation of Glutamate Receptors

D1-like receptors interact with glutamate receptors at multiple levels, including signal integration by CREB and PKA-dependent phosphorylation of the glutamate receptors. D1-like receptors increase NMDA receptor-mediated responses in neostriatal (Citation[83], Citation[84], Citation[85], Citation[86]), hippocampal (Citation[87]), and cortical neurons (Citation[88], Citation[89], Citation[90], Citation[91]) and enhance phosphorylation of the NMDA-NR1 subunit. DARPP-32 is required for D1 receptor-stimulated phosphorylation of NR1 and for the increased NMDA-evoked responses (Citation[20], Citation[86], Citation[92]). A contribution of D1-like receptor activation of L-type Ca2+ channels and protein kinase C has also been described (Citation[93], Citation[94]), whereas other studies have focused on the role of PKA (Citation[95]). PKA/DARPP-32 may enhance NMDA receptor function by a complex combination of mechanisms including direct phosphorylation of NR1 by PKA, DARPP-32-mediated inhibition of dephosphorylation of NR1, and depolarization via DARPP-32-mediated activation of L-type Ca2+ channels (Citation[20], Citation[91]).

An additional consideration is that D1 effects may be state dependent. If the target cell is in a depolarized state, then D1 receptor stimulation increases NMDA currents, but if it is in a hyperpolarized state when dopamine is applied, then D1-like receptor activation decreases NMDA receptor currents (Citation[27], Citation[96], Citation[97]). A positive feedback situation may exist in that NMDA receptor activation in depolarized cells may benefit from the simultaneous D1 receptor-mediated modulation of INap, Ca2+ channels, and NMDA receptors; greater depolarization is produced, these voltage-dependent currents become larger, thus evoking more depolarization, thereby enhancing the subtle actions of dopamine.

Dopamine appears to exert opposing effects on AMPA currents via activation of D1 and D2 receptors. Both receptor subtypes can activate kinases that target specific residues on AMPA receptors, but as for NMDA receptors, experiments to elucidate signaling pathways regulating these voltage-dependent receptors are complicated by the necessity to exclude dopamine receptor effects on other ion currents.

D1 receptor activation may have variable effects on AMPA currents in the striatum, with a small effect to enhance the currents, and a larger effect to stabilize them by delaying AMPA receptor current “rundown” (Citation[98], Citation[99], Citation[100]). The stabilization may be mediated by both PKA-catalyzed phosphorylation of the GluR1 subunit at Ser845 and DARPP-32 inhibition of PP1-catalyzed dephosphorylation of the residue (Citation[100], Citation[101], Citation[102], Citation[103]), whereas enhanced amplitude may reflect D1-like receptor activation of L-type Ca2+ channels (Citation[62], Citation[93], Citation[99]), also mediated by PKA/DARPP-32. Thus, D1 receptor-mediated enhancement of AMPA currents would depend on the co-presence of the Ca2+ channels, perhaps accounting for some variability in results. To complicate matters further, there is some debate about whether the effects of D1 stimulation occur pre- or postsynaptically. In prefrontal cortex, D1 receptor stimulation attenuates AMPA currents by decreasing the probability of glutamate release from presynaptic cells. This implies that rather than a direct effect of D1 receptor activation on AMPA receptors, decreased synaptic glutamate levels manifest themselves as decreases in AMPA currents (Citation[89]). Later work supported this idea by removing the presynaptic component of this response. When only the postsynaptic effect is examined, then D1-like receptor stimulation slightly increases AMPA currents via a Ca2+-dependent mechanism (Citation[104]).

D1-like receptor stimulation also enhances the trafficking of NMDA receptors and the GluR1 subunit of AMPA receptors to the membrane in neurons (Citation[105], Citation[106]). Enhanced trafficking of GluR1 is mimicked by forskolin-induced activation of adenylate cyclase (Citation[106]), whereas that of NMDA receptors is independent of DARPP-32 but requires the Src-family protein tyrosine kinase Fyn (Citation[107]). Finally, a direct protein-protein interaction between the C termini of the D1 receptor and the NR1-1a and NR2A NMDA receptor subunits mediates reciprocal regulation of receptor function and trafficking (Citation[108], Citation[109], Citation[110]). In particular, this physical interaction permits D1 receptor inhibition of NMDA receptor currents (Citation[108]).

D1-Like Receptor Regulation of GABA Receptors

Dopamine exerts varied effects on GABAA currents depending on the brain area and cell type being examined. One reason for these various effects could be the subunit composition of the GABA receptors located in these different areas, because different isoforms of α-, β-, and γ-subunits are differentially affected by protein kinases. D1-like receptor stimulation decreases GABA receptor activation in medium spiny neurons from neostriatum and nucleus accumbens (Citation[24], Citation[111]), an effect associated with phosphorylation of β1/β3 subunits and activation of PKA/DARPP-32 (Citation[24]). In large cholinergic interneurons of the neostriatum, D1-like agonists have been reported to have no effect on GABA receptor IPSPs (Citation[112]) or to enhance the activity of a subpopulation of zinc-sensitive GABAA receptors via the D5 receptor, PKA, and PP1 (Citation[113]).

As for the D1 and NMDA receptors, mutually inhibitory modulation between D5 and GABAA receptors results from a protein-protein interaction between the D5 receptor and the γ2 subunit of the GABAA receptor (Citation[114]). These direct interactions between D1-like and ion channel-coupled receptors may represent G protein-independent mechanisms of signaling, although it is also possible that G protein activation is required for formation of the multiprotein signaling complex.

D1-Like Receptor Regulation of Other Signaling Pathways

Inhibition of Na+-,K+-ATPase

D1-like receptors inhibit Na+-,K+-ATPase in many peripheral and neural tissues (Citation[115], Citation[116]). Multiple mechanisms mediate this effect. Activation of DARPP-32 plays a key role in both renal tubule cells (Citation[117]), where inhibition of Na+-, K+-ATPase contributes to D1 receptor-stimulated natriuresis and diuresis, and neostriatal neurons (Citation[23]). Phospholipase C-dependent mechanisms have also been described (Citation[118], Citation[119]), and D1-like receptor inhibition of Na+/H+ exchange would also be expected to inhibit Na+-,K+-ATPase activity by decreasing the intracellular concentration of Na+ (for an extensive review, see 115).

Activation of Mitogen-Activated Protein Kinases

Several reports describe D1-like receptor activation of mitogen-activated protein (MAP) kinases, including ERK (Citation[35], Citation[120]), p38 MAP kinase, and c-jun amino-terminal kinase (Citation[121]). Regulation of the latter two MAP kinase pathways is mediated by PKA, whereas D1-like receptor activation of ERK may be partially independent of PKA or cyclic AMP. One possible PKA-independent but cyclic AMP-dependent mechanism for activation of ERK by the D1 receptor is activation of the Rap GTPase (Citation[122]) by the cyclic AMP-activated guanine nucleotide-exchange factor Epac (Citation[123]).

D2-Like Receptors and G Proteins

D2-like receptor signaling is mediated primarily by activation of the heterotrimeric G proteins Gαi/o, a class of G proteins inactivated by pertussis toxin-catalyzed ADP-ribosylation (Citation[124], Citation[125]). For the D2 receptor, the possibility that the alternatively spliced insert in the third cytoplasmic loop of D2L might influence G protein interactions and result in differential G protein selection by D2S and D2L has meant that analyses of G protein selection often focus on comparisons between the two isoforms. There is considerable disagreement in the literature concerning which G proteins interact with D2S and D2L (Citation[126], Citation[127]). It seems likely that both receptor isoforms are inherently able to activate multiple Gαi/o subtypes, including Gαi2, Gαi3, and Gαo (Citation[128], Citation[129]), but that interactions with particular G proteins are restricted in a cell-type dependent manner due to compartmentalization or the availability of appropriate effectors and scaffolding proteins. D2S and D2L can also activate the pertussis toxin-insensitive G protein Gαz (Citation[130], Citation[131]). This interaction could explain some reports of pertussis toxin-insensitive signaling by the D2 receptor (Citation[132], Citation[133], Citation[134]). Nevertheless, accumulating evidence from a variety of approaches has identified Gαo as the Gαi/o subtype that is most robustly activated by D2L (Citation[135], Citation[136], Citation[137]) and by D2S (Citation[138], Citation[139], Citation[140]) and, furthermore, the G protein subtype that is predominantly coupled to D2-like receptors in mouse brain (Citation[141]).

The human D4 receptor is similar to D2 in that it activates multiple pertussis toxin-sensitive G proteins, including Gαi2, Gαi3, and Gαo (Citation[142], Citation[143]). The rat D4 receptor has been reported to couple preferentially to Gαz (Citation[131]) and to the pertussis toxin-sensitive transducin subtype, Gαt2 (Citation[144]). Work by several groups has identified Gαo as being activated by the D3 receptor and mediating D3 signaling, with some evidence for D3 receptor signaling via Gαz and Gαq/11 (Citation[131], Citation[142], Citation[145], Citation[146]). An example of the importance of cell-type specific factors in the selective activation of G proteins by GPCRs is that the D3 receptor couples more efficiently to Gαo in SH-SY5Y cells than in HEK293 cells, despite the abundance of that G protein subtype in both cell lines (Citation[146]). Zaworski et al. (Citation[146]) suggest that the additional presence in SH-SY5Y cells of effectors regulated by the D3 receptor contributes to the efficient activation of Gαo by the D3 receptor in those cells, a hypothesis consistent with other work showing that receptors form complexes with effectors and that G proteins participate in complex formation (Citation[147]).

An unusual feature of the D3 receptor is that it binds agonists with a high affinity that is relatively insensitive to GTP (Citation[126]). The affinity of GPCRs for agonists is typically reduced by GTP, reflecting GTP-induced destabilization of the agonist-receptor-G protein ternary complex. The insensitivity of the D3 receptor to GTP could reflect GTP-resistant coupling to G proteins or a receptor structure that is constrained in a comformation with high affinity for agonists regardless of interactions with G proteins; interesting work by Leysen and colleagues expressing the D3 receptor in E. coli, and thus in the absence of endogenous G proteins with which the receptor can interact, indicates that the latter explanation is more likely (Citation[148]).

D2-like Dopamine Receptor Signaling

D2-Like Receptor Signaling via Gαi/o—Inhibition of Adenylate Cyclase

The first signaling pathway identified for D2-like receptors was inhibition of cyclic AMP accumulation (Citation[149], Citation[150]). As for D1 receptor stimulation of adenylate cyclase, genetic deletion of adenylate cyclase 5 abolishes D2 receptor-mediated inhibition of adenylate cyclase in the mouse neostriatum. That this null mutation also eliminates the locomotor inhibitory effects of D2 receptor-blocking antipsychotic drugs shows the behavioral significance of this signaling pathway (Citation[16]). The lack of responsiveness to antipsychotic drugs is a phenotype also seen in D2 receptor (Citation[151]) and DARPP-32 (Citation[20]) null mutant mice, further indicating that this signaling pathway contributes to D2 receptor-stimulated locomotor activity. D2 and D4 receptors inhibit adenylate cyclase activity in a variety of tissues and cell lines (Citation[2], Citation[152]). Inhibition of adenylate cyclase by the D3 receptor is weaker and often undetectable although it is of interest that the D3 receptor robustly inhibits adenylate cyclase type 5 (Citation[153], Citation[154]), in contrast to several other adenylate cyclase subtypes including the closely related type 6. On the other hand, co-expression of D2 and D3 receptors in COS-7 cells with adenylate cyclase 6 substantially increases the potency of D2-like receptor agonists for inhibition of cyclic AMP accumulation, suggesting that the D2/D3 heteromer has increased potency for agonists and/or couples more efficiently to adenylate cyclase 6 (Citation[154], Citation[155]).

D2-like receptor inhibition of adenylate cyclase is presumed to be mediated by Gαi/o, because adenylate cyclase 5 is directly inhibited by Gαi and is insensitive to Gβγ (Citation[156]). Gαi binds primarily to the C1 cytosolic domain of Gαi-inhibited forms of adenylate cyclase and reduces C1/C2 domain interaction (Citation[157]). One inconsistency is that adenylate cyclase 5 expressed in insect Sf9 cells is insensitive to purified Gαo (Citation[156]), whereas work cited above supports a prominent D2 → Gαo → adenylate cyclase 5 signaling path.

D2 receptor signaling via inhibition of adenylate cyclase would be expected to act in opposition to agents that stimulate adenylate cyclase, decreasing the phosphorylation of PKA substrates (). For example, stimulation of D2-like receptors decreases PKA-stimulated phosphorylation of DARPP-32 at Thr34 and increases phosphorylation at Thr75 (Citation[22], Citation[158]). Both of these effects may be at least partially mediated by Gαi-dependent inhibition of cyclic AMP, although for Thr34, the lack of effect of quinpirole in Ca2+-free medium is difficult to reconcile with a major role for adenylate cyclase inhibition; calcium-dependent stimulation of calcineurin (protein phosphatase 2B) also contributes to D2 receptor dephosphoryation of Thr34 (Citation[158]). It is of interest that the opposing effects of D1 and D2 receptors on DARPP-32 both lead to inhibition of the Na+,K+-ATPase in neostriatal neurons (Citation[159]). Another response potentially mediated by D2-like receptor inhibition of adenylate cyclase is autoreceptor suppression of tyrosine hydroxylase activity. Stimulation of D2-like synthesis-inhibiting autoreceptors reverses PKA-dependent phosphorylation of tyrosine hydroxylase at Ser40 and activation of the enzyme (Citation[160]). Although it is reasonable to speculate that inhibition of adenylate cyclase contributes to this response, a role for other mechanisms, such as calcium-stimulated protein phosphatases, has not been excluded. Inhibition of adenylate cyclase is also likely to be the mechanism of D4 receptor-induced inhibition of GABA currents in rat globus pallidus, perhaps by decreasing PKA-dependent phosphorylation of the β3 subunit of the GABAA receptor (Citation[161]), and the proximal mechanism of D4 receptor-induced inhibition of NMDA receptor currents in rat prefrontal cortex, for which the complete pathway is apparently disinhibition of PP1, dephosphorylation of autophosphorylated calcium-, calmodulin-dependent kinase II thus reducing its calcium-independent kinase activity, and decreased phosphorylation of the NR1 subunit of the NMDA receptor which in turn decreases NMDA receptor expression in the plasma membrane (Citation[162]). On the other hand, inhibition of NMDA receptor transmission by the D4 receptor in rat hippocampus apparently involves platelet-derived growth factor receptor-dependent mobilization of calcium rather than inhibition of PKA-dependent phosphorylation (Citation[163]).

D2-Like Receptor Signaling Mediated by G Protein βγ Subunits

As is typical of Gαi/o-coupled receptors, D2-like receptors modulate many signaling pathways in addition to adenylate cyclase, including phospholipases, ion channels, MAP kinases, and the Na+/H+ exchanger (Citation[2]). Many of these pathways are regulated by G protein βγ subunits that are released by receptor activation of Gαi/o proteins ().

Gβγ-Stimulated Adenylate Cyclase

Gβγ has a permissive effect on adenylate cyclase 2 and 4, so that the stimulatory effect of other activators, including Gαs and protein kinase C, is enhanced in the presence of free Gβγ (Citation[52], Citation[53], Citation[156]). Gβγ binds to residues in several cytosolic domains of adenylate cyclase 2, with stimulation primarily due to binding to the less conserved region of cytosolic loop 1, C1b (Citation[164], Citation[165]). This Gβγ-mediated stimulatory effect is primarily observed for Gαi/o-coupled receptors, perhaps because activation of only this relatively abundant class of G proteins liberates a sufficient concentration of Gβγ subunits. D2 and D4 receptors markedly increase the activity of adenylate cyclase 2, whereas the D3 receptor has little or no effect (Citation[53], Citation[153]). D2-like receptor activation of Gβγ-stimulated adenylate cyclase response has only been observed by using heterologously expressed receptors and adenylate cyclase. It is not known if it contributes to D2-like receptor signaling in neurons, although it has been speculated that Gβγ-stimulated adenylate cyclase contributes to synergistic activation of spike firing in nucleus accumbens neurons by D1-like and D2-like receptors (Citation[51]).

Gβγ-Stimulated K+ Channels

D2 stimulation exerts a powerful influence over K+ currents likely via dissociation of Gβγ subunits rather than by Gαi-dependent inhibition of adenylate cyclase activity. Unlike the generally excitatory effect of D1 receptor stimulation, D2 stimulation decreases cell excitability by increasing K+ currents in most brain areas ().

All of the D2-like receptors activate a G protein-regulated inwardly rectifying potassium channel (GIRK or Kir3), a channel that carries one of several potassium currents modulated by dopamine in midbrain dopamine neurons (Citation[166], Citation[167]), and neostriatal D2 receptor-expressing neurons (Citation[168]), with activation presumably via Gβγ (Citation[169], Citation[170], Citation[171]). The D3 receptor is approximately as efficient as the D2L receptor at coupling to homomeric GIRK2 (Citation[172]), the GIRK subtype predominantly expressed by dopamine neurons in the rat ventral mesencephalon (Citation[173], Citation[174]), and regulation of GIRK channels contributes to inhibition of secretion by the D3 receptor heterologously expressed in AtT-20 mouse pituitary cells (Citation[175]). D2 and D4 receptors both co-precipitate with GIRK channels in a heterologous expression system, and the rat neostriatal D2 receptor co-precipitates with GIRK2, suggesting the existence of a stable complex that forms during receptor/channel biosynthesis (Citation[147]). The formation of large multiprotein complexes that include GPCRs and their effectors may be a general characteristic of GPCR signaling (Citation[176]). Evidence that dopamine release-regulating autoreceptors are coupled to potassium channels (Citation[177]) rather than to inhibition of adenylate cyclase (Citation[178]), together with the robust regulation of GIRK currents by D2 receptors in substantia nigra dopamine neurons (Citation[179]), suggests that D2 receptor activation of GIRK currents contributes to D2 autoreceptor inhibition of dopamine release and dopamine neuronal activity. The hyperactivity and facilitation of D1 receptor signaling observed in GIRK2 null mutant mice (Citation[180]) is also consistent with a loss of inhibitory autoreceptor function.

The effect of D2 stimulation on voltage-gated outward potassium currents appears to depend on the brain area being examined. Medium spiny neurons in the striatum show an increase in the amplitude of the slowly inactivated K+ current (ID) in the presence of D2 agonists (Citation[181]), but pyramidal neurons of the cortex show no effect of D2 stimulation on this same current (Citation[57]). More work is needed to understand the mechanism by which D2 receptors exert their effects on voltage-gated K+ channels.

Gβγ-Mediated Regulation of Ca2+ Channels

All D2-like receptors decrease the activity L, N, and P/Q-type channels via pertussis toxin-sensitive G proteins (Citation[128], Citation[182], Citation[183], Citation[184], Citation[185]). D2 receptors in neostriatal large aspiny (cholinergic) interneurons inhibit N-type Ca2+ channels by a membrane-delimited pathway that probably involves Gβγ subunits (Citation[186], Citation[187]). Gβγ subunits target the I-II linker region and/or the COOH terminus of the α-subunit of these Ca2+ channels, decreasing the amount of current carried (Citation[188], Citation[189], Citation[190]). D2 receptors in striatal medium spiny neurons decrease L-type Ca2+ currents (Citation[191]). This response is also mediated by Gβγ subunits, although rather than being membrane delimited the pathway involves Gβγ stimulation of cytosolic phospholipase C, mobilization of Ca2+, and activation of calcineurin ().

The functional effects of this modulation are possibly to regulate neurotransmitter release when the N-type Ca2+ channel is targeted; thus, D2 receptor activation of neostriatal interneurons inhibits the release of acetylcholine (Citation[192]), and D2-like receptor stimulation decreases glutamate release in striatum, presumably via effects on N-type Ca2+ channels (Citation[193], Citation[194]). Inhibition of glutamate release may implicate tonic D2 receptor activity in the prevention of excessive glutamate release; together with D2 receptor inhibition of L-type channels in neostriatal medium spiny neurons, this would be expected to promote the downstate in neostriatal projection neurons. Facilitory effects of D1-like receptor stimulation would take over when synchronized inputs arrive drive the cell into the upstate. In this way, D2 receptor activation may serve as a gatekeeper over striatal upstates (Citation[85]). Voltage-dependent Ca2+ channels are inhibited by D2 receptors in the anterior pituitary (Citation[128]) and by the D3 receptor heterologously expressed in AtT-20 cells (Citation[184]); inhibition of Ca2+ channels in these cells would be expected to decrease secretion of pituitary hormones.

Gβγ-Stimulated MAP Kinases

MAP kinases are components of parallel protein kinase cascades that transmit signals from a variety of extracellular stimuli to the cell nucleus, thus participating in cell proliferation, differentiation, and survival (Citation[195]). Like many other GPCRs, including those coupled to Gαi/o (Citation[195], Citation[196]), activation of the D2 receptor stimulates MAP kinases, including the two isozymes of extracellular signal-regulated kinase (ERK) (Citation[197], Citation[198], Citation[199], Citation[200], Citation[201], Citation[202], Citation[203], Citation[204]) and stress-activated protein kinase/Jun amino-terminal kinase (SAPK/JNK) (Citation[199]). D3 (Citation[205]) and D4 (Citation[203], Citation[206]) dopamine receptors also activate ERK. D2-like receptors activate ERK in brain slices (Citation[35], Citation[207]) and in rat brain after administration of agonist in vivo (Citation[208]).

Although the pathway from D2-like receptors to ERK has not been thoroughly elucidated and may differ depending on cell type and receptor subtype, D2-like receptor activation of ERK is frequently mediated by pertussis toxin-sensitive G proteins (Citation[200], Citation[201], Citation[205], Citation[209]), Gβγ (Citation[197], Citation[201], Citation[202]), phosphatidylinositol 3-kinase (Citation[200], Citation[205]), Ras (Citation[199], Citation[206]), and the MAP kinase kinase MEK (Citation[199], Citation[201], Citation[207], Citation[208]). D2-like receptor activation of ERK is in at least some cases mediated by transactivation of a receptor tyrosine kinase (RTK), thus recruiting the RTK-signaling cascade in response to dopamine. Although the epidermal growth factor receptor has frequently been identified as an RTK that is transactivated by GPCRs (Citation[210], Citation[211]), transactivation of the platelet-derived growth factor receptor can be a necessary intermediate step in the activation of ERK by recombinant and endogenous D2 and D4 receptors (Citation[163], Citation[203], Citation[204]). We have observed that the identity of the RTK that is transactivated by D2-like receptors depends on cell type, with D2 receptor stimulation of ERK mediated by the platelet-derived growth factor receptor in nonneuronal cells, but by the epidermal growth factor receptor in neuroblastoma cells and primary neuronal cultures from embryonic rat neostriatum (Citation[209]). It is not clear which of the several identified mechanisms for RTK transactivation (Citation[212]) are used by D2-like receptors, although in some cells D2 receptor stimulation enhances a direct interaction between the D2 receptor and the platelet-derived growth factor receptor (Citation[203]) or the epidermal growth factor receptor (Citation[209], Citation[213]).

D2 receptor activation of ERK stimulates DNA synthesis and mitogenesis in many different cell types (Citation[199], Citation[202], Citation[214], Citation[215]). In post-mitotic neurons, activation of MAP kinases is involved not only in cell survival and in synaptic plasticity (Citation[216], Citation[217], Citation[218]) but also in acute behavioral responses to dopamine receptor stimulation (Citation[208]). D2 receptor signaling to ERK in pituitary lactotrophs may be more complicated; in both primary lactotrophs and a prolactin-secreting cell line, the D2 receptor is reported to inhibit ERK, leading to suppression of prolactin promoter function (Citation[139]). A conflicting report using a different prolactin-secreting cell line describes D2 receptor stimulation of ERK leading to inhibition of cell proliferation (Citation[219]).

Epidermal growth factor receptors signal not only through a Ras-MEK-ERK pathway but also through a phosphoinositide 3-kinase → protein kinase B (Akt) pathway. D2 and D4 receptors activate Akt in several cell types with neuronal characteristics and in immortalized dopaminergic neurons (Citation[206], Citation[213]). In PC12 cells expressing a recombinant D2 receptor, D2 receptor activation of Akt is mediated by Src-dependent transactivation of the epidermal growth factor receptor. This pathway mediates D2 receptor neuroprotection against oxidative stress (Citation[213]).

Other Signaling Pathways Regulated by D2-Like Receptors

D2-Like Receptor Regulation of Phospholipases

D2 receptors in neostriatal medium spiny neurons activate a cytosolic, Gβγ-stimulated form of phospholipase C, PLCβ1, causing inositol trisphosphate-induced calcium mobilization that activates calcium-dependent proteins such as the protein phosphatase calcineurin, ultimately reducing L-type Ca2+ currents (Citation[220]). This pathway may contribute to activation of both ERK and CREB by D2-like receptors in neostriatal neurons (Citation[207]) and also to the PKA-independent regulation of DARPP-32 phosphorylation by D2-like receptors described above (Citation[158]). This pathway and the D4 receptor-stimulated transactivation of the platelet-derived growth factor receptor leading to depression of NMDA receptor signaling in hippocampal neurons have interesting similarities, such as dependence on Gβγ, phospholipase C, and calcium mobilization, which are suggestive of a more general role for RTK transactivation in D2-like receptor signaling.

The D2 receptor potentiates arachadonic acid release induced by calcium-mobilizing receptors in heterologous expression systems (Citation[220], Citation[221], Citation[222]), a response that is mediated by cytosolic phospholipase A2 (Citation[223]). The D4 receptor also activates this pathway (Citation[223]), whereas the D3 receptor has no effect or is inhibitory (Citation[224]). Although the response is inhibited by pertussis toxin, the lack of effect of cyclic AMP and the sensitivity of cytosolic phospholipase A2 to Gβγ (Citation[225], Citation[226]) suggest that this is a Gβ-mediated response. D2 receptor-stimulated arachidonate release in the absence of a calcium-mobilizing agent has also been observed (Citation[227]). Arachidonic acid and its bioactive lipooxygenase and cyclooxygense metabolites (e.g., prostaglandin E2) have numerous effects on cellular function and may feed back to regulate D2-like receptor signaling and uptake (Citation[228], Citation[229], Citation[230], Citation[231]).

Many GPCRs, including the D2 receptor, stimulate phospholipase D, which catalyzes the hydrolysis of phosphatidylcholine to form choline and phosphatidic acid (Citation[232], Citation[233]). The low molecular weight G protein RhoA is required for this D2 receptor-mediated response (Citation[234]), and work with other phospholipase D-activating GPCRs has identified a direct interaction between the receptors and two monomeric G proteins RhoA and ARF (Citation[232]). D2 receptor stimulation of phospholipase D is not mediated by Gαi/o, because stimulation is insensitive to pertussis toxin but does require activation of protein kinase Cε (Citation[233]). Activation of Gα13, which in turn stimulates the Rho guanine nucleotide exchange activator (p115RhoGEF), is a mechanism by which some GPCRs activate phospholipase D (Citation[235], Citation[236], Citation[237]).

D2-Like Receptor Regulation of Na+ Channels

D2-like receptor activation in the neostriatum exerts variable effects on Na+ channels possibly via several intracellular signaling pathways. In the prefrontal cortex, on the other hand, D2-like receptor stimulation does not appear to exert a consistent or strong effect over Na+ channels in some studies (81, but see 238), perhaps because D1-like receptors are somehow in a better position to control these channels or simply more abundant. The strong effect of prefrontal cortical D2-like receptors on other ion channels may reflect differences in scaffolding with these channels.

Surmeier et al. (Citation[27], Citation[181]) discovered that D2-like receptor stimulation can either increase or decrease Na+ currents in neostriatal neurons, perhaps depending on the subtype of D2-like receptors expressed by a given cell. Enhancement of Na+ currents requires a soluble second messenger and may represent a process reciprocal to attenuation of voltage-gated Na+ currents by D1 receptor-stimulated activation of PKA and DARPP-32 (Citation[23], Citation[116]). That is, the D2-like receptor stimulated increase in Na+ current observed in a minority of neostriatal neurons may be due to inhibition of adenylate cyclase and PKA, preventing phosphorylation of the channel. In most D2 agonist-responsive neurons, D2-like receptor stimulation decreases Na+ currents in a membrane-delimited manner (Citation[181]) that may involve binding of Gβγ subunits to the Na+ channels. The Na+ current attenuation involved a shift in inactivation potential to more negative values, with no decrease in the peak amplitude of the current. These results mimic the effects of inhibition of pertussis toxin-sensitive G-proteins (presumably Gi/o) on recombinant Na+ channels in CHO cells that are similar to the type expressed in the striatum (Citation[239]).

D2-Like Receptor Regulation of Na+/H+ Exchange

The Na+/H+ exchangers are a family of integral membrane proteins that regulate intracellular pH and transcellular Na+ absorption (Citation[240]). Heterologously expressed D2 (Citation[133]), D3 (Citation[241], Citation[242]), and D4 receptors (Citation[222]) activate the widely expressed Na+/H+ exchanger NHE1. Like GPCR activation of phospholipase D, D2 receptor activation of Na+/H+ exchange is insensitive to pertussis toxin in some cell lines (Citation[133]), and the G protein Gα13 can activate the exchanger via Rho (Citation[243]). It is of interest that several GPCR subtypes, including endogenous D2-like receptors in primary lactotrophs, mediate pertussis toxin-insensitive and cyclic AMP-independent inhibition of Na+/H+ exchanger activity (Citation[132], Citation[244]). Lin et al. (Citation[244]) have proposed that a potential mechanism for this inhibition is Gα12 competition for the interaction between Gα13 and p115RhoGEF. Na+/H+ exchange is also inhibited by D1-like receptors by both cyclic AMP-dependent and -independent mechanisms (Citation[115], Citation[245]).

D2-Like Receptor Regulation of Receptor-Linked Ion Channels

Glutamatergic neurotransmission is enhanced in D2 receptor null mutant mice, which probably reflects the loss of both presynaptic control of glutamate release and postsynaptic inhibition of glutamatergic responses by the D2 receptor (Citation[246]). D2-like receptor stimulation decreases NMDA currents, but the mechanism for this effect is still debated. One idea is that D2 stimulation results in changes in K+ and Na+ permeabilities that hyperpolarize cells and prevent the removal of the Mg2+ blockade over these channels, resulting in a decrease in the open probability of the channel (Citation[98]). Another idea is that D2 stimulation causes dephosphorylation of the NR1 subunit by antagonizing D1-like receptor stimulation of the DARPP-32 signaling cascade (Citation[92]). In prefrontal cortex, the D4 receptor decreases NMDA currents by a similar mechanism involving disinhibition of PP1 (Citation[162]). Another means by which D2 activation attenuates NMDA currents in hippocampus is via transactivation of the platelet-derived growth factor receptor (Citation[163]).

A consistent role for the D2 receptor has not been described for GABA transmission, perhaps because both D1-like and D2-like receptors modulate GABAergic transmission by altering GABA release (Citation[247]). In addition to decreasing the probability of GABA release in the prefrontal cortex, D2 receptor agonists reduce postsynaptic responsiveness to a GABAA agonist by a mechanism that includes transactivation of the platelet-derived growth factor receptor (Citation[247], Citation[248]). This mechanism is consistent with work showing that activation of the platelet-derived growth factor receptor decreases GABAA currents via activation of phospholipase C and subsequent mobilization of intracellular Ca2+ to alter the phosphorylation state of the receptor (Citation[249]).

Novel Protein–Protein Interactions Involved in D2-Like Receptor Signaling

D2-Like Receptor Signaling via Receptor Heteromerization

We have discussed some functional consequences of dopamine receptor heteromerization with ion channels (D1 and NMDA, D5 and GABAA), receptor tyrosine kinases (D2/D4 and the epidermal growth factor and platelet-derived growth factor receptors), and other dopamine receptor subtypes (D2 and D3) in other contexts in this review. The D2 receptor can also participate in heteromers with other GPCRs including the somatostatin receptor subtype SSTR5 (Citation[251]), and the adenosine A2A receptor (Citation[251], Citation[252]). Intramembrane interactions of the D2 receptor with neurotensin and metabotropic glutamate receptors are also potentially mediated by direct receptor:receptor interactions (Citation[253]), although competition for a limiting pool of another receptor-interacting protein is another possible mechanism. The formation of heteromers can be constitutive (Citation[252]) or stimulated by the binding of ligands, particularly agonists (Citation[250]), and may underlie mutually inhibitory (Citation[253]) or stimulatory (Citation[250]) interactions. There is considerable behavioral and functional evidence for interactions between brain dopamine and adenosine systems and between dopamine and somatostatin, and receptor heteromerization is likely to be a molecular mechanism of these interactions (Citation[254]).

Other Dopamine Receptor-Interacting Proteins That Modulate Signaling

A number of interactions between the third cytoplasmic loop of D2-like receptors and other proteins are likely to influence D2-like receptor signaling. D2 and D3 receptors, but not D1 or D4 receptors, bind the actin-binding protein filamin A, or ABP-280. Zhou and colleagues report that binding is to a segment in the carboxyl terminus of the third cytoplasmic loop, where both D2 and D3 receptors have a potential site of phosphorylation by PKC, and that D2 and D3 receptors expressed in cells that lack ABP-280 have diminished ability to inhibit adenylate cyclase (Citation[255], Citation[256]). Furthermore, PKC-catalyzed phosphorylation of the D2 receptor on Ser358 may inhibit binding of ABP-280, thus attenuating D2 receptor signaling (Citation[255]). The third cytoplasmic loop of the D2 receptor includes a binding site for spinophilin, a scaffolding protein that also binds and targets PP1 to dendritic spines (Citation[257]). PP1 anchoring by spinophilin is critical for modulation of glutamate receptor activity by dopamine (Citation[100]). Calmodulin binds in a calcium-dependent manner to the amino terminal end of the D2 receptor third cytoplasmic loop and inhibits D2 receptor activation of, but not binding to, Gαi (Citation[258]). Proteins, such as Nck, Grb2, and c-Src which contain Src homology 3 (SH3) domains, a modular protein-protein interaction domain that is essential for the formation of functional signaling complexes, bind to the third cytoplasmic loop of the D4 receptor, which has multiple copies of the proline-rich SH3 binding motif (Citation[259]). Several SH3 domain-containing proteins also bind to the D3 receptor, although the site of binding has not been identified (Citation[259], Citation[260]). The functional role of SH3 protein binding to D2-like receptors is unknown, although mutation of the SH3 binding motifs in the D4 receptor causes constitutive internalization of the receptor (Citation[260]), and binding of the protein tyrosine kinase c-Src to other GPCRs has important consequences for receptor signaling and desensitization (Citation[261]).

Conclusion

Dopamine receptor signal transduction pathways are complex, so that just one dopamine receptor subtype can activate multiple effectors by distinct or partially overlapping pathways that have many points of intersection. One of the goals of this review was to show how these effectors work in concert to alter cellular function in response to dopamine receptor activation, an area in which considerable progress has been made in recent years. On the other hand, much less is known about how these cellular processes contribute to the function of neuronal networks and, ultimately, to behavior. Another emerging area that is only touched on in this review, despite its probable importance for GPCR function, is the role of protein-protein interactions and signaling complexes in dopamine receptor signaling. Elucidating the mechanisms and significance of receptor oligomerization, of interactions with signal-switching proteins, such as calcyon, and of interactions with scaffolding proteins that support the formation of the signaling complex and target the complex to particular subcellular domains, will be crucial to understanding dopamine receptor function at the molecular level.

References

  • Neve KA, Neve RL. Molecular biology of dopamine receptors. The Dopamine Receptors, KA Neve, RL Neve. Humana Press, Totawa, NJ 1997; 27–76
  • Huff RM. Signaling pathways modulated by dopamine receptors. The Dopamine Receptors, KA Neve, RL Neve. Humana Press, Totowa, NJ 1997; 167–192
  • Sidhu A. Coupling of D1 and D5 dopamine receptors to multiple G proteins—implications for understanding the diversity in receptor-G protein coupling. Mol Neurobiol 1998; 16: 125–134
  • Jin L-Q, Wang H-Y, Friedman E. Stimulated D1 dopamine receptors couple to multiple Gα proteins in different brain regions. J Neurochem 2001; 78: 981–990
  • Zhuang X, Belluscio L, Hen R. Golfα mediates dopamine D1 receptor signaling. J Neurosci 2000; 20: NIL1–NIL5
  • Hervé D, Le Moine C, Corvol JC, Belluscio L, Ledent C, Fienberg AA, Jaber M, Studler JM, Girault JA. Gαolf levels are regulated by receptor usage and control dopamine and adenosine action in the striatum. J Neurosci 2001; 21: 4390–4399
  • Jones DT, Reed RR. Golf: an olfactory neuron specific-G protein involved in odorant signal transduction. Science 1989; 244: 790–795
  • Corvol JC, Studler JM, Schonn JS, Girault JA, Hervé D. Gαolf is necessary for coupling D1 and A2a receptors to adenylyl cyclase in the striatum. J Neurochem 2001; 76: 1585–1588
  • Wang Q, Jolly JP, Surmeier JD, Mullah BM, Lidow MS, Bergson CM, Robishaw JD. Differential dependence of the D1 and D5 dopamine receptors on the G protein γ7 subunit for activation of adenylylcyclase. J Biol Chem 2001; 276: 39386–39393
  • Watson JB, Coulter PM II, Margulies JE, De Lecea L, Danielson PE, Erlander MG, Sutcliffe JG. G-protein γ7 subunit is selectively expressed in medium-sized neurons and dendrites of the rat neostriatum. J Neurosci Res 1994; 39: 108–116
  • Kimura K, White BH, Sidhu A. Coupling of human D-1 dopamine receptors to different guanine nucleotide binding proteins. Evidence that D-1 dopamine receptors can couple to both Gs and Go. J Biol Chem 1995; 270: 14672–14678
  • Brown JH, Makman MH. Stimulation by dopamine of adenylate cyclase in retinal homogenates and of adenosine-3′:5′-cyclic monophosphate formation in intact retina. Proc Natl Acad Sci USA 1972; 69: 539–543
  • Kebabian JW, Petzold GL, Greengard P. Dopamine-sensitive adenylate cyclase in caudate nucleus of rat brain, and its similarity to the “dopamine receptor.”. Proc Natl Acad Sci USA 1972; 69: 2145–2149
  • Tesmer JJG, Sunahara RK, Gilman AG, Sprang SR. Crystal structure of the catalytic domains of adenylyl cyclase in a complex with Gsα.GTPγS. Science 1997; 278: 1907–1916
  • Glatt CE, Snyder SH. Cloning and expression of an adenylyl cyclase localized to the corpus striatum. Nature 1993; 361: 536–538
  • Lee KW, Hong JH, Choi IY, Che YZ, Lee JK, Yang SD, Song CW, Kang HS, Lee JH, Noh JS, Shin HS, Han PL. Impaired D2 dopamine receptor function in mice lacking type 5 adenylyl cyclase. J Neurosci 2002; 22: 7931–7940
  • Iwamoto T, Okumura S, Iwatsubo K, Kawabe JI, Ohtsu K, Sakai I, Hashimoto Y, Izumitani A, Sango K, Ajiki K, Toya Y, Umemura S, Goshima Y, Arai N, Vatner SF, Ishikawa Y. Motor dysfunction in type 5 adenylyl cyclase-null mice. J Biol Chem 2003; 278: 16936–16940
  • Demchyshyn LL, O’Dowd BF, George SR. Structure of mammalian D1 and D5 dopamine receptors and their function and regulation in cells. Dopamine Receptors and Transporters: Function, Imaging, and Clinical Implication, A Sidhu, M Laruelle, P Vernier. Marcel Dekker, Inc., New York 2003; 45–76
  • Hemmings HC, Jr, Greengard P, Tung HY, Cohen P. DARPP-32, a dopamine-regulated neuronal phosphoprotein, is a potent inhibitor of protein phosphatase-1. Nature 1984; 310: 503–505
  • Greengard P, Allen PB, Nairn AC. Beyond the dopamine receptor: the DARPP-32/Protein phosphatase-1 cascade. Neuron 1999; 23: 435–447
  • Bibb JA, Snyder GL, Nishi A, Yan Z, Meijer L, Fienberg AA, Tsai LH, Kwon YT, Girault JA, Czernik AJ, Huganir RL, Hemmings HC Jr, Nairn AC, Greengard P. Phosphorylation of DARPP-32 by Cdk5 modulates dopamine signalling in neurons. Nature 1999; 402: 669–671
  • Nishi A, Bibb JA, Snyder GL, Higashi H, Nairn AC, Greengard P. Amplification of dopaminergic signaling by a positive feedback loop. Proc Natl Acad Sci USA 2000; 97: 12840–12845
  • Fienberg AA, Hiroi N, Mermelstein PG, Song WJ, Snyder GL, Nishi A, Cheramy A, O’Callaghan JP, Miller DB, Cole DG, Corbett R, Haile CN, Cooper DC, Onn SP, Grace AA, Ouimet CC, White FJ, Hyman SE, Surmeier DJ, Girault JA, Nestler EJ, Greengard P. DARPP-32: Regulator of the efficacy of dopaminergic neurotransmission. Science 1998; 281: 838–839
  • Flores-Hernandez J, Hernandez S, Snyder GL, Yan Z, Fienberg AA, Moss SJ, Greengard P, Surmeier DJ. D1 dopamine receptor activation reduces GABAA receptor currents in neostriatal neurons through a PKA/DARPP-32/PP1 signaling cascade. J Neurophysiol 2000; 83: 2996–3004
  • Nally RE, McNamara FN, Clifford JJ, Kinsella A, Tighe O, Croke DT, Fienberg AA, Greengard P, Waddington JL. Topographical assessment of ethological and dopamine receptor agonist-induced behavioral phenotype in mutants with congenic DARPP-32 ‘knockout.’. Neuropsychopharmacology 2003; 28: 2055–2063
  • Cantrell AR, Smith RD, Goldin AL, Scheuer T, Catterall WA. Dopaminergic modulation of sodium current in hippocampal neurons via cAMP-dependent phosphorylation of specific sites in the sodium channel α subunit. J Neurosci 1997; 17: 7330–7338
  • Surmeier DJ, Eberwine J, Wilson CJ, Cao Y, Stefani A, Kitai ST. Dopamine receptor subtypes colocalize in rat striatonigral neurons. Proc Natl Acad Sci USA 1992; 89: 10178–10182
  • Li M, West JW, Lai Y, Scheuer T, Catterall WA. Functional modulation of brain sodium channels by cAMP-dependent phosphorylation. Neuron 1992; 8: 1151–1159
  • Smith RD, Goldin AL. Phosphorylation at a single site in the rat brain sodium channel is necessary and sufficient for current reduction by protein kinase A. J Neurosci 1997; 17: 6086–6093
  • Murphy BJ, Rossie S, De Jongh KS, Catterall WA. Identification of the sites of selective phosphorylation and dephosphorylation of the rat brain Na+ channel α subunit by cAMP-dependent protein kinase and phosphoprotein phosphatases. J Biol Chem 1993; 268: 27355–27362
  • Liu FC, Graybiel AM. Spatiotemporal dynamics of CREB phosphorylation: transient versus sustained phosphorylation in the developing striatum. Neuron 1996; 17: 1133–1144
  • Zhang DS, Zhang L, Lou DW, Nakabeppu Y, Zhang JH, Xu M. The dopamine D1 receptor is a critical mediator for cocaine-induced gene expression. J Neurochem 2002; 82: 1453–1464
  • Konradi C, Cole RL, Heckers S, Hyman SE. Amphetamine regulates gene expression in rat striatum via transcription factor CREB. J Neurosci 1994; 14: 5623–5634
  • Cole RL, Konradi C, Douglass J, Hyman SE. Neuronal adaptation to amphetamine and dopamine: Molecular mechanisms of prodynorphin gene regulation in rat striatum. Neuron 1995; 14: 813–823
  • Brami-Cherrier K, Valjent E, Garcia M, Pagès C, Hipskind RA, Caboche J. Dopamine induces a PI3-kinase-independent activation of Akt in striatal neurons: A new route to cAMP response element-binding protein phosphorylation. J Neurosci 2002; 22: 8911–8921
  • Berke JD, Hyman SE. Addiction, dopamine, and the molecular mechanisms of memory. Neuron 2000; 25: 515–532
  • Mahan LC, Burch RM, Monsma FJ, Jr, Sibley DR. Expression of striatal D1 dopamine receptors coupled to inositol phosphate production and Ca2+ mobilization in Xenopus oocytes. Proc Natl Acad Sci USA 1990; 87: 2196–2200
  • Lezcano N, Mrzljak L, Eubanks S, Levenson R, Goldman-Rakic P, Bergson C. Dual signaling regulated by calcyon, a D1 dopamine receptor interacting protein. Science 2000; 287: 1660–1664
  • Lezcano N, Bergson C. D1/D5 dopamine receptors stimulate intracellular calcium release in primary cultures of neocortical and hippocampal neurons. J Neurophysiol 2002; 87: 2167–2175
  • Undie AS, Friedman E. Stimulation of a dopamine D1 receptor enhances inositol phosphates formation in rat brain. J Pharmacol Exp Ther 1990; 253: 987–992
  • Pacheco MA, Jope RS. Comparison of [3H]phosphatidylinositol and [3H]phosphatidylinositol 4,5-bisphosphate hydrolysis in postmortem human brain membranes and characterization of stimulation by dopamine D1 receptors. J Neurochem 1997; 69: 639–644
  • Undie AS, Weinstock J, Sarau HM, Friedman E. Evidence for a distinct D1-like dopamine receptor that couples to activation of phosphoinositide metabolism in brain. J Neurochem 1994; 62: 2045–2048
  • Jin LQ, Goswami S, Cai GP, Zhen XC, Friedman E. SKF83959 selectively regulates phosphatidylinositol-linked D1 dopamine receptors in rat brain. J Neurochem 2003; 85: 378–386
  • Leonard SK, Anderson CM, Lachowicz JE, Schulz DW, Kilts CD, Mailman RB. Amygdaloid D1 receptors are not linked to stimulation of adenylate cyclase. Synapse 2003; 50: 320–333
  • Wang HY, Undie AS, Friedman E. Evidence for the coupling of Gq protein to D1 -like dopamine sites in rat striatum: possible role in dopamine-mediated inositol phosphate formation. Mol Pharmacol 1995; 48: 988–994
  • Friedman E, Jin LQ, Cai GP, Hollon TR, Drago J, Sibley DR, Wang HY. D1-like dopaminergic activation of phosphoinositide hydrolysis is independent of D1A dopamine receptors: evidence from D1A knockout mice. Mol Pharmacol 1997; 51: 6–11
  • Clifford JJ, Tighe O, Croke DT, Kinsella A, Sibley DR, Drago J, Waddington JL. Conservation of behavioural topography to dopamine D1-like receptor agonists in mutant mice lacking the D1A receptor implicates a D1-like receptor not coupled to adenylyl cyclase. Neuroscience 1999; 93: 1483–1489
  • McNamara FN, Clifford JJ, Tighe O, Kinsella A, Drago J, Croke DT, Waddington JL. Congenic D1A dopamine receptor mutants: ethologically based resolution of behavioural topography indicates genetic background as a determinant of knockout phenotype. Neuropsychopharmacology 2003; 28: 86–99
  • Montague DM, Striplin CD, Overcash JS, Drago J, Lawler CP, Mailman RB. Quantification of D1B (D5) receptors in dopamine D1A receptor-deficient mice. Synapse 2001; 39: 319–322
  • Foehring RC, Surmeier DJ. Voltage-gated potassium currents in acutely dissociated rat cortical neurons. J Neurophysiol 1993; 70: 51–63
  • Hopf FW, Cascini MG, Gordon AS, Diamond I, Bonci A. Cooperative activation of dopamine D1 and D2 receptors increases spike firing of nucleus accumbens neurons via G-protein β, γ subunits. J Neurosci 2003; 23: 5079–5087
  • Sunahara RK, Dessauer CW, Gilman AG. Complexity and diversity of mammalian adenylyl cyclases. Annu Rev Pharmacol Toxicol 1996; 36: 461–480
  • Watts VJ, Neve KA. Activation of type II adenylate cyclase by D2 and D4 but not D3 dopamine receptors. Mol Pharmacol 1997; 52: 181–186
  • Marshall JF, Ruskin DN, Lahoste GJ. D1/D2 dopamine receptor interactions in basal ganglia. The Dopamine Receptors, KA Neve, RL Neve. Humana Press, Totawa, NJ 1997; 193–219
  • Gorelova N, Seamans JK, Yang CR. Mechanisms of dopamine activation of fast-spiking interneurons that exert inhibition in rat prefrontal cortex. J Neurophysiol 2002; 88: 3150–3166
  • Yang CR, Seamans JK. Dopamine D1 receptor actions in layers V–VI rat prefrontal cortex neurons in vitro: modulation of dendritic-somatic signal integration. J Neurosci 1996; 16: 1922–1935
  • Dong Y, White FJ. Dopamine D1-class receptors selectively modulate a slowly inactivating potassium current in rat medial prefrontal cortex pyramidal neurons. J Neurosci 2003; 23: 2686–2695
  • Yang CR, Seamans JK, Gorelova N. Developing a neuronal model for the pathophysiology of schizophrenia based on the nature of electrophysiological actions of dopamine in the prefrontal cortex. Neuropsychopharmacology 1999; 21: 161–194
  • Dong Y, Cooper D, Nasif F, Hu XT, White FJ. Dopamine modulates inwardly rectifying potassium currents in medial prefrontal cortex pyramidal neurons. J Neurosci 2004; 24: 3077–3085
  • Paupardin-Tritsch D, Colombaioni L, Deterre P, Gerschenfeld HM. Two different mechanisms of calcium spike modulation by dopamine. J Neurosci 1985; 5: 2522–2532
  • Marchetti C, Carbone E, Lux HD. Effects of dopamine and noradrenaline on Ca channels of cultured sensory and sympathetic neurons of chick. Pflugers Arch 1986; 406: 104–111
  • Surmeier DJ, Bargas J, Hemmings HC Jr, Nairn AC, Greengard P. Modulation of calcium currents by a D1 dopaminergic protein kinase/phosphatase cascade in rat neostriatal neurons. Neuron 1995; 14: 385–397
  • Hernández-López S, Bargas J, Surmeier DJ, Reyes A, Galarraga E. D1 receptor activation enhances evoked discharge in neostriatal medium spiny neurons by modulating an L-type Ca2+ conductance. J Neurosci 1997; 17: 3334–3342
  • Schiller J, Schiller Y, Stuart G, Sakmann B. Calcium action potentials restricted to distal apical dendrites of rat neocortical pyramidal neurons. J Physiol 1997; 505: 605–616
  • Seamans JK, Gorelova NA, Yang CR. Contributions of voltage-gated Ca2+ channels in the proximal versus distal dendrites to synaptic integration in prefrontal cortical neurons. J Neurosci 1997; 17: 5936–5948
  • Gross RA, Uhler MD, Macdonald RL. The cyclic AMP-dependent protein kinase catalytic subunit selectively enhances calcium currents in rat nodose neurones. J Physiol 1990; 429: 483–496
  • Lai Y, Peterson BZ, Catterall WA. Selective dephosphorylation of the subunits of skeletal muscle calcium channels by purified phosphoprotein phosphatases. J Neurochem 1993; 61: 1333–1339
  • Young CE, Yang CR. Dopamine D1/D5 receptor modulates state-dependent switching of soma-dendritic Ca2+ potentials via differential protein kinase A and C activation in rat prefrontal cortical neurons. J Neurosci 2004; 24: 8–23
  • Colbert CM, Johnston D. Axonal action-potential initiation and Na+ channel densities in the soma and axon initial segment of subicular pyramidal neurons. J Neurosci 1996; 16: 6676–6686
  • Stuart G, Sakmann B. Amplification of EPSPs by axosomatic sodium channels in neocortical pyramidal neurons. Neuron 1995; 15: 1065–1076
  • Hutcheon B, Miura RM, Puil E. Models of subthreshold membrane resonance in neocortical neurons. J Neurophysiol 1996; 76: 698–714
  • Calabresi P, Mercuri N, Stanzione P, Stefani A, Bernardi G. Intracellular studies on the dopamine-induced firing inhibition of neostriatal neurons in vitro: evidence for D1 receptor involvement. Neuroscience 1987; 20: 757–771
  • Schiffmann SN, Lledo P-M, Vincent J-D. Dopamine D1 receptor modulates the voltage-gated sodium current in rat striatal neurones through a protein kinase A. J Physiol (Lond) 1995; 483: 95–107
  • Schiffmann SN, Desdouits F, Menu R, Greengard P, Vincent JD, Vanderhaeghen JJ, Girault JA. Modulation of the voltage-gated sodium current in rat striatal neurons by DARPP-32, an inhibitor of protein phosphatase. Eur J Neurosci 1998; 10: 1312–1320
  • Moorman JR, Kirsch GE, VanDongen AM, Joho RH, Brown AM. Fast and slow gating of sodium channels encoded by a single mRNA. Neuron 1990; 4: 243–252
  • Alzheimer C, Schwindt PC, Crill WE. Modal gating of Na+ channels as a mechanism of persistent Na+ current in pyramidal neurons from rat and cat sensorimotor cortex. J Neurosci 1993; 13: 660–673
  • Brown AM, Schwindt PC, Crill WE. Different voltage dependence of transient and persistent Na+ currents is compatible with modal-gating hypothesis for sodium channels. J Neurophysiol 1994; 71: 2562–2565
  • Connors BW, Gutnick MJ, Prince DA. Electrophysiological properties of neocortical neurons in vitro. J Neurophysiol 1982; 48: 1302–1320
  • Crill WE. Persistent sodium current in mammalian central neurons. Annu Rev Physiol 1996; 58: 349–362
  • Maurice N, Tkatch T, Meisler M, Sprunger LK, Surmeier DJ. D1/D5 dopamine receptor activation differentially modulates rapidly inactivating and persistent sodium currents in prefrontal cortex pyramidal neurons. J Neurosci 2001; 21: 2268–2277
  • Gorelova NA, Yang CR. Dopamine D1/D5 receptor activation modulates a persistent sodium current in rat prefrontal cortical neurons in vitro. J Neurophysiol 2000; 84: 75–87
  • Geijo-Barrientos E, Pastore C. The effects of dopamine on the subthreshold electrophysiological responses of rat prefrontal cortex neurons in vitro. Eur J Neurosci 1995; 7: 358–366
  • Cepeda C, Radisavljevic Z, Peacock W, Levine MS, Buchwald NA. Differential modulation by dopamine of responses evoked by excitatory amino acids in human cortex. Synapse 1992; 11: 330–341
  • Levine MS, Altemus KL, Cepeda C, Cromwell HC, Crawford C, Ariano MA, Drago J, Sibley DR, Westphal H. Modulatory actions of dopamine on NMDA receptor-mediated responses are reduced in D1A-deficient mutant mice. J Neurosci 1996; 16: 5870–5882
  • Cepeda C, Levine MS. Dopamine and N-methyl-D-aspartate receptor interactions in the neostriatum. Dev Neurosci 1998; 20: 1–18
  • Flores-Hernández J, Cepeda C, Hernández-Echeagaray E, Calvert CR, Jokel ES, Fienberg AA, Greengard P, Levine MS. Dopamine enhancement of NMDA currents in dissociated medium-sized striatal neurons: Role of D1 receptors and DARPP-32. J Neurophysiol 2002; 88: 3010–3020
  • Huang Y-Y, Kandel ER. D1/D5 receptor agonists induce a protein synthesis-dependent late potentiation in the CA1 region of the hippocampus. Proc Natl Acad Sci USA 1995; 92: 2446–2450
  • Zheng P, Zhang XX, Bunney BS, Shi WX. Opposite modulation of cortical N-methyl-D-aspartate receptor-mediated responses by low and high concentrations of dopamine. Neuroscience 1999; 91: 527–535
  • Seamans JK, Durstewitz D, Christie BR, Stevens CF, Sejnowski TJ. Dopamine D1/D5 receptor modulation of excitatory synaptic inputs to layer V prefrontal cortex neurons. Proc Natl Acad Sci USA 2001; 98: 301–306
  • Wang J, O’Donnell P. D1 dopamine receptors potentiate NMDA-mediated excitability increase in layer V prefrontal cortical pyramidal neurons. Cereb Cortex 2001; 11: 452–462
  • Tseng KY, O’Donnell P. Dopamine-glutamate interactions controlling prefrontal cortical pyramidal cell excitability involve multiple signaling mechanisms. J Neurosci 2004; 24: 5131–5139
  • Snyder GL, Fienberg AA, Huganir RL, Greengard P. A dopamine/D1 receptor/protein kinase A/dopamine- and cAMP-regulated phosphoprotein (Mr 32 kDa)/protein phosphatase-1 pathway regulates dephosphorylation of the NMDA receptor. J Neurosci 1998; 18: 10297–10303
  • Cepeda C, Colwell CS, Itri JN, Chandler SH, Levine MS. Dopaminergic modulation of NMDA-induced whole cell currents in neostriatal neurons in slices: contribution of calcium conductances. J Neurophysiol 1998; 79: 82–94
  • Chergui K, Lacey MG. Modulation by dopamine D1-like receptors of synaptic transmission and NMDA receptors in rat nucleus accumbens is attenuated by the protein kinase C inhibitor Ro 32-0432. Neuropharmacology 1999; 38: 223–231
  • Colwell CS, Levine MS. Excitatory synaptic transmission in neostriatal neurons: regulation by cyclic AMP-dependent mechanisms. J Neurosci 1995; 15: 1704–1713
  • Durstewitz D, Seamans JK, Sejnowski TJ. Dopamine-mediated stabilization of delay-period activity in a network model of prefrontal cortex. J Neurophysiol 2000; 83: 1733–1750
  • Lavin A, Grace AA. Stimulation of D1-type dopamine receptors enhances excitability in prefrontal cortical pyramidal neurons in a state-dependent manner. Neuroscience 2001; 104: 335–346
  • Cepeda C, Buchwald NA, Levine MS. Neuromodulatory actions of dopamine in the neostriatum are dependent upon the excitatory amino acid receptor subtypes activated. Proc Natl Acad Sci USA 1993; 90: 9576–9580
  • Galarraga E, Hernández-López S, Reyes A, Barral J, Bargas J. Dopamine facilitates striatal EPSPs through an L-type Ca2+ conductance. Neuroreport 1997; 8: 2183–2186
  • Yan Z, Hsieh-Wilson L, Feng J, Tomizawa K, Allen PB, Fienberg AA, Nairn AC, Greengard P. Protein phosphatase 1 modulation of neostriatal AMPA channels: regulation by DARPP-32 and spinophilin. Nat Neurosci 1999; 2: 13–17
  • Snyder GL, Allen PB, Fienberg AA, Valle CG, Huganir RL, Nairn AC, Greengard P. Regulation of phosphorylation of the GluR1 AMPA receptor in the neostriatum by dopamine and psychostimulants in vivo. J Neurosci 2000; 20: 4480–4488
  • Chao SZ, Lu WX, Lee HK, Huganir RL, Wolf ME. D1 dopamine receptor stimulation increases GluR1 phosphorylation in postnatal nucleus accumbens cultures. J Neurochem 2002; 81: 984–992
  • Li AJ, Suzuki M, Suzuki S, Ikemoto M, Imamura T. Differential phosphorylation at serine sites in glutamate receptor-1 within neonatal rat hippocampus. Neurosci Lett 2003; 341: 41–44
  • Gonzalez-Islas C, Hablitz JJ. Dopamine enhances EPSCs in layer II–III pyramidal neurons in rat prefrontal cortex. J Neurosci 2003; 23: 867–875
  • Dunah AW, Standaert DG. Dopamine D1 receptor-dependent trafficking of striatal NMDA glutamate receptors to the postsynaptic membrane. J Neurosci 2001; 21: 5546–5558
  • Chao SZ, Ariano MA, Peterson DA, Wolf ME. D1 dopamine receptor stimulation increases GluR1 surface expression in nucleus accumbens neurons. J Neurochem 2002; 83: 704–712
  • Dunah AW, Sirianni AC, Fienberg AA, Bastia E, Schwarzschild MA, Standaert DG. Dopamine D1-dependent trafficking of striatal N-methyl-D-aspartate glutamate receptors requires Fyn protein tyrosine kinase but not DARPP-32. Mol Pharmacol 2004; 65: 121–129
  • Lee FJ, Xue S, Pei L, Vukusic B, Chéry N, Wang Y, Wang YT, Niznik HB, Yu X, Liu F. Dual regulation of NMDA receptor functions by direct protein–protein interactions with the dopamine D1 receptor. Cell 2002; 111: 219–230
  • Fiorentini C, Gardoni F, Spano PF, 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
  • Pei L, Lee FJS, Moszczynsak A, Vukusic B, Liu F. Regulation of dopamine D1 receptor function by physical interaction with the NMDA receptors. J Neurosci 2004; 24: 1149–1158
  • Nicola SM, Malenka RC. Modulation of synaptic transmission by dopamine and norepinephrine in ventral but not dorsal striatum. J Neurophysiol 1998; 79: 1768–1776
  • Pisani A, Bonsi P, Centonze D, Calabresi P, Bernardi G. Activation of D2-like dopamine receptors reduces synaptic inputs to striatal cholinergic interneurons. J Neurosci 2000; 20: RC69
  • Yan Z, Surmeier DJ. D5 dopamine receptors enhance Zn2+-sensitive GABAA currents in striatal cholinergic interneurons through a PKA/PP1 cascade. Neuron 1997; 19: 1115–1126
  • Liu F, Wan Q, Pristupa ZB, Yu XM, Wang YT, Niznik HB. Direct protein-protein coupling enables cross-talk between dopamine D5 and γ-aminobutyric acid A receptors. Nature 2000; 403: 274–280
  • Jose PA, Eisner GM, Felder RA. Renal dopamine receptors in health and hypertension. Pharmacol Ther 1998; 80: 149–182
  • Aizman O, Brismar H, Greengard P, Zettergren E, Levey AI, Forssberg H, Greengard P, Aperia A. Anatomical and physiological evidence for D1 and D2 dopamine receptor colocalization in neowstriatal neurons. Nat Neurosci 2000; 3: 226–230
  • Aperia A, Fryckstedt J, Svensson L, Hemmings HC Jr, Nairn AC, Greengard P. Phosphorylated Mr 32,000 dopamine- and cAMP-regulated phosphoprotein inhibits Na+,K+-ATPase activity in renal tubule cells. Proc Natl Acad Sci USA 1991; 88: 2798–2801
  • Shahedi M, Laborde K, Azimi S, Hamdani S, Sachs C. Mechanisms of dopamine effects on Na-K-ATPase activity in Madin-Darby canine kidney (MDCK) epithelial cells. Pflügers Arch 1995; 429: 832–840
  • Gomes P, Soares-da-Silva P. Role of cAMP-PKA-PLC signaling cascade on dopamine-induced PKC-mediated inhibition of renal Na+-K+-ATPase activity. Am J Physiol Renal Physiol 2002; 282: F1084–F1096
  • Gerfen CR, Miyachi S, Paletzki R, Brown P. D1 dopamine receptor supersensitivity in the dopamine-depleted striatum results from a switch in the regulation of ERK1/2/MAP kinase. J Neurosci 2002; 22: 5042–5054
  • Zhen XC, Uryu K, Wang HY, Friedman E. D1 dopamine receptor agonists mediate activation of p38 mitogen-activated protein kinase and c-jun amino-terminal kinase by a protein kinase A dependent mechanism in SK-N-MC human neuroblastoma cells. Mol Pharmacol 1998; 54: 453–458
  • Weissman JT, Ma JN, Essex A, Gao Y, Burstein ES. G-protein-coupled receptor-mediated activation of rap GTPases: characterization of a novel Gαi regulated pathway. Oncogene 2004; 23: 241–249
  • de Rooij J, Zwartkruis FJ, Verheijen MH, Cool RH, Nijman SM, Wittinghofer A, Bos JL. Epac is a Rap1 guanine-nucleotide-exchange factor directly activated by cyclic AMP. Nature 1998; 396: 474–477
  • Kurose H, Katada T, Amano T, Ui M. Specific uncoupling by islet-activating protein, pertussis toxin, of negative signal transduction via α-adrenergic, cholinergic, and opiate receptors in neuroblastoma x glioma hybrid cells. J Biol Chem 1983; 258: 4870–4875
  • Bokoch GM, Katada T, Northup JK, Hewlett EL, Gilman AG. Identification of the predominant substrate for ADP-ribosylation by islet activating protein. J Biol Chem 1983; 258: 2072–2075
  • Robinson SW, Caron MG. Interactions of dopamine receptors with G proteins. The Dopamine Receptors, KA Neve, RL Neve. Humana Press, Totawa, NJ 1997; 137–165
  • Neve KA, DuRand CJ, Teeter MM. Structural analysis of the mammalian D2, D3, and D4 dopamine receptors. Dopamine Receptors and Transporters: Function, Imaging, and Clinical Implication, A Sidhu, M Laruelle, P Vernier. Marcel Dekker Inc., New York 2003; 77–144
  • Lledo PM, Homburger V, Bockaert J, Vincent J-D. Differential G protein-mediated coupling of D2 dopamine receptors to K+ and Ca2+ currents in rat anterior pituitary cells. Neuron 1992; 8: 455–463
  • Liu YF, Jakobs KH, Rasenick MM, Albert PR. G protein specificity in receptor-effector coupling: analysis of the roles of Go and Gi2 in GH4C1 pituitary cells. J Biol Chem 1994; 269: 13880–13886
  • Wong YH, Conklin BR, Bourne HR. Gz-Mediated hormonal inhibition of cyclic AMP accumulation. Science 1992; 255: 339–342
  • Obadiah J, Avidor-Reiss T, Fishburn CS, Carmon S, Bayewitch M, Vogel Z, Fuchs S, Levavi-Sivan B. Adenylyl cyclase interaction with the D2 dopamine receptor family: differential coupling to Gi, Gz, and Gs. Cell Mol Neurobiol 1999; 19: 653–664
  • Ganz MB, Pachter JA, Barber DL. Multiple receptors coupled to adenylate cyclase regulate Na–H exchange independent of cAMP. J Biol Chem 1990; 265: 8989–8992
  • Neve KA, Kozlowski MR, Rosser MP. Dopamine D2 receptor stimulation of Na+/H+ exchange assessed by quantification of extracellular acidification. J Biol Chem 1992; 267: 25748–25753
  • Senogles SE. The D2 dopamine receptor mediates inhibition of growth in GH4ZR7 cells: involvement of protein kinase-Cε. Endocrinology 1994; 134: 783–789
  • Watts VJ, Wiens BL, Cumbay MG, Vu MN, Neve RL, Neve KA. Selective activation of Gαo by D2L dopamine receptors in NS20Y neuroblastoma cells. J Neurosci 1998; 18: 8692–8699
  • Cordeaux Y, Nickolls SA, Flood LA, Graber SG, Strange PG. Agonist regulation of D2 dopamine receptor/G protein interaction—evidence for agonist selection of G protein subtype. J Biol Chem 2001; 276: 28667–28675
  • Gazi L, Nickolls SA, Strange PG. Functional coupling of the human dopamine D2 receptor with Gαi1, Gαi2, Gαi3 and Gαo G proteins: evidence for agonist regulation of G protein selectivity. Br J Pharmacol 2003; 138: 775–786
  • Leaney JL, Tinker A. The role of members of the pertussis toxin-sensitive family of G proteins in coupling receptors to the activation of the G protein-gated inwardly rectifying potassium channel. Proc Natl Acad Sci USA 2000; 97: 5651–5656
  • Liu JC, Baker RE, Sun C, Sundmark VC, Elsholtz HP. Activation of Go-coupled dopamine D2 receptors inhibits ERK1/ERK2 in pituitary cells—a key step in the transcriptional suppression of the prolactin gene. J Biol Chem 2002; 277: 35819–35825
  • Nickolls SA, Strange PG. Interaction of the D2short dopamine receptor with G proteins: analysis of receptor G protein selectivity. Biochem Pharmacol 2003; 65: 1139–1150
  • Jiang MS, Spicher K, Boulay G, Wang Y, Birnbaumer L. Most central nervous system D2 dopamine receptors are coupled to their effecters by Go. Proc Natl Acad Sci USA 2001; 98: 3577–3582
  • Liu LX, Burgess LH, Gonzalez AM, Sibley DR, Chiodo LA. D2S, D2L, D3, and D4 dopamine receptors couple to a voltage-dependent potassium current in N18TG2 x mesencephalon hybrid cell (MES-23.5) via distinct G proteins. Synapse 1999; 31: 108–118
  • Kazmi MA, Snyder LA, Cypess AM, Graber SG, Sakmar TP. Selective reconstitution of human D4 dopamine receptor variants with Giα subtypes. Biochemistry 2000; 39: 3734–3744
  • Yamaguchi I, Harmon SK, Todd RD, O’Malley KL. The rat D4 dopamine receptor couples to cone transducin (Gαt2) to inhibit forskolin-stimulated cAMP accumulation. J Biol Chem 1997; 272: 16599–16602
  • Newman-Tancredi A, Cussac D, Audinot V, Pasteau V, Gavaudan S, Millan MJ. G protein activation by human dopamine D3 receptors in high-expressing Chinese hamster ovary cells: a guanosine-5′-O-(3-[35S]thio)-triphosphate binding and antibody study. Mol Pharmacol 1999; 55: 564–574
  • Zaworski PG, Alberts GL, Pregenzer JF, Bin Im W, Slightom JL, Gill GS. Efficient functional coupling of the human D3 dopamine receptor to Go subtype of G proteins in SH-SY5Y cells. Br J Pharmacol 1999; 128: 1181–1188
  • Lavine N, Ethier N, Oak JN, Pei L, Liu F, Trieu P, Rebois RV, Bouvier M, Hébert TE, Van Tol HHM. G protein-coupled receptors form stable complexes with inwardly rectifying potassium channels and adenylyl cyclase. J Biol Chem 2002; 277: 46010–46019
  • Vanhauwe J.F.M., Josson K, Luyten WHML, Driessen AJ, Leysen JE. G-protein sensitivity of ligand binding to human dopamine D2 and D3 receptors expressed in Escherichia coli: clues for a constrained D3 receptor structure. J Pharmacol Exp Ther 2000; 295: 274–283
  • De Camilli P, Macconi D, Spada A. Dopamine inhibits adenylate cyclase in human prolactin-secreting pituitary adenomas. Nature 1979; 278: 252–254
  • Stoof JC, Kebabian JW. Opposing roles for D-1 and D-2 dopamine receptors in efflux of cyclic AMP from rat neostriatum. Nature 1981; 294: 366–368
  • Kelly MA, Rubinstein M, Phillips TJ, Lessov CN, Burkhart-Kasch S, Zhang G, Bunzow JR, Fang Y, Gerhardt GA, Grandy DK, Low MJ. Locomotor activity in D2 dopamine receptor-deficient mice is determined by gene dosage, genetic background, and developmental adaptations. J Neurosci 1998; 18: 3470–3479
  • Oak JN, Oldenhof J, Van Tol HHM. The dopamine D4 receptor: one decade of research. Eur J Pharmacol 2000; 405: 303–327
  • Robinson SW, Caron MG. Selective inhibition of adenylyl cyclase type V by dopamine D3 receptor. Mol Pharmacol 1997; 52: 508–514
  • Scarselli M, Novi F, Schallmach E, Ling RW, Baragli A, Colzi A, Griffon N, Corsini GU, Sokoloff P, Levenson R, Vogel Z, Maggio R. D2/D3 dopamine receptor heterodimers exhibit unique functional properties. J Biol Chem 2001; 276: 30308–30314
  • Maggio R, Scarselli M, Novi F, Millan MJ, Corsini GU. Potent activation of dopamine D3/D2 heterodimers by the antiparkinsonian agents, S32504, pramipexole and ropinirole. J Neurochem 2003; 87: 631–641
  • Taussig R, Tang W-J, Hepler JR, Gilman AG. Distinct patterns of bidirectional regulation of mammalian adenylyl cyclases. J Biol Chem 1994; 269: 6093–6100
  • Dessauer CW, Chen-Goodspeed M, Chen J. Mechanism of Gαi-mediated inhibition of type V adenylyl cyclase. J Biol Chem 2002; 277: 28823–28829
  • Nishi A, Snyder GL, Greengard P. Bidirectional regulation of DARPP-32 phosphorylation by dopamine. J Neurosci 1997; 17: 8147–8155
  • Nishi A, Snyder GL, Fienberg AA, Fisone G, Aperia A, Naim AC, Greengard P. Requirement for DARPP-32 in mediating effect of dopamine D2 receptor activation. Eur. J Neurosci 1999; 11: 2589–2592
  • Lew JY, Garcia-Espana A, Lee KY, Carr KD, Goldstein M, Haycock JW, Meller E. Increased site-specific phosphorylation of tyrosine hydroxylase accompanies stimulation of enzymatic activity induced by cessation of dopamine neuronal activity. Mol Pharmacol 1999; 55: 202–209
  • Shin R-M, Masuda M, Miura M, Sano H, Shirasawa T, Song W-J, Kobayashi K, Aosaki T. Dopamine D4 receptor-Induced postsynaptic inhibition of GABAergic currents in mouse globus pallidus neurons. J Neurosci 2003; 23: 11662–11672
  • Wang X, Zhong P, Gu Z, Yan Z. Regulation of NMDA receptors by dopamine D4 signaling in prefrontal cortex. J Neurosci 2003; 23: 9852–9861
  • Kotecha SA, Oak JN, Jackson MF, Perez Y, Orser BA, Van Tol HHM, MacDonald JF. A D2 class dopamine receptor transactivates a receptor tyrosine kinase to inhibit NMDA receptor transmission. Neuron 2002; 35: 1111–1122
  • Chen J, Devivo M, Dingus J, Harry A, Li J, Sui J, Carty DJ, Blank JL, Exton JH, Stoffel RH, Inglese J, Lefkowitz RJ, Logothetis DE, Hildebrandt JD, Iyengar R. A region of adenylyl cyclase 2 critical for regulation by G protein β, γ subunits. Science 1995; 268: 1166–1169
  • Weitmann S, Schultz G, Kleuss C. Adenylyl cyclase type II domains involved in Gβ, γ stimulation. Biochemistry 2001; 40: 10853–10858
  • Lacey MG, Mercuri NB, North RA. Dopamine acts on D2 receptors to increase potassium conductance in neurones of rat substantia nigra zona compacta. J Physiol (Lond) 1987; 392: 397–416
  • Liu L, Shen R-Y, Kapatos G, Chiodo LA. Dopamine neuron membrane physiology: characterization of the transient outward current (IA) and demonstration of a common signal transduction pathway for IA and IK. Synapse 1994; 17: 230–240
  • Greif GJ, Lin Y-J, Liu J-C, Freedman JE. Dopamine-modulated potassium channels on rat striatal neurons: Specific activation and cellular expression. J Neurosci 1995; 15: 4533–4544
  • Wickman KD, Iñiguez-Lluhi JA, Davenport PA, Taussig R, Krapivinsky GB, Linder ME, Gilman AG, Clapham DE. Recombinant G-protein β, γ-subunits activate the muscarinic-gated atrial potassium channel. Nature 1994; 368: 255–257
  • Brown NA, McAllister G, Weinberg D, Milligan G, Seabrook GR. Involvement of G-protein αi1 subunits in activation of G-protein gated inward rectifying K+ channels (GIRK1) by human NPY1 receptors. Br J Pharmacol 1995; 116: 2346–2348
  • Dascal N. Signalling via the G protein-activated K+ channels. Cell Signal 1997; 9: 551–573
  • Kuzhikandathil EV, Yu WF, 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
  • Karschin C, Dißmann E, Stühmer W, Karschin A. IRK(1–3) and GIRK(1–4) inwardly rectifying K+ channel mRNAs are differentially expressed in the adult rat brain. J Neurosci 1996; 16: 3559
  • Inanobe A, Yoshimoto Y, Horio Y, Morishige K-I, Hibino H, Matsumoto S, Tokunaga Y, Maeda T, Hata Y, Takai Y, Kurachi Y. Characterization of G-protein-gated K+ channels composed of Kir3.2 subunits in dopaminergic neurons of the substantia nigra. J Neurosci 1999; 19: 1006–1017
  • Kuzhikandathil EV, Oxford GS. Dominant-negative mutants identify a role for GIRK channels in D3 dopamine receptor-mediated regulation of spontaneous secretory activity. J Gen Physiol 2000; 115: 697–706
  • Rebois RV, Hebert TE. Protein complexes involved in heptahelical receptor-mediated signal transduction. Receptors Channels 2003; 9: 169–194
  • Cass WA, Zahniser NR. Potassium channel blockers inhibit D2 dopamine, but not A1 adenosine, receptor-mediated inhibition of striatal dopamine release. J Neurochem 1991; 57: 147–152
  • Memo M, Missale C, Carruba MO, Spano PF. D2 dopamine receptors associated with inhibition of dopamine release from rat neostriatum are independent of cyclic AMP. Neurosci Lett 1986; 71: 192–196
  • Davila V, Yan Z, Craciun LC, Logothetis D, Sulzer D. D3 dopamine autoreceptors do not activate G-protein-gated inwardly rectifying potassium channel currents in substantia nigra dopamine neurons. J Neurosci 2003; 23: 5693–5697
  • Blednov YA, Stoffel M, Cooper R, Wallace D, Mane N, Harris RA. Hyperactivity and dopamine D1 receptor activation in mice lacking girk2 channels. Psychopharmacology 2002; 159: 370–378
  • Surmeier DJ, Kitai ST. D1 and D2 dopamine receptor modulation of sodium and potassium currents in rat neostriatal neurons. Prog Brain Res 1993; 99(309–24)309–324
  • Seabrook GR, Kemp JA, Freedman SB, Patel S, Sinclair HA, McAllister G. Functional expression of human D3 dopamine receptors in differentiated neuroblastoma X glioma NG108–15 cells. Br J Pharmacol 1994; 111: 391–393
  • Seabrook GR, Knowles M, Brown N, Myers J, Sinclair H, Patel S, Freedman SB, McAllister G. Pharmacology of high-threshold calcium currents in GH4C1 pituitary cells and their regulation by activation of human D2 and D4 dopamine receptors. Br J Pharmacol 1994; 112: 728–734
  • Kuzhikandathil EV, Oxford GS. Activation of human D3 dopamine receptor inhibits P/Q-type calcium channels and secretory activity in AtT-20 cells. J Neurosci 1999; 19: 1698–1707
  • Okada Y, Miyamoto T, Toda K. Dopamine modulates a voltage-gated calcium channel in rat olfactory receptor neurons. Brain Res 2003; 968: 248–255
  • Yan Z, Song WJ, Surmeier DJ. 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
  • Zamponi GW, Snutch TP. Decay of prepulse facilitation of N type calcium channels during G protein inhibition is consistent with binding of a single Gβγ subunit. Proc Natl Acad Sci USA 1998; 95: 4035–4039
  • Page KM, Stephens GJ, Berrow NS, Dolphin AC. The intracellular loop between domains I and II of the B-type calcium channel confers aspects of G-protein sensitivity to the E-type calcium channel. J Neurosci 1997; 17: 1330–1338
  • Herlitze S, Hockerman GH, Scheuer T, Catterall WA. Molecular determinants of inactivation and G protein modulation in the intracellular loop connecting domains I and II of the calcium channel α1A subunit. Proc Natl Acad Sci USA 1997; 94: 1512–1516
  • Qin N, Platano D, Olcese R, Stefani E, Birnbaumer L. Direct interaction of Gβ, γ with a C-terminal Gβ, γ-binding domain of the Ca2+ channel α1 subunit is responsible for channel inhibition by G protein-coupled receptors. Proc Natl Acad Sci USA 1997; 94: 8866–8871
  • Hernández-López S, Tkatch T, Perez-Garci E, Galarraga E, Bargas J, Hamm H, Surmeier J. D2 dopamine receptors in striatal medium spiny neurons reduce L-type Ca2+ currents and excitability via a novel PLCβ1-IP3-calcineurin-signaling cascade. J Neurosci 2000; 20: 8987–8995
  • Dunlap K, Luebke JI, Turner TJ. Exocytotic Ca2+ channels in mammalian central neurons. Trends Neurosci 1995; 18: 89–98
  • Koga E, Momiyama T. Presynaptic dopamine D2-like receptors inhibit excitatory transmission onto rat ventral tegmental dopaminergic neurones. J Physiol 2000; 523: 163–173
  • Svensson E, Wikström MA, Hill RH, Grillner S. Endogenous and exogenous dopamine presynaptically inhibits glutamatergic reticulospinal transmission via an action of D2-receptors on N-type Ca2+ channels. Eur J Neurosci 2003; 17: 447–454
  • Gutkind JS. The pathways connecting G protein-coupled receptors to the nucleus through divergent mitogen-activated protein kinase cascades. J Biol Chem 1998; 273: 1839–1842
  • Alblas J, Van Corven EJ, Hordijk PL, Milligan G, Moolenaar WH. Gi-mediated activation of the p21ras-mitogen-activated protein kinase pathway by α2-adrenergic receptors expressed in fibroblasts. J Biol Chem 1993; 268: 22235–22238
  • Faure M, Voyno-Yasenetskaya TA, Bourne HR. cAMP and β, γ subunits of heterotrimeric G proteins stimulate the mitogen-activated protein kinase pathway in COS-7 cells. J Biol Chem 1994; 269: 7851–7854
  • Huff RM. Signal transduction pathways modulated by the D2 subfamily of dopamine receptors. Cell Signal 1996; 8: 453–459
  • Luo YQ, Kokkonen GC, Wang XT, Neve KA, Roth GS. D2 dopamine receptors stimulate mitogenesis through pertussis toxin-sensitive G proteins and ras-involved ERK and SAP/JNK pathways in rat C6-D2L glioma cells. J Neurochem 1998; 71: 980–990
  • Welsh GI, Hall DA, Warnes A, Strange PG, Proud CG. Activation of microtubule-associated protein kinase (Erk) and p70 S6 kinase by D2 dopamine receptors. J Neurochem 1998; 70: 2139–2146
  • Choi EY, Jeong DW, Park KW, Baik JH. G protein-mediated mitogen-activated protein kinase activation by two dopamine D2 receptors. Biochem Biophys Res Commun 1999; 256: 33–40
  • Ghahremani MH, Forget C, Albert PR. Distinct roles for Gαi2 and Gβ, γ in signaling to DNA synthesis and Gαi3 in cellular transformation by dopamine D2S receptor activation in BALB/c 3T3 cells. Mol Cell Biol 2000; 20: 1497–1506
  • Oak JN, Lavine N, Van Tol HHM. Dopamine D4 and D2L receptor stimulation of the mitogen-activated protein kinase pathway is dependent on transactivation of the platelet-derived growth factor receptor. Mol Pharmacol 2001; 60: 92–103
  • Kim SJ, Kim MY, Lee EJ, Ahn YS, Baik JH. Distinct regulation of internalization and mitogen-activated protein kinase activation by two isoforms of the dopamine D2 receptor. Mol Endocrinol 2004; 18: 640–652
  • Cussac D, Newman-Tancredi A, Pasteau V, Millan MJ. Human dopamine D3 receptors mediate mitogen-activated protein kinase activation via a phosphatidylinositol 3-kinase and an atypical protein kinase C-dependent mechanism. Mol Pharmacol 1999; 56: 1025–1030
  • Zhen XC, Zhang J, Johnson GP, Friedman E. D4 dopamine receptor differentially regulates Akt/nuclear factor-kappaB and extracellular signal-regulated kinase pathways in D4MN9D cells. Mol Pharmacol 2001; 60: 857–864
  • Yan Z, Feng J, Fienberg AA, Greengard P. D2 dopamine receptors induce mitogen-activated protein kinase and cAMP response element-binding protein phosphorylation in neurons. Proc Natl Acad Sci USA 1999; 96: 11607–11612
  • Cai GP, Zhen XC, Uryu K, Friedman E. Activation of extracellular signal-regulated protein kinases is associated with a sensitized locomotor response to D2 dopamine receptor stimulation in unilateral 6-hydroxydopamine-lesioned rats. J Neurosci 2000; 20: 1849–1857
  • Wang C, Buck DC, Yang R, Macey TA, Neve KA. Dopamine D2 receptor stimulation of mitogen-activated protein kinases mediated by cell type-dependent transactivation of receptor tyrosine kinases. J Biol Chem 2004, Submitted for publication
  • Daub H, Weiss FU, Wallasch C, Ullrich A. Role of transactivation of the EGF receptor in signalling by G-protein-coupled receptors. Nature 1996; 379: 557–560
  • Maudsley S, Pierce KL, Zamah AM, Miller WE, Ahn S, Daaka Y, Lefkowitz RJ, Luttrell LM. The β2-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
  • Wetzker R, Böhmer F-D. Transactivation joins multiple tracks to the ERK/MAPK cascade. Nat Rev Mol Cell Biol 2003; 4: 651–657
  • Nair VD, Sealfon SC. Agonist-specific transactivation of phosphoinositide 3-kinase signaling pathway mediated by the dopamine D2 receptor. J Biol Chem 2003; 278: 47053–47061
  • Lajiness ME, Chio CL, Huff RM. D2 dopamine receptor stimulation of mitogenesis in transfected Chinese hamster ovary cells: relationship to dopamine stimulation of tyrosine phosphorylations. J Pharmacol Exp Ther 1993; 267: 1573–1581
  • Hill CS, Treisman R. Transcriptional regulation by extracellular signals: mechanisms and specificity. Cell 1995; 80: 199–211
  • Fukunaga K, Miyamoto E. Role of MAP kinase in neurons. Mol Neurobiol 1998; 16: 79–95
  • Otani S, Auclair N, Desce JM, Roisin MP, Crépel F. Dopamine receptors and groups I and II mGluRs cooperate for long-term depression induction in rat prefrontal cortex through converging postsynaptic activation of MAP kinases. J Neurosci 1999; 19: 9788–9802
  • Impey S, Obrietan K, Storm DR. Making new connections: role of ERK/MAP kinase signaling in neuronal plasticity. Neuron 1999; 23: 11–14
  • Iaccarino C, Samad TA, Mathis C, Kercret H, Picetti R, Borrelli E. Control of lactotrop proliferation by dopamine: Essential role of signaling through D2 receptors and ERKs. Proc Natl Acad Sci USA 2002; 99: 14530–14535
  • Kanterman RY, Mahan LC, Briley EM, Monsma FJ, Sibley DR, Axelrod J, Felder CC. Transfected D2 dopamine receptors mediate the potentiation of arachidonic acid release in chinese hamster ovary cells. Mol Pharmacol 1991; 39: 364–369
  • Piomelli D, Pilon C, Giros B, Sokoloff P, Martres M-P, Schwartz J-C. Dopamine activation of the arachidonic acid cascade as a basis for D1/D2 receptor synergism. Nature 1991; 353: 164–167
  • Chio CL, Drong RF, Riley DT, Gill GS, Slightom JL, Huff RM. D4 dopamine receptor-mediated signaling events determined in transfected Chinese hamster ovary cells. J Biol Chem 1994; 269: 11813–11819
  • Vial D, Piomelli D. Dopamine D2 receptors potentiate arachidonate release via activation of cytosolic, arachidonate-specific phospholipase A2. J Neurochem 1995; 64: 2765–2772
  • Nilsson CL, Hellstrand M, Ekman A, Eriksson E. Both dopamine and the putative dopamine D3 receptor antagonist PNU-99194A induce a biphasic inhibition of phorbol ester-stimulated arachidonic acid release from CHO cells transfected with the dopamine D3 receptor. Life Sci 1999; 64: 939–951
  • Jelsema CL, Axelrod J. Stimulation of phospholipase A2 activity in bovine rod outer segments by the β, γ subunits of transducin and its inhibition by the α subunit. Proc Natl Acad Sci USA 1987; 84: 3623–3627
  • van Tol-Steye H, Lodder JC, Mansvelder HD, Planta RJ, Van Heerikhuizen H, Kits KS. Roles of G-protein β, γ, arachidonic acid, and phosphorylation in convergent activation of an S-like potassium conductance by dopamine, Ala-Pro-Gly-Trp-NH2, and Phe-Met-Arg-Phe-NH2. J Neurosci 1999; 19: 3739–3751
  • Nilsson CL, Hellstrand M, Ekman A, Eriksson E. Direct dopamine D2-receptor-mediated modulation of arachidonic acid release in transfected CHO cells without the concomitant administration of a Ca2+-mobilizing agent. Br J Pharmacol 1998; 124: 1651–1658
  • Piomelli D, Greengard P. Lipoxygenase metabolites of arachidonic acid in neuronal transmembrane signalling. Trends Pharmacol Sci 1990; 11: 367–373
  • Di Marzo V, Piomelli D. Participation of prostaglandin E2 in dopamine D2 receptor-dependent potentiation of arachidonic acid release. J Neurochem 1992; 59: 379–382
  • L’hirondel M, Chéramy A, Godeheu G, Glowinski J. Effects of arachidonic acid on dopamine synthesis, spontaneous release, and uptake in striatal synaptosomes from the rat. J Neurochem 1995; 64: 1406–1409
  • Zhang L, Reith MEA. Regulation of the functional activity of the human dopamine transporter by the arachidonic acid pathway. Eur J Pharmacol 1996; 315: 345–354
  • Mitchell R, McCulloch D, Lutz E, Johnson M, MacKenzie C, Fennell M, Fink G, Zhou W, Sealfon SC. Rhodopsin-family receptors associate with small G proteins to activate phospholipase D. Nature 1998; 392: 411–414
  • Senogles SE. The D2s dopamine receptor stimulates phospholipase D activity: a novel signaling pathway for dopamine. Mol Pharmacol 2000; 58: 455–462
  • Senogles SE. D2s dopamine receptor mediates phospholipase D and antiproliferation. Mol Cell Endocrinol 2003; 209: 61–69
  • Buhl AM, Johnson NL, Dhanasekaran N, Johnson GL. Gα12 and Gα13 stimulate Rho-dependent stress fiber formation and focal adhesion assembly. J Biol Chem 1995; 270: 24631–24634
  • Hart MJ, Jiang X, Kozasa T, Roscoe W, Singer WD, Gilman AG, Sternweis PC, Bollag G. Direct stimulation of the guanine nucleotide exchange activity of p115 RhoGEF by Gα13. Science 1998; 280: 2112–2114
  • Mao J, Yuan H, Xie W, Wu D. Guanine nucleotide exchange factor GEF115 specifically mediates activation of Rho and serum response factor by the G protein α subunit Gα 13. Proc Natl Acad Sci USA 1998; 95: 12973–12976
  • Gulledge AT, Jaffe DB. Multiple effects of dopamine on layer V pyramidal cell excitability in rat prefrontal cortex. J Neurophysiol 2001; 86: 586–595
  • Ma JY, Li M, Catterall WA, Scheuer T. Modulation of brain Na+ channels by a G-protein-coupled pathway. Proc Natl Acad Sci USA 1994; 91: 12351–12355
  • Yun CHC, Tse C-M, Nath SK, Levine SA, Brant SR, Donowitz M. Mammalian Na+/H+ exchanger gene family: structure and function studies. Am J Physiol Gastrointest. Liver Physiol 1995; 269: G1–G11
  • Cox BA, Rosser MP, Kozlowski MR, Duwe KM, Neve RL, Neve KA. Regulation and functional characterization of a rat recombinant dopamine D3 receptor. Synapse 1995; 21: 1–9
  • Chio CL, Lajiness ME, Huff RM. Activation of heterologously expressed D3 dopamine receptors: comparison with D2 dopamine receptors. Mol Pharmacol 1994; 45: 51–60
  • Hooley R, Yu CY, Symons M, Barber DL. G alpha 13 stimulates Na+–H+ exchange through distinct Cdc42-dependent and RhoA-dependent pathways. J Biol Chem 1996; 271: 6152–6158
  • Lin C-Y, Varma MG, Joubel A, Madabushi S, Lichtarge O, Barber DL. Conserved motifs in somatostatin D2-dopamine, and α2B-adrenergic receptors for inhibiting the Na-H exchanger, NHE1. J Biol Chem 2003; 278: 15128–15135
  • Felder CC, Albrecht FE, Campbell T, Eisner GM, Jose PA. cAMP-independent, G-protein-linked inhibition of Na+/H+ exchange in renal brush border by D1 dopamine agonists. Am J Physiol 1993; 264: F1032–F1037
  • Cepeda C, Hurst RS, Altemus KL, Flores-Hernández J, Calvert CR, Jokel ES, Grandy DK, Low MJ, Rubinstein M, Ariano MA, Levine MS. Facilitated glutamatergic transmission in the striatum of D2 dopamine receptor-deficient mice. J Neurophysiol 2001; 85: 659–670
  • Seamans JK, Gorelova N, Durstewitz D, Yang CR. Bidirectional dopamine modulation of GABAergic inhibition in prefrontal cortical pyramidal neurons. J Neurosci 2001; 21: 3628–3638
  • Trantham-Davidson H, Lavin A, Seamans JK. Concentration specific modulation of GABAA currents in prefrontal neurons via different dopamine receptor subtypes. Soc Neurosci Abstr 2003; 573: 16
  • Valenzuela CF, Kazlauskas A, Brozowski SJ, Weiner JL, Demali KA, McDonald BJ, Moss SJ, Dunwiddie TV, Harris RA. Platelet-derived growth factor receptor is a novel modulator of type A γ-aminobutyric acid-gated ion channels. Mol Pharmacol 1995; 48: 1099–1107
  • Rocheville M, Lange DC, Kumar U, Patel SC, Patel RC, Patel YC. Receptors for dopamine and somatostatin: formation of hetero-oligomers with enhanced functional activity. Science 2000; 288: 154–157
  • Hillion J, Canals M, Torvinen M, Casadó V, Scott R, Terasmaa A, Hansson A, Watson S, Olah ME, Mallol J, Canela EI, Zoli M, Agnati LF, Ibáñez CF, Lluis C, Franco R, Ferré S, Fuxe K. Coaggregation, cointernalization, and codesensitization of adenosine A2A receptors and dopamine D2 receptors. J Biol Chem 2002; 277: 18091–18097
  • Canals M, Marcellino D, Fanelli F, Ciruela F, de Benedetti P, Goldberg SR, Neve K, Fuxe K, Agnati LF, Woods AS, Ferré S, Lluis C, Bouvier M, Franco R. Adenosine A2A-dopamine D2 receptor–receptor heteromerization: qualitative and quantitative assessment by fluorescence and bioluminescence energy transfer. J Biol Chem 2003; 278: 46741–46749
  • Rimondini R, Fuxe K, Ferré S. Multiple intramembrane receptor-receptor interactions in the regulation of striatal dopamine D2 receptors. Neuroreport 1999; 10: 2051–2054
  • Agnati LF, Ferré S, Lluis C, Franco R, Fuxe K. Molecular mechanisms and therapeutical implications of intramembrane receptor/receptor interactions among heptahelical receptors with examples from the striatopallidal GABA neurons. Pharmacol Rev 2003; 55: 509–550
  • Li M, Bermak JC, Wang ZW, Zhou QY. Modulation of dopamine D2 receptor signaling by actin-binding protein (ABP-280). Mol Pharmacol 2000; 57: 446–452
  • Li M, Li CY, Weingarten P, Bunzow JR, Grandy DK, Zhou QY. Association of dopamine D3 receptors with actin-binding protein 280 (ABP-280). Biochem Pharmacol 2002; 63: 859–863
  • Smith FD, Oxford GS, Milgram SL. Association of the D2 dopamine receptor third cytoplasmic loop with spinophilin, a protein phosphatase-1-interacting protein. J Biol Chem 1999; 274: 19894–19900
  • Bofill-Cardona E, Kudlacek O, Yang Q, Ahorn H, Freissmuth M, Nanoff C. Binding of calmodulin to the D2-dopamine receptor reduces receptor signaling by arresting the G protein activation switch. J Biol Chem 2000; 275: 32672–32680
  • Oldenhof J, Vickery R, Anafi M, Oak J, Ray A, Schoots O, Pawson T, von Zastrow M, Van Tol HHM. SH3 binding domains in the dopamine D4 receptor. Biochemistry 1998; 37: 15726–15736
  • Oldenhof J, Ray A, Vickery R, Van Tol HHM. SH3 ligands in the dopamine D3 receptor. Cell Signal 2001; 13: 411–416
  • Cao WH, Luttrell LM, Medvedev AV, Pierce KL, Daniel KW, Dixon TM, Lefkowitz RJ, Collins S. Direct binding of activated c-Src to the β3-adrenergic receptor is required for MAP kinase activation. J Biol Chem 2000; 275: 38131–38134

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.