SUMMARY
G protein Coupled Receptors (GPCRs) represent the largest family of drug targets. Upon activation GPCRs signal primarily via a diverse set of heterotrimeric G proteins. Most GPCRs can couple to several different G protein subtypes. However, how drugs act at GPCRs contributing to selectivity of G protein recognition is poorly understood. Here, we examined the G protein selectivity profile of the dopamine D2 receptor (D2), a GPCR targeted by antipsychotic drugs. We show that D2 discriminates between 6 individual members of the Gi/o family and its profile of functional selectivity is remarkably different across its ligands which all engaged D2 with a distinct G protein coupling pattern. Using structural modeling, receptor mutagenesis and pharmacological evaluation, we identified residues in the D2 binding pocket that shape these ligand-directed biases. We further provide pharmacogenomic evidence that natural variants in D2 differentially impact its G protein biases in response to different ligands.
eTOC Blurb
Moo et al. report that the dopamine D2 receptor engages different G proteins when activated by dopamine and antipsychotics. They elucidate the structural mechanism underlying G protein bias and show how it can be altered by natural variants and rationally designed mutations.
Graphical Abstract

INTRODUCTION
G protein-coupled receptors (GPCRs) form the largest class of receptors that recognize diverse types of ligands, including ions, small molecules and peptides, linking them to downstream signals (Alexander et al., 2019). They are prominent drug targets, responsible for the therapeutic effect of ~30–40% of approved drugs (Hauser et al., 2017; Hauser et al., 2018; Insel et al., 2019; Santos et al., 2017). All canonical GPCRs transduce their signals by activating heterotrimeric G proteins that entails the release of the GTP-bound Gα subunit from the inseparable Gβγ complex (Flock et al., 2015; Olsen et al., 2020; Wettschureck and Offermanns, 2005). Mammalian genomes encode a diverse set of 16 Gα proteins, divided into four major families based on sequence homology and function: Gi/o, Gs, Gq/11 and G12/13 which uniquely regulate downstream effectors (Hermans, 2003; Milligan and Kostenis, 2006). It is now well-established that individual GPCRs can couple to multiple different Gα proteins and that specificity of this recognition is the key parameter defining GPCR properties (Inoue et al., 2019; Masuho et al., 2015b; Michel and Charlton, 2018). In addition, GPCRs can signal by engaging β-arrestin adaptors that upon activation scaffold a variety of signaling molecules also generating cellular responses (Shenoy and Lefkowitz, 2011; Shukla et al., 2011; Wootten et al., 2018).
One of the most exciting developments in understanding GPCRs has been the realization that they can differentially route the signals depending on the nature of the ligand engaged, a phenomenon termed biased signaling or functional selectivity (Kenakin, 2011, 2019; Spongier et al., 1993; Urban et al., 2007). Biased agonists are theorized to stabilize different receptor conformations, which exhibit different affinity for signal transducers to produce distinct cellular responses (Kenakin and Miller, 2010; Michel and Charlton, 2018; Smith et al., 2018; Wootten et al., 2018). The concept of biased signaling offers an attractive possibility for achieving selective pharmacological modulation of GPCR signaling outcomes (Hoffmann et al., 2008). This may allow to dissociate desirable therapeutic effects associated with favorable signaling pathways from the side-effects driven by unfavorable signals, a concept fueling an immense drug discovery effort for identifying and characterizing GPCR ligands with biased signaling properties (Kenakin, 2019; Kenakin and Christopoulos, 2013; Violin et al., 2014).
The best-known example of biased signaling involves differential recruitment of β-arrestin vs. activation of G proteins, a discrimination that many GPCRs have been shown to be capable of (Rajagopal et al., 2010; Reiter et al., 2012; Wacker et al., 2017). Exploiting this phenomenon, a number of synthetic biased ligands has been developed for several receptors. They are capable of preferential engagement of either β-arrestin or G protein and show substantial promise for enhancing the therapeutic effect window (Becker et al., 2016; Bonifazi et al., 2019; Ryba et al., 2017; Singla et al., 2017; Viscusi et al., 2016). Ultimately, the concept of biased singling can be generalized to differential engagement of various downstream pathways initiated by GPCRs which occurs upon alteration of the balance in activation of different proximal signaling mediators (Urban et al., 2007). Considering that G proteins are universal transducers of signals engaged by most GPCRs in a physiological setting, their differential activation has been suggested to greatly contribute to biased signaling (Kenakin, 2019). Indeed, several examples attest to a possibility that GPCRs can differentially engage G proteins in a biased fashion (Bonifazi et al., 2019; Masuho et al., 2015b; Randáková et al., 2020). However, biased GPCR signaling across G proteins remains poorly explored, especially from the angle of understanding the actions of drugs in clinical use. Furthermore, the impact of genetic variations that prominently affect many GPCRs on their G protein selectivity and signaling bias is largely an uncharted territory (Hauser et al., 2018).
Receptors for dopamine are among the most canonical GPCRs with important roles in physiology and disease (Beaulieu et al., 2015). Dysfunction in dopamine signaling has been linked to numerous neuropsychiatric disorders, including schizophrenia, depression and Parkinson’s disease (Beaulieu and Gainetdinov, 2011). In mammals, there are two subfamilies of dopamine receptors. The stimulatory D1 class includes dopamine D1 and D5, which activate primarily the Gs/olf subfamily of G proteins and signal to increase production of the second messenger, cAMP. The D2 class comprises inhibitory dopamine D2, D3, and D4 receptors, which couple to the Gi/o proteins and engage several signaling mediators, (Neve et al., 2004). Most members of the Gi/o family, except for sensory subunits Gαt1, Gαt2 and Gαgust, are ubiquitously expressed across the nervous system (Jiang and Bajpayee, 2009; Syrovatkina et al., 2016; Uhlen et al., 2015). However, activation of individual members can produce varying outcomes. For example, Gαi1, Gαi2 and Gαi3 proteins inhibit most adenylyl cyclase (AC) isoforms, reducing cAMP production (Sunahara et al., 1996). In contrast, Gαo does not directly interact with AC and instead acts as signal modifier via the release of Gβγ subunits which differentially modulate activities of various AC isoforms (Dessauer et al., 2017). Gαz has a number of unique properties, including extremely long lifetime in the active state, insensitivity to pertussin toxin and ability to stimulate potassium channels (Fields and Casey, 1997; Ho and Wong, 2001).
Among the dopamine receptors, D2 is a major drug target for the treatment of schizophrenia and many last generation atypical antipsychotic drugs in clinical use act as D2 partial agonists (Ginovart and Kapur, 2012; Mauri et al., 2014; Miyamoto et al., 2012). Interestingly, these drugs produce a range of the distinct efficacies and side effects. One particularly interesting hypothesis with respect to explaining varying effects relates to D2 signaling bias (Mailman and Murthy, 2010; Urs et al., 2017). Biased signaling of D2 has been investigated intensely, in respect to its activation of G protein vs. β-arrestin pathways. Studies suggest that β-arrestin2 recruitment is essential for the antipsychotic activity of D2 acting compounds, yet there is conflicting evidence in respect to β-arrestin bias and antagonism in effectiveness of psychosis treatment (Allen et al., 2011; Madras, 2013; Masri et al., 2008; Urs et al., 2017). Hence, the link between antipsychotic efficiency and signaling bias of D2 remains unclear.
In this study, we report that dopamine and several antipsychotics with partial agonism at D2 are able to induce biases amongst different Gα subtypes at the D2. We identified the structural determinants in D2 involved in determining G protein signaling biases induced by various ligands. Finally, we showed that genetic variants of D2 display biased G protein activation. Taken together, our study highlights the importance of studying the initial step of receptor activation to produce a comprehensive profile of ligand signaling and provide information that can aid in the design of better D2-targeted therapeutics. These findings also suggest that ligand directed bias across G protein subtypes may be a universal phenomenon applicable to many other GPCRs.
RESULTS
Dopamine activates D2 with distinct G protein selectivity profile favoring Gαo.
Previous studies established that D2 couples exclusively to the Gi/o family of Gα proteins with seemingly varying efficiency across individual subtypes (Beaulieu et al., 2015; Masuho et al., 2015b). To follow up on previously observed qualitative differences in D2 coupling to G proteins (Masuho et al., 2015b), we quantitatively measured the ability of D2 to activate different non-sensory Gαi/o family members: Gαi1, Gαi2, Gαi3, GαoA, GαoB and Gαz using a NanoBRET-based biosensor approach (Masuho et al., 2015a)(Fig. 1A). Stimulation of D2 with a saturating concentration of the endogenous agonist, dopamine, revealed that it differentially activated various G proteins (Fig. 1B). We have previously established equivalence of stoichiometric ratios of all G protein heterotrimers used in this assay as well as specificity of the signal not observed when G protein subunits or receptors are skipped from the transfection (Masuho et al., 2015a). With these assay conditions, we observed the largest BRET signal amplitudes for GαoA and GαoB followed by Gαi1–3 and low but significant activation of Gαz (Fig. 1C). Because the extent of G protein activation measured by the signal amplitude may be influenced by intrinsic G protein properties, we focused our subsequent analysis on evaluating the kinetics of the BRET signal generation (Fig. 1D). The onset kinetics reflects the direct measure of the catalytic reaction of G protein activation and thus provides a more accurate measure of GPCR activity. Using exponential fitting of the rising phase of the BRET response we found that application of saturating dopamine concentration activated GαoA and GαoB the fastest (2.43 ± 0.27 s−1 and 2.28 ± 0.15 s−1, respectively), followed by the markedly slower activation of Gαi1–3 proteins (Gαi1, 1.12 ± 0.01 s−1; Gαi3, 0.74 ± 0.04 s−1; Gαi2, 0.58 ± 0.06 s−1) and very slow Gαz activation (0.03 ± 0.01 s−1).
Figure 1. Activation profile of Gai/o proteins by D2 stimulated by dopamine.

(A) Schematic representation of the NanoBRET assay used to measure D2R G protein activation. Gβγ is tagged with Venus and masGRK3ct is tagged with Nano luciferase (Nluc). Agonist stimulation of D2 leads to dissociation of Gαβγ trimer. Gβγ-Venus then associates with Gβγ effector mimetic masGRK3ct-Nluc to product BRET response. Different Gα responses are recorded by transient transfection with Gα of choice. (B) Representative traces of the effect of 100 μM dopamine on Gi1, Gi2, Gi3, GoA, GoB, Gz activation and no G protein transfected by D2 measured over 40 seconds. Four independent experiments were performed in triplicates. (C) Maximum BRET response of dopamine for D2R individual G protein response. (D) Activation kinetics of dopamine for D2R individual G protein response. (E) Dopamine concentration-response curves for D2R Gi/o protein activation. Three independent experiments were performed in duplicates. (F) Potency (pEC50) of dopamine at D2R individual G protein response. (G) Correlation plot showing the relationship between D activation rate and F potency of dopamine Gi/o activation. (H) The differences in activation rate of Gi/o proteins are illustrated in web plot constructed from data in D. * denotes significantly different when compared to value for Gi1, * denotes significantly different when compared to value for Gi2, * denotes significantly different when compared to value for Gi3, * denotes significantly different when compared to value for GoA, * denotes significantly different when compared to value for GoB, * denotes significantly different when compared to value for Gz, p<0.05, one-way ANOVA with Tukey’s post hoc test. The values in B-G are expressed as mean ± SEM. See also Figure S1.
We have previously noted that G protein activation kinetics can be used to approximate the potency of GPCR responses (Masuho et al., 2015b), suggesting that dopamine-stimulated D2 activates various Gαi/o subtypes with different potencies. To directly test this possibility, we conducted concentration-response studies measuring BRET response amplitudes mediated by individual Gαi/o proteins at different dopamine concentrations (Fig. 1E). The analysis of the data showed that potencies of the dopamine responses mediated by different Gαi/o subtypes also varied and had the same rank order as kinetic differences. Namely, the Gαo signaling was the most sensitive, followed by Gαi1–3 and least response sensitivity was observed for the Gαz (Fig. 1F). Furthermore, we found a nearly perfect correlation (R2 = 0.93) between activation rates and pEC50 values for dopamine mediated by different Gαi/o subtypes (Fig. 1G). Importantly, the GαoA-B > Gαi1–3 > Gαz rank order in activation rates was maintained across dopamine concentrations (Fig. S1). As the concentration of dopamine declined to the lowest range, D2 lost coupling to Gαz first, followed by abolished coupling to Gαi1–3 while maintaining detectable activation of GαoA-B. These observations indicate that potency of D2 responses to dopamine varies depending on the Gα subtype transducing the signal and stems from differences in efficiencies with which D2 can catalyze the activation of its individual G protein substrates (Fig. 1H). Therefore, dopamine induces a distinct signaling profile at D2 with differential selectivity amongst the members of the Gαi/o family.
Differential ligand-directed G protein biases at the D2.
Given the multitude of small-molecule agonists developed for D2 and their documented differences in signaling properties such as G protein vs. β-arrestin bias (Allen et al., 2011; Bonifazi et al., 2019; Free et al., 2014), we next investigated if they have different Gα selectivity bias of D2. To address this experimentally, we evaluated a panel of D2-targeting drugs with divergent chemical scaffolds (Fig. 2A; Table S1). When used at the saturating concentrations, all ligands produced smaller responses as compared to dopamine, with the maximum BRET amplitudes ranging from 14% to 90% of dopamine response consistent with their partial agonism (Fig. S2). As expected, we observed that the rank order of efficacy defined by maximal BRET amplitudes of GαoA-B > Gαi1–3 > Gαz remained equivalent to dopamine responses consistent with the notion that response efficacies are mostly driven by intrinsic Gα subtype properties rather than their activation by D2.
Figure 2. Antipsychotics show distinct Gi/o protein activation profile.

(A) The ability of five antipsychotics with divergent chemical scaffolds (MLS1547, bifeprunox, cariprazine, aripiprazole and brexpiprazole) to activate Gi1, Gi2, Gi3, GoA, GoB and Gz at D2 were measured over 60 seconds. (B) 100 μM of each agonist was used. The data was normalized to the maximal response induced by the agonist at that pathway. Four independent experiments were performed in triplicates. (C) Activation kinetics of agonists for D2R individual G protein response. * denotes significantly different when compared to value for Gi1, * denotes significantly different when compared to value for Gi2, * denotes significantly different when compared to value for Gi3, * denotes significantly different when compared to value for GoA, * denotes significantly different when compared to value for GoB, * denotes significantly different when compared to value for Gz, p<0.05, one-way ANOVA with Tukey’s post hoc test. + denotes significantly different when compared to corresponding values for dopamine, p<0.05, two-way ANOVA with Dunnett’s post hoc test. The values are presented as mean ± SEM. (D) The differences in activation rate of Gi/o proteins are illustrated in web plot constructed from data in C. The values are represented as mean only. (E) The 1/τ activation rates from C were normalized to reference agonist dopamine, and then normalized to reference pathway GoA activation to obtain 1/τ activation rate ratio. The values are showed in web plots. Open circles indicate significant differences when compared to 1/τ activation rate of dopamine at the same pathway, p<0.05, determined by one-way ANOVA with Tukey’s post hoc test. See also Figure S2, Table S1.
In contrast, analysis of the activation kinetics (1/τ), as the main parameter defining catalytic D2 action and its response potencies revealed substantial heterogeneity in the behavior of the ligands, which displayed distinct activation profile of Gα subtypes (Fig. 2B). The response profile triggered by MLS1547 largely resembled that of dopamine with all Gαi/o subtypes being engaged (Fig. 2C, D). It still activated Gαo better than Gαi, however the extent of this preference was diminished relative to dopamine. Bifeprunox was also able to activate all Gαi/o subtypes, however it no longer favored GαoA-B over Gαi1–3 (Fig. 2C, D). Similar lack of Gαi vs. Gαo preferences was exhibited by cariprazine and aripiprazole, which additionally lost their ability to activate Gαz. Finally, brexpiprazole showed an extreme preference for both Gαo isoforms and was unable to activate any other G proteins (Fig. 2C, D). None of these agonists showed G protein activation in the absence of transfected D2 (Fig. S2) showing that the effects are driven by changes in the D2 properties.
To probe whether G protein biases seen with different ligands are related to the induction of various agonist states we evaluated the effects of antipsychotics in the antagonist mode studying their ability to inhibit signaling via Gαi1 or GαoA initiated by dopamine. As a reference, we used a canonical D2 antagonist, haloperidol, which showed equivalent potency in inhibiting both Gαi1 and GαoA signaling (Fig. S3). All representative antipsychotics used in these experiments behaved similar to haloperidol and were equipotent in inhibiting D2 signaling via both G proteins, suggesting that their biased signaling properties may indeed be related to the induction of specific agonist states.
In order to determine whether the observed changes in G protein selectivity truly reflect biases in G protein activation, we compared the response profiles induced by antipsychotics in reference to dopamine selectivity profile (Fig 2E), an approach commonly used to estimate biased agonism (Kenakin et al., 2012). In this analysis, the coupling profile elicited by dopamine is set to an equal value for each G protein and deviations produced by other ligands become apparent as increases or decreases in values relative to the dopamine. Analysis of the data showed that different agonists indeed generated distinct fingerprints of G protein selectivity different from the profile triggered by dopamine. The unifying theme of these changes was a modulation of either Gαi or Gαo selectivity of D2. Overall, these results show that D2 exhibits a clear ligand-directed bias in the activation of G proteins.
Dopamine-induced G protein bias of D2 is determined by ligand-binding residues.
Differences in G protein bias of D2 observed from ligands with varied molecular structures suggest that interactions with different residues in the binding pocket can contribute to selective recognition of different Gαi/o subtypes. To test this hypothesis, we sought to identify residues determining Gα selectivity bias by modeling of ligand-receptor structure complexes constructed from induced-fit docking in an active state homology model of D2 generated with the GPCRdb pipeline (Pandy-Szekeres et al., 2018) (Fig. 3). All five antipsychotics form the canonical salt-bridge to D1143×32 (GPCRdb generic residue numbering scheme (Isberg et al., 2015) in superscript) and aromatic edge-to-face π-π interactions to F3896×51 and F3906×52 (Fig. S4A); all of which are conserved residues and interactions of aminergic ligand-receptor complexes (Peng et al., 2018). All ligands except MLS1547 share binding mode in the dopamine-binding (orthosteric) pocket (Vass et al., 2019; Wang et al., 2017) lined by side chains of eight interacting residues: V1153×33, C1183×36, I18445×52, S1935×43, S1975×461, H3936×55, Y4087×34, T4127×38 and Y4167×42. In contrast, all ligands but cariprazine display two spatially different binding modes in sites extending towards Y4087×34 in TM7 or F1103×28 in TM3 and capped by W10023×50 in the first extracellular loop (ECL1) (Fig. 3 and Fig. S4B–C). Notably, MLS1547 flips not only horizontally, but also vertically, in the two best scoring binding poses. Our selection is reinforced by the experimental evidence that residues S1935×43 and S1975×461, which are conserved in all dopamine receptor subtypes, influence agonist binding and efficacy (Chemel et al., 2012; Cummings et al., 2010; Fowler et al., 2012; Sartania and Strange, 1999) and that residue H3936×55 influences D2 signaling bias and efficacy (Tschammer et al., 2011).
Figure 3. Putative binding modes of antipsychotics in D2.

One (cariprazine) or two proposed binding modes for the five studied antipsychotics from induced-fit docking in a D2 active state homology model. Antipsychotics are displayed as thin sticks with carbon atoms colored as in Figure 2 with a slight nuance difference between the two suggested poses for the same compounds. D2 is depicted as beige cartoon and thick sticks for the backbone and the carbon atoms of residues selected for mutagenesis. Other atoms have the following colors: oxygen – red, nitrogen – blue, sulphur – yellow and chlorine – green. The receptor is viewed from TM1+2 and, for clarity reasons, these two helices together with hydrogen atoms are not displayed. Additionally, only one receptor conformation is depicted, though, induced-fit docking produces varying amino acid rotamers corresponding to individual binding poses. GPCRdb generic numbers accompany the D2 sequence numbers in superscript (Isberg et al., 2015). See also Figure S4, Table S4. Figure prepared with the PyMOL Molecular Graphics System, Version 2.0 Schrödinger, LLC.
To evaluate their roles as determinants for ligand-dependent biased signaling, we designed 17 single-point mutants of the 11 antipsychotics interacting residues and evaluated the effect of the mutations on dopamine-induced activation of representative members along the axis of bias: Gαi1 and GαoA (Fig. 4). We found that all D2 mutants were present on the surface in levels comparable to the wild-type receptor (Fig. S5), ensuring that any alterations in their function would not be due to differences in expression or localization. Most of the 17 mutations significantly slowed the G protein activation kinetics of D2 when activated by dopamine (Fig. 4A–C; Table S2). Nine mutations affected Gαi1 activation while 13 slowed activation of GαoA. Three mutations (I18445×52F, Y4087×34F and T4127×38V) had no significant impact on the activation of either Gα, while one mutation (S1975×461A) completely abrogated the functional activity of D2 on both. In order to evaluate whether the mutations affected Gαo vs. Gαi signaling balance thereby changing receptor preferences we calculated the G protein bias (Fig. 4E). With this metric we found that ten of the mutations significantly affected selectivity of D2 diminishing its preference for GαoA over Gαi1. The degree of this effect varied amongst the affected mutants and in two cases (S1935×43A and Y4167×42W) completely eliminated the GαoA preference seen in the wild-type D2 making respective receptors equipotent in activation of both the Gαi and Gαo branches. Dopamine binding affinity was only affected by the mutations, S1975×461A and Y4167×42W (Table S2). In summary, we uncovered a range of spatially and physicochemically different determinant residues for the Gαo versus Gαi protein selectivity of dopamine-activated D2.
Figure 4. D2 mutants identified using structural modeling display dopamine-induced Gi/o proteins bias.

(A) Snake plot of D2R with mutations studied highlighted in their respective colors. (B) The ability of 100 μM dopamine to activate Gi1 and GoA at D2R mutations were measured with BRET G protein activation assay over 60 seconds. The data were normalised to the maximal response induced by dopamine at GoA activation. Activation kinetics of dopamine for D2 WT and mutants (C) Gi1 and (D) GoA protein response. * denotes significantly different values when compared to D2 WT, p<0.05, determined by one-way ANOVA with Tukey’s post hoc test. (E) GoA - Gi1 bias was determined by normalising 1/τ of GoA to Gi1 activation pathway. Open circles denote significantly different values when compared to D2 WT, p<0.05, determined by one-way ANOVA with Tukey’s post hoc test. Data are presented as mean ± SEM of three experiments performed in triplicates. ND: not determinable. See also Figure S5, S6, Table S2.
Determinants underlying ligand-induced alteration of G protein biases.
We next evaluated if mutation of the four ligand interacting residues in the two extended binding sites would influence, or even switch, the Gαi1 vs. GαoA bias of the most structurally distinct ligands we evaluated: MLS1547, bifeprunox and aripiprazole on D2 (Fig. 5; Table S3). Remarkably, the W100A23×50, F110A3×28, Y408A7×34 and S193A5×43 mutations exhibited a diverse spectrum of effects on the different ligands and G proteins. MLS1547-D2 signaling was affected significantly by all four mutations, but in different ways, as W100A23×5, S193A5×43 and Y408A7×34 selectively reduced GαoA activation, whereas F110A3×28 abolished signaling through both Gαi1 and GαoA. Aripiprazole-D2 signaling changes were more selective, as only the S193A5×43 and F110A3×28 mutations had effects which again differed – augmenting and diminishing the responses of the two Gα subtypes, respectively. Finally, bifeprunox-D2 signaling was only influenced by the Y408A7×34 mutation, which results in a slight reduction specific for GαoA.
Figure 5. Antipsychotics display Gi/o protein bias at D2R mutants.

(A) The ability of 100 μM of MLS1547, bifeprunox and aripiprazole to activate Gi1 and GoA at D2 mutations were measured over 60 seconds. The data were normalized to the maximal response induced by dopamine at GoA activation. Activation kinetics of antipsychotics for D2R WT and mutants (B) Gi1 and (C) GoA protein response. * denotes significantly different values when compared to D2 WT, p<0.05, determined by one-way ANOVA with Tukey’s post hoc test. (D) GoA - Gi1 bias was determined by normalizing 1/τ of GoA to Gi1 activation pathway. Open circles denote significantly different values when compared to D2 WT, p<0.05, determined by one-way ANOVA with Tukey’s post hoc test. Data are presented as mean ± SEM of three experiments performed in triplicates. ND not determinable. # denotes that the value was constrained to GoA bias because Gi1 pathway was not activated. See also Table S3.
To better understand the impact of mutations on the agonist bias of Gα preferences we calculated G protein bias (Fig. 5D). This analysis revealed that mutations produced selective bidirectional effects on Gα selection by D2 in an agonist selective manner. For instance, both W100A23×50 and Y408A7×34 flipped the selectivity of D2 to a strong preference for Gαi1 instead of GαoA when stimulated with MLS1547. In contrast the Y408A7×34 mutation made aripiprazole selective for GαoA at the D2 by abolishing Gαi1 coupling. Similar results were observed using an orthogonal assay platform, the TRUPATH technology (Fig. S6) (Olsen et al., 2020). Taken together, these observations identified structural determinants in the dopamine-binding site of D2 that uniquely modulate the G protein bias of antipsychotics.
Genetic variants influence agonist-directed bias in G proteins selectivity of D2.
Since natural GPCR mutations have been linked to different drug therapy efficacy, toxicity and diseases (Hauser et al., 2018; Insel et al., 2007), we next tested if reported naturally occurring genetic variations in D2 alter the G protein selectivity of dopamine and antipsychotics causing or perturbing the medication of disease, respectively. V1544×44I (rs10489422; Allele Frequency: 0.00004602), located at TM4 close to the intracellular domain of the receptor, has been associated with myoclonus dystonia without noting any functional alterations of traditional measures for agonist or antagonist affinities and functional responses (Klein et al., 1999; Klein et al., 2000). S311ICL3C (rs1801028; Allele Frequency: 0.02572, the most frequently observed missense mutation in D2) (Karczewski et al., 2020), in the middle of intracellular loop 3 (ICL3), has been associated with an increased risk for schizophrenia (Arinami et al., 1996; Glatt and Jonsson, 2006; Yao et al., 2015) and described to lower the affinity of dopamine and diminish its efficacy in inhibiting cAMP synthesis (Cravchik et al., 1996). We have then examined the signaling properties of both mutants in our functional assays determining the impact on the Gα biases. The V1544×44I variant selectively increases dopamine- and aripiprazole-D2 activation of Gαi1 and augments MLS1547-D2 GαoA signaling (Fig. 6A, B). Thus, this variant produces two types of D2 signaling changes that switch from Gαo to Gαi selectivity across all three agonists. In contrast, the S311ICL3C variant only significantly changes the response of aripiprazole for which it augments both Gαi1 and GαoA signaling. Evaluation of the signaling bias indicated significant change with Gαi over Gαo selectivity induced by both mutations in response to aripiprazole, and S311C showed preference for Gαi signaling when activated by dopamine (Fig. 6C). We conclude that naturally occurring variants in D2 can change the balance of its G protein preferences in an agonist selective manner. These changes may lead to altered drug responses with physiological implications.
Figure 6. Natural genetic variants of D2R show different G protein activation.

Activation kinetics of 100 μM dopamine, MLS1547 and aripiprazole for D2 WT and the natural genetic variants V154I and S311C at (A) Gi1 and (B) GoA protein response. * denotes significantly different values when compared to D2 WT, p<0.05, determined by one-way ANOVA with Tukey’s post hoc test. (C) GoA - Gi1 bias factor was determined by normalizing 1/τ of GoA to Gi1 activation pathway. Open circles denote significantly different values when compared to D2 WT, p<0.05, determined by one-way ANOVA with Tukey’s post hoc test. Data are presented as mean ± SEM of three experiments performed in triplicates.
DISCUSSION
Biased G protein signalling profile of the D2 dopamine receptor
In this study we show, that the prototypical GPCR, the D2 dopamine receptor shows prominent ligand-dependent functional selectivity at the level of G protein activation based on its ability to discriminate between individual members of the Gαi/o subfamily. The endogenous agonist dopamine is capable of triggering D2 activity on all non-sensory members of the Gαi/o subfamily: GαoA, GαoB, Gαi1, Gαi2, Gαi3 and Gαz but has an intrinsic signaling profile of preferring Gαo over Gαi and Gαz activation. It should be noted that D2 is exquisitely selective for the Gαi/o members and does not show appreciable activation of other G protein subtypes (Masuho et al., 2015b) making this investigation an exhaustive profiling of changes in D2-Gα preferences. Our results indicate that dopamine-bound D2 is functionally selective at the first step of signaling following receptor activation, prior to any consequent downstream second messenger system. This measure of functional outcome allows for direct comparison between diverse G protein pathways, uncovering new bias activities, different from previous bias studies that focused on downstream second messenger readouts (Allen et al., 2011; Chen et al., 2016; Tschammer et al., 2011). Even though the individual Gαi/o members are classified in the same Gα family, their activation have significantly diverse functional outcomes pertaining to effector engagement selectivity and kinetic profiles (Ho and Wong, 2001; Jiang and Bajpayee, 2009), suggesting that bias within the Gαi/o subfamily may have physiological and/or therapeutic implications (Anderson et al., 2020).
Our results are consistent with several prior studies that noted differences in G protein activation by the D2 and its preference for Gαo (Bonifazi et al., 2019; Klein Herenbrink et al., 2016; Masuho et al., 2015a; Sanchez-Soto et al., 2016). We substantially extended these observations by providing a systematic comparative analysis of all Gα subunits coupled to D2 including the often neglected Gαz protein that binds to D2 in vivo (Leck et al., 2006). Uniquely, we also systematically compare the G protein profiles engaged by dopamine to those in response to a structurally diverse class of other D2 ligands to reveal their differential biases. We report that partial agonists with varying degrees of clinical efficacy also have distinct G protein activation profiles compared to the full agonist dopamine. In contrast to the clear preference for Gαo signaling of dopamine, partial agonists bifeprunox and cariprazine have a more balanced Gαi/o activation profile. Importantly, we observe that partial agonists do not simply subdue the signaling via the weaker coupled Gα subtypes, which would be expected to proportionally scale down the response to result in a perceived bias due to loss of D2 coupling to less preferred substrates, a phenomenon observed for μ-opioid (Gillis et al., 2020) and M1 muscarinic (Masuho et al., 2015b) receptors. Instead, we observe a rank-order change in the D2 preferences for individual Gα subtypes induced by antipsychotics as compared to dopamine reflecting a true functional bias. Together with previous findings, our results form a comprehensive picture of the G protein biased signaling profile of the D2 and uncovers its potential for modulation by drugs.
Determinants of G protein discrimination by the D2 receptor
From the structural standpoint, biased signaling is thought to be underpinned by the ability of ligands to stabilize specific receptor conformation that lead to differential engagement of signaling partners and cellular outcomes (Kenakin, 2011). The partial agonists included in this study are bivalent molecules that are able to simultaneously occupy the orthosteric binding site and one of two spatially distinct secondary extended binding pockets (Free et al., 2014; Klein Herenbrink et al., 2019; Szabo et al., 2014). The nature of the head group can allosterically influence ligand binding mode in both spaces and thus affect the global receptor conformation (Weichert et al., 2015). Besides aripiprazole and cariprazine, all partial agonists have a distinct chemical scaffold with diverse primary and secondary structures. Indeed, molecular dynamics has shown that dopamine and aripiprazole are able to induce different conformations of important GPCR structural motifs, including the extracellular loops, ligand binding sites and intracellular G protein-binding domains, contributing to their biased signaling profiles (Kling et al., 2014).
Our study presents new insights into the structural basis of the identified D2 biased signaling. Dopamine at the wild type receptor has a clear Gαo bias over Gαi subtypes, and based on docking to a D2 homology model we were able to pinpoint amino acid residues involved in setting this selectivity and overriding it. As only an antagonist bound D2 structure (Wang et al., 2018) was available when the current project was started, we modelled an active state D2 receptor to take structural changes in the binding cavity into account. Two additional inactive (Fan et al., 2020; Im et al., 2020) and two active state (Yin et al., 2020; Zhuang et al., 2021) D2 structures are now available. Comparison of all five structures to our model shows a similar overall location of the binding site residues. Furthermore, the induced-fit docking protocol employed herein compensates for different side chain rotamers making it likely to obtain similar binding poses independent of D2 structure employed. Nevertheless, mutational experiments based on the model showed that the S1935×43A mutation in the orthosteric site was able to completely abolish the intrinsic bias of D2 towards Gαo indicating its importance in dictating G protein preferences of the receptor. We also find that mutating residues in the extended binding pockets that indirectly coordinate dopamine reduced D2 bias for Gαo but did not erase its preference over the Gαi upon activation by dopamine. This structural information can be utilized to design ligands that differentially engage the residues with the hope of generating small molecules mimicking the signaling profile on the wild-type receptor. Of note, only one of the two proposed binding modes for MLS1547 and aripiprazole provides a structural explanation for the marked effect of the F1103×28A mutation, which should be taken into account using our results for structure-based design of bias ligands. We envision that the mutants with altered G protein selectivity that we describe could be used for in vivo studies to explore the behavioral consequence of changing the balance of Gαi/o signaling at D2.
Significant insights are generated from the analysis of how mutations alter the D2 responses to antipsychotics introducing another dimension of biased signaling. Previous data showed that aripiprazole displayed bias towards cAMP over pERK1/2 and mutating ligand binding site residues did not cause a significant change in bias (Klein Herenbrink et al., 2019; Szabo et al., 2014). However, we find that aripiprazole, MLS1547 and bifeprunox have drastically different profiles of G protein activation at the D2 mutants making the ligands functionally selective for Gαi signaling. Most noteworthy, the F1103×28A mutation displayed no G protein activation by MLS1547. The residues identified are also able to affect antipsychotic activity, creating different antipsychotic signaling profiles at the mutant receptors. Overall, we were able to identify receptor microdomains that participate in mediating ligand-specific conformations associated with biased signaling, and also detected residues that are not involved in this process.
Pharmacogenomic implications of analyzing D2 signaling biases
It is interesting to consider our observations with rationally designed mutations in D2 that altered its G protein selectivity profile from the perspective of naturally occurring variants. It is possible that drug-induced biases can be broadly extended when one considers non-synonymous polymorphisms in D2. We have evaluated two such representative naturally occurring alterations in D2, V1544.44I and S311ICL3C, and found that they also have vastly different G protein signaling profiles relative to wild type receptor. Generally, dopamine was less biased for the Gαo pathway, while MLS1547 had enhanced bias towards Gαo with these variants. Meanwhile, the D2 genetic variants preferred the Gαi pathway when the receptor was activated by aripiprazole. Interestingly, S311ICL3C was found in patients with psychotic affective disorders, making it plausible that people with these mutations could be prescribed antipsychotics as part of their treatment (Arinami et al., 1996). Therefore, with information on more genetic variants, it may be required to examine putative antipsychotics in genotyped-matched clinical trials. Additionally, it would be ideal to study patient’s genotype before initializing treatment to prevent unexpected drug responses and adverse reactions. The outcome highlights the importance of a personalized approach in prescribing medications based on genetic make-up, a concept also applicable to other GPCR variants (Hauser et al., 2018).
Conclusion
In summary, we propose that it is important to consider biased signaling across various Gα subtypes when profiling ligands and genetic variants, and when developing drugs with better therapeutic efficacy and reduced side effects. We provide a framework for the analysis that includes direct evaluation of GPCR selectivity for the spectrum of G proteins using proximal assays, predictive structural modeling and the functional surveillance of ligand panels across receptors carrying mutations and natural variants. With the rising importance of personalized medicine, pharmacogenomic discrimination among related G proteins should provide an additional dimension to the development and prescription of antipsychotics.
STAR METHODS
Resource Availability
Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Kirill Martemyanov (kirill@scripps.edu).
Materials availability
Plasmids generated in this study will be distributed upon request without restriction.
Data and code availability
The source data for raw traces of GPCR responses used for quantitative analysis reported in this study have not been deposited in a publicly available repository because of their trivial nature and custom format they are generated in. They have been archived locally on the institutional cloud service. To request access, please contact the Lead Contact. The paper does not report any original code. Any additional information to reanalyse the data reported in this paper is available from the lead contact upon request.
Experimental Model and Subject Details
Cell lines
HEK293T/17 cells were obtained from ATTC (Manassas, VA) and grown in DMEM supplemented with 10% FBS, minimum Eagle’s medium non-essential amino acids, 1mM sodium pyruvate, and antibiotics (100 units/ml penicillin and 100 μg/ml streptomycin) at 37°C in a humidified incubator containing 5% CO2.
Method Details
cDNA constructs
Flag-tagged dopamine D2 receptors (NM_000795) containing the hemagglutinin signal sequence (KTIIALSYIFCLVFA) at the N terminus was a gift from Dr. Abraham Kovoor. The pCMV5 plasmids encoding rat GαoA, rat Gαi1, rat Gαi2 and rat Gαi3, were gifts from Dr. Hiroshi Itoh. pcDNA3.1+ plasmids encoding GαoB (AH002708) and Gαz (J03260) were purchased from the cDNA Resource Center (www.cDNA.org). Venus 156–239-Gβ1 (amino acids 156–239 of Venus fused to a GGSGGG linker at the N terminus of Gβ1 without the first methionine (NM_002074)) and Venus 1–155-Gγ2 (amino acids 1–155 of Venus fused to a GGSGGG linker at the N terminus of Gγ2 (NM_053064)) were gifts from Dr. Nevin A. Lambert (Hollins et al., 2009). The masGRK3ct-Nluc-HA and PTX-S1 were reported previously (Gulati et al., 2018; Masuho et al., 2015a; Raveh et al., 2010). HiBiT tagged D2 and D2 mutants were obtained from Genscript. TRUPATH assay related cDNA was a gift from Bryan Roth (Addgene kit #1000000163) (Olsen et al., 2020). GenBank accession number for each sequence is given in parentheses.
Cell transfection
On the day of transfection, HEK293T/17 cells were split into 2×106 cells onto 35mm plates in culture medium supplemented with 10μg/ml Matrigel. Four hours later, the cells were transfected using desired constructs (total of 5μg DNA) with Lipofectamine LTX (5μL) and Plus (5μL) reagent. For BRET G protein activation assay, the cells were transfected with the Venus 156–239-Gβ (1), Venus 1–155-Gγ2 (1), and masGRK3ct-Nluc (1) constructs in addition to D2 WT or mutants (1) and the Gα of interest: GαoA (2), GαoB (1), Gαi1 (1), Gαi2 (2), Gαi3 (1.5), Gαz (1.5). PTX-S1 (1) was added to cells transfected with Gαz to avoid coupling of endogenous Gai/o to the receptor (the number in parentheses indicates the ratio of transfected DNA, ratio 1 = 0.21μg). The empty vector pcDNA3.1(+) was used to normalize the amount of DNA to 5μg in each transfection.
BRET G protein activation assay
Agonist-dependent BRET measurements between Venus-Gβ1γ2 and masGRK3ct-Nluc were performed as described previously (Masuho et al., 2015a; Masuho et al., 2015b). 16 to 24 hours after transfection, HEK 293T/17 were washed once with BRET buffer (PBS containing 0.1% glucose and 0.5mM MgCl2) and then were gently detached using BRET buffer. The cells suspension was centrifuged at 500g for five minutes and were resuspended in BRET buffer. Approximately 50,000 to 100,000 cells were added into 96-well flat-bottomed white microplates (Greiner Bio-One). The Nano-Glo Luciferase assay substrate was prepared according to manufacturer’s instructions. The substrate was dissolved in BRET buffer immediately before use and added to the cells at a final dilution factor of 1:250, then a 5 second baseline measurement was recorded. 100μM of agonist was injected into each well and activation of D2 was measured at 20ms intervals using a LUMIstar plate reader (BMG LabTech). All measurements were performed at room temperature. The BRET ratio was calculated as ratio of light emitted at 535nm by light emitted at 475nm. The average baseline value was subtracted from the agonist-stimulated BRET value to obtain the ΔBRET ratio.
Receptor cell surface expression HiBiT assay
Cell surface expression of D2 WT and mutants were determined using Promega’s NanoGlo® HiBiT Extracellular Detection System. HEK 293T/17 cells were transfected with 0.21 μg HiBiT-tagged D2 and incubated overnight. The next day, cells were added at 100,000 cells per well in culture medium without phenol-red into 96-well flat-bottomed white microplates. Nanoglo HiBiT extracellular reagent containing substrate and buffer was prepared per manufacturer’s protocol. 25 μL of reagent was added into each well to a final volume of 50 μL. Luminenscene was measured every 2.5 minutes for 20 minutes using the LUMIstar plate reader to ensure maximum, steady signal was achieved. Data was normalized to mock-transfected cells as background control.
TRUPATH assay
5×106 cells were plated into poly-D-lysine coated 96-well flat-bottomed white microplates (Greiner Bio-One) and transfected using a 6:1:1:1 ratio of receptor:Gα-RLuc8:Gβ:Gγ-GFP2 (0.4μg per 1 ratio). A combination of GαoA-RLuc8, Gβ3 and Gγ8-GFP2, or Gαi1-RLuc8, Gβ3 and Gγ9-GFP2 was used. The next day, culture media was removed and replaced immediately with 80μL of assay buffer (Hank’s balanced salt solution (HBSS) + 20mM HEPES + 1mM CaCl2 + 1mM MgCl2, pH 7.4), and 10μL of freshly prepared 50μM coelenterazine 400a (Nanolight Technologies). After a 5 minutes incubation period, cells were treated with 10μL of ligand addition. Plates were read using a LUMIstar plate reader (BMG Labtech), at integration times of 1 second per well. Plates were read at 1-minute interval, over 10 minutes. Measurements over the read was averaged and used in all analyses. BRET2 rations were calculated as the ratio of the GFP2 emission (510nm) to Rluc8-coelenterazine 400a (395nm) emission.
Radioligand binding assay
7×106 cells were plated onto 150mm dish and transfected using a 1:6 ratio of receptor and polyethyleneimine as transfection reagent (10μg per 1 ratio). 48 hours later, the cells were harvested with PBS containing 2mM EDTA and centrifuged at 500g for 5 minutes. The resulting pellet was resuspended in ice-cold binding assay buffer (20mM HEPES, 100mM NaCl2, 6mM MgCl2, 1mM EGTA and 1mM EDTA, pH 7.4) and homogenised using UltraTurrax homogenizer before being centrifuged at 1000g for 10 minutes. After centrifugation, the supernatant was recentrifuged at 30000g for 60 minutes at 4°C. The resulting pellet was resuspended in assay buffer and stored at −80°C. Membrane protein concentration was determined using the BCA protein assay kit (Thermo Scientific).
The radioligand binding assay was performed using [3H]spiperone in a 96-well format. 4nM [3H]spiperone was incubated with 10μg/well protein in a total volume of 200μL binding buffer containing 100μM GppNHp and 0.1% ascorbic acid for 3 hours at −80°C. Non-specific binding was determined using 10μM haloperidol. After the incubation period, the binding reaction was terminated by rapid filtration through GF/C unifilters (PerkinElmer) using a 96-well Packard FilterMate cell harvester (PerkinElmer), followed by 3 washes with 250μL ice-cold 0.9% NaCl. After drying at 50°C, microscintillation fluid was added to the dried filters and the boud radioactivity was measured in a Packard TopCounter microplate scintillation counter.
Ligand-receptor docking and mutation design
The active state D2 homology model was created with the automated protocol from the G protein-coupled database, GPCRdb.org (Pandy-Szekeres et al., 2018), using the β1 receptor structure with isoprenaline, (PDB ID 6H7J)(Warne et al., 2019) as the main template. Available active state structures were ranked based on sequence similarity to the 7TM helices plus ECL2 and the mentioned template ranked second was selected due to the high similarity of the co-crystalized ligand to dopamine.
Incorporating isoprenaline from the template into the model and deleting the OH- and iPr-substituents gave the dopamine molecule in the D2 binding site. All molecular modelling was performed within using applications in the Schrödinger Drug Discovery Software Suite (Schrödinger Release 2019–2, Schrödinger, LLC, New York, NY, 2019). The D2 structure model was prepared for docking with the Protein Preparation Wizard using default settings. The chemical structures of the five antipsychotics were downloaded from the PubChem database (http://pubchem.ncbi.nlm.nih.gov/ – CIDs: 1093278, 208951, 11154555, 60795 and 11978813)(Kim et al., 2019) and prepared for docking with default settings in LigPrep and docked in the D2 model using the standard Induced-fit Docking Protocol. The binding site center was defined by the ligand from the main template and the ligand length was set to ≦ 18 Å and XP precision was used in the re-docking step, while all other settings were default. Among the top three ranked output poses with similar IFD scores, four of the antipsychotics produced two markedly different binding modes, which were selected as possible and for cariprazine only the highest-ranking pose was selected.
17 mutations in 11 sequence positions were designed based on the binding modes obtained from the homology model and the induced fit docking. Residue positions predicted to, directly or indirectly, influence ligand binding were selected and mutant amino acids were chosen to remove/introduce interactions or change the shape, size or property of the binding site as described in Table S4.
Quantification and Statistical Analysis
Data Analysis
The maximum BRET amplitude and the rate constants (1/τ) of activation phase were obtained by fitting a single exponential curve to the traces with Clampfit Ver. 10.3 software (Molecular Devices).
Dose response curves were analyzed using Prism 8.4 (GraphPad Software Inc.). The results were fitted using the following three parameter equation:
Where Top and Bottom represent the maximal and minimal asymptote of the concentration response curve, [A] represents the concentration of agonist and EC50 is the concentration of agonist required to elicit a response halfway between top and bottom. To exclude the impact of system and observational bias effects on the observed agonism of each agonist, the 1/τ activation rate constant of reference agonist, dopamine was normalized to the 1/τ activation rate constant of each agonist. Then the ratio was normalized to 1/τ of reference pathway, GαoA activation from 1/τ of the pathways to yield 1/τ activation rate constant ratio. To calculate bias factor of D2 mutants, the 1/τ of GαoA pathway was subtracted by 1/τ of Gαi1 pathway. IC50 values obtained from the radioligand inhibition curves were converted to Ki values using the Cheng and Prusoff equation.
Statistical analysis
One-way ANOVA followed by Tukey’s post hoc test was used for comparing results and determining statistical significance. The statistical analysis performed using GraphPad Prism Ver 8.4.2. Statistical details of the experiments can be found in the figure legend.
Supplementary Material
KEY RESOURCES TABLE.
| REAGENT or RESOURCE | SOURCE | IDENTIFIER |
|---|---|---|
| Chemicals, peptides, and recombinant proteins | ||
| Dulbecco’s modified Eagle’s medium | Thermo Fisher Scientific | Cat #11965–092 |
| Fetal bovine serum | Genesse Scientific | Cat #25–550 |
| Sodium pyruvate | Thermo Fisher Scientific | Cat #11360–070 |
| MEM non-essential amino acids | Thermo Fisher Scientific | Cat #11140–050 |
| Penicillin-streptomycin | Thermo Fisher Scientific | Cat# 15140–122 |
| Matrigel | Corning | Cat# 356230 |
| Lipofectamine LTX and Plus reagent | Thermo Fisher Scientific | Cat# 15338–100 |
| Dulbecco’s phosphate-buffered saline | MilliporeSigma | Cat# D5652 |
| Hank’s balanced salt solution | Thermo Fisher Scientific | Cat# 14175053 |
| Guanosine 5′-[β,γ-imido]triphosphate trisodium salt hydrate | Sigma-Aldrich | Cat# 3655 |
| Dopamine hydrochloride | MilliporeSigma | Cat# H8502 |
| Aripiprazole | Cayman Chemicals | Cat# 19989 |
| Cariprazine | Cayman Chemicals | Cat# 31446 |
| Brexpiprazole | Cayman Chemicals | Cat# 22906 |
| MLS1547 | Sigma-Aldrich | Cat# SML1331 |
| Bifeprunox mesylate | Sigma-Aldrich | Cat# SML1670 |
| Haloperidol | Sigma-Aldrich | Cat# H1512 |
| Critical commercial assays | ||
| Nano-Glo Luciferase Assay substrate (furimazine) | Promega | Cat# N1120 |
| Nano-Glo HiBiT Extracellular Dectection System | Promega | Cat# N2420 |
| [3H]Spiperone | Perkin Elmer | Cat# NET1187250UC |
| Coelentrazine 400a | Cayman Chemicals | Cat# 16157 |
| Deposited data | ||
| β1 structure | (Warne et al., 2019) | PDB ID 6H7J |
| Aripiprazole chemical structure | (Kim et al., 2019) | CID: 60795 |
| Cariprazine chemical structure | (Kim et al., 2019) | CID: 11154555 |
| Brexpiprazole chemical structure | (Kim et al., 2019) | CID: 11978813 |
| MLS1547 chemical structure | (Kim et al., 2019) | CID: 1093278 |
| Bifeprunox mesylate chemical structure | (Kim et al., 2019) | CID: 208951 |
| Experimental models: Cell lines | ||
| HEK293T/17 | ATCC | ATCC: CRL-11268 |
| Recombinant DNA | ||
| Plasmid: Flag-D2R | Dr Abraham Kovoor, Univesity of Rhode Island | N/A |
| Plasmid: GαoA | Dr Hiroshi Itoh, Nara Institute of Science and Technology | N/A |
| Plasmid: GαoB | cDNA resource center | Cat# GNA0OB0000 |
| Plasmid: Gαi1 | Dr Hiroshi Itoh | N/A |
| Plasmid: Gαi2 | Dr Hiroshi Itoh | N/A |
| Plasmid: Gαi3 | Dr Hiroshi Itoh | N/A |
| Plasmid: Gαz | cDNA resource center | Cat# GNA0Z00000 |
| Plasmid: Venus-156-239-Gβ1 | (Hollins et al., 2009) | N/A |
| Plasmid: Venus-1-155-Gγ2 | (Hollins et al., 2009) | N/A |
| Plasmid: masGRK3ct-Nluc-HA | (Gulati et al., 2018) | N/A |
| PTX-S1 | (Raveh et al., 2010) | N/A |
| Plasmid: HiBiT-D2R | This paper | N/A |
| Plasmid: HiBiT-D2R W100A | This paper | N/A |
| Plasmid: HiBiT-D2R W100L | This paper | N/A |
| Plasmid: HiBiT-D2R F110A | This paper | N/A |
| Plasmid: HiBiT-D2R F110L | This paper | N/A |
| Plasmid: HiBiT-D2R V115I | This paper | N/A |
| Plasmid: HiBiT-D2R C118S | This paper | N/A |
| Plasmid: HiBiT-D2R C118V | This paper | N/A |
| Plasmid: HiBiT-D2R I184F | This paper | N/A |
| Plasmid: HiBiT-D2R I184S | This paper | N/A |
| Plasmid: HiBiT-D2R S193A | This paper | N/A |
| Plasmid: HiBiT-D2R S197A | This paper | N/A |
| Plasmid: HiBiT-D2R H393N | This paper | N/A |
| Plasmid: HiBiT-D2R Y408A | This paper | N/A |
| Plasmid: HiBiT-D2R Y408F | This paper | N/A |
| Plasmid: HiBiT-D2R Y408V | This paper | N/A |
| Plasmid: HiBiT-D2R T412V | This paper | N/A |
| Plasmid: HiBiT-D2R Y416W | This paper | N/A |
| Plasmid: HiBiT-D2R V154I | This paper | N/A |
| Plasmid: HiBiT-D2R S311C | This paper | N/A |
| Plasmid: Gαi1-RLuc8 | (Olsen et al., 2020) | Cat# 1000000163 |
| Plasmid: GαoA-RLuc8 | (Olsen et al., 2020) | Cat# 1000000163 |
| Plasmid: Gβ3 | (Olsen et al., 2020) | Cat# 1000000163 |
| Plasmid: Gγ8-GFP2 | (Olsen et al., 2020) | Cat# 1000000163 |
| Plasmid: Gγ9-GFP2 | (Olsen et al., 2020) | Cat# 1000000163 |
| Software and algorithms | ||
| GraphPad Prism8.4.2 | GraphPad software | https://www.graphpad.com/ |
| Clampfit 10.3 | Molecular Devices | http://www.moleculardevices.com/products/software/pclamp.html |
| Microsoft Excel | Microsoft | https://www.microsoft.com/en-ww/microsoft-365/excel |
| Schrödinger Drug Discovery Software Suite | Schrödinger, LLC | Version 2019–2 |
| PyMOL | Schrödinger, LLC | Version 2.4 |
| Other | ||
| GPCRdb | https://gpcrdb.org/ | N/A |
| PubChem | http://pubchem.ncbi.nlm.nih.gov | N/A |
SIGNIFICANCE.
The dopamine D2 receptor (D2) is an extensively studied prototype of G protein-coupled receptors (GPCRs) and is a prominent target for the development of antipsychotic drugs. Although widely used in the clinic, antipsychotics exhibit a range of the poorly explained pharmacological effects while sharing a common property: partial agonism at D2. The variability of D2 properties require their empirical prescription and limit the development of precise interventions. Explaining how different drugs with common pharmacological properties elicit different effects also presents a conundrum in basic receptor biology. The discovery of drug-induced functional discrimination among G proteins activated by the D2 variants reported here should prompt the design of precise pharmacological tools and personalized prescriptions for the treatment of schizophrenia.
Highlights.
The D2 dopamine receptor differentially activates various Gαi/o proteins
Dopamine and antipsychotic drugs produce distinct Gα activation profiles
Rationally designed mutations in D2 can override G protein biases
Genetic variants in D2 alter its G protein preferences
ACKNOWLEDGEMENTS
This work was supported by NIH grants DA036596 and MH105482 to K.A.M; the Lundbeck Foundation R313-2019-526 and the Novo Nordisk Foundation NNF18OC0031226 to D.E.G.; the Lundbeck Foundation R278-2018-180 to H.B.-O. and A.S.H.; and the Carlsberg Foundation CF20-0248 to H.B.-O.
Footnotes
DECLARATIONS OF INTEREST
The authors declare no competing interests.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
REFERENCES
- Alexander SPH, Christopoulos A, Davenport AP, Kelly E, Mathie A, Peters JA, Veale EL, Armstrong JF, Faccenda E, Harding SD, et al. (2019). THE CONCISE GUIDE TO PHARMACOLOGY 2019/20: G protein-coupled receptors. Br J Pharmacol 176 Suppl 1, S21–S141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Allen JA, Yost JM, Setola V, Chen X, Sassano MF, Chen M, Peterson S, Yadav PN, Huang XP, Feng B, et al. (2011). Discovery of beta-arrestin-biased dopamine D2 ligands for probing signal transduction pathways essential for antipsychotic efficacy. Proc Natl Acad Sci U S A 108, 18488–18493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anderson A, Masuho I, Marron Fernandez de Velasco E, Nakano A, Birnbaumer L, Martemyanov KA, and Wickman K (2020). GPCR-dependent biasing of GIRK channel signaling dynamics by RGS6 in mouse sinoatrial nodal cells. Proc Natl Acad Sci U S A 117, 14522–14531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arinami T, Itokawa M, Aoki J, Shibuya H, Ookubo Y, Atsushi I, Katsuya O, Shimizu H, Hamaguchi H, and Toru M (1996). Further association study on dopamine D2 receptor variant S311C in schizophrenia and affective disorders. Am J Med Genet 67, 133–138. [DOI] [PubMed] [Google Scholar]
- Beaulieu JM, Espinoza S, and Gainetdinov RR (2015). Dopamine receptors - IUPHAR Review 13. Br J Pharmacol 172, 1–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beaulieu JM, and Gainetdinov RR (2011). The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol Rev 63, 182–217. [DOI] [PubMed] [Google Scholar]
- Becker G, Bolbos R, Costes N, Redoute J, Newman-Tancredi A, and Zimmer L (2016). Selective serotonin 5-HT1A receptor biased agonists elicitdistinct brain activation patterns: a pharmacoMRI study. Sci Rep 6, 26633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonifazi A, Yano H, Guerrero AM, Kumar V, Hoffman AF, Lupica CR, Shi L, and Newman AH (2019). Novel and Potent Dopamine D2 Receptor Go-Protein Biased Agonists. ACS Pharmacol Transl Sci 2, 52–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chemel BR, Bonner LA, Watts VJ, and Nichols DE (2012). Ligand-specific roles for transmembrane 5 serine residues in the binding and efficacy of dopamine D(1) receptor catechol agonists. Mol Pharmacol 81, 729–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen X, McCorvy JD, Fischer MG, Butler KV, Shen Y, Roth BL, and Jin J (2016). Discovery of G Protein-Biased D2 Dopamine Receptor Partial Agonists. J Med Chem 59, 10601–10618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cravchik A, Sibley DR, and Gejman PV (1996). Functional analysis of the human D2 dopamine receptor missense variants. J Biol Chem 271, 26013–26017. [DOI] [PubMed] [Google Scholar]
- Cummings DF, Ericksen SS, Goetz A, and Schetz JA (2010). Transmembrane segment five serines of the D4 dopamine receptor uniquely influence the interactions of dopamine, norepinephrine, and Ro10–4548. J Pharmacol Exp Ther 333, 682–695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dessauer CW, Watts VJ, Ostrom RS, Conti M, Dove S, and Seifert R (2017). International Union of Basic and Clinical Pharmacology. CI. Structures and Small Molecule Modulators of Mammalian Adenylyl Cyclases. Pharmacol Rev 69, 93–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fan L, Tan L, Chen Z, Qi J, Nie F, Luo Z, Cheng J, and Wang S (2020). Haloperidol bound D2 dopamine receptor structure inspired the discovery of subtype selective ligands. Nat Commun 11, 1074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fields TA, and Casey PJ (1997). Signalling functions and biochemical properties of pertussis toxin-resistant G-proteins. Biochem J 321, 561–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flock T, Ravarani CNJ, Sun D, Venkatakrishnan AJ, Kayikci M, Tate CG, Veprintsev DB, and Babu MM (2015). Universal allosteric mechanism for Galpha activation by GPCRs. Nature 524, 173–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fowler JC, Bhattacharya S, Urban JD, Vaidehi N, and Mailman RB (2012). Receptor conformations involved in dopamine D(2L) receptor functional selectivity induced by selected transmembrane-5 serine mutations. Mol Pharmacol 81, 820–831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Free RB, Chun LS, Moritz AE, Miller BN, Doyle TB, Conroy JL, Padron A, Meade JA, Xiao J, Hu X, et al. (2014). Discovery and characterization of a G protein-biased agonist that inhibits beta-arrestin recruitment to the D2 dopamine receptor. Mol Pharmacol 86, 96–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gillis A, Gondin AB, Kliewer A, Sanchez J, Lim HD, Alamein C, Manandhar P, Santiago M, Fritzwanker S, Schmiedel F, et al. (2020). Low intrinsic efficacy for G protein activation can explain the improved side effect profiles of new opioid agonists. Sci Signal 13, 1–18. [DOI] [PubMed] [Google Scholar]
- Ginovart N, and Kapur S (2012). Role of dopamine D(2) receptors for antipsychotic activity. Handb Exp Pharmacol, 27–52. [DOI] [PubMed] [Google Scholar]
- Glatt SJ, and Jonsson EG (2006). The Cys allele of the DRD2 Ser311Cys polymorphism has a dominant effect on risk for schizophrenia: evidence from fixed- and random-effects meta-analyses. Am J Med Genet B Neuropsychiatr Genet 141B, 149–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gulati S, Jin H, Masuho I, Orban T, Cai Y, Pardon E, Martemyanov KA, Kiser PD, Stewart PL, Ford CP, et al. (2018). Targeting G protein-coupled receptor signaling at the G protein level with a selective nanobody inhibitor. Nat Commun 9, 1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hauser AS, Attwood MM, Rask-Andersen M, Schioth HB, and Gloriam DE (2017). Trends in GPCR drug discovery: new agents, targets and indications. Nat Rev Drug Discov 16, 829–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hauser AS, Chavali S, Masuho I, Jahn LJ, Martemyanov KA, Gloriam DE, and Babu MM (2018). Pharmacogenomics of GPCR Drug Targets. Cell 172, 41–54 e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hermans E (2003). Biochemical and pharmacological control of the multiplicity of coupling at G-protein-coupled receptors. Pharmacology & Therapeutics 99, 25–44. [DOI] [PubMed] [Google Scholar]
- Ho MKC, and Wong YH (2001). Gz signaling: emerging divergence from Gi signaling. Oncogene 20, 1615–1625. [DOI] [PubMed] [Google Scholar]
- Hoffmann C, Zurn A, Bunemann M, and Lohse MJ (2008). Conformational changes in G-protein-coupled receptors-the quest for functionally selective conformations is open. Br J Pharmacol 153 Suppl 1, S358–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hollins B, Kuravi S, Digby GJ, and Lambert NA (2009). The c-terminus of GRK3 indicates rapid dissociation of G protein heterotrimers. Cell Signal 21, 1015–1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Im D, Inoue A, Fujiwara T, Nakane T, Yamanaka Y, Uemura T, Mori C, Shiimura Y, Kimura KT, Asada H, et al. (2020). Structure of the dopamine D2 receptor in complex with the antipsychotic drug spiperone. Nat Commun 11, 6442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Inoue A, Raimondi F, Kadji FMN, Singh G, Kishi T, Uwamizu A, Ono Y, Shinjo Y, Ishida S, Arang N, et al. (2019). Illuminating G-Protein-Coupling Selectivity of GPCRs. Cell 177, 1933–1947 e1925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Insel PA, Sriram K, Gorr MW, Wiley SZ, Michkov A, Salmeron C, and Chinn AM (2019). GPCRomics: An Approach to Discover GPCR Drug Targets. Trends Pharmacol Sci 40, 378–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Insel PA, Tang CM, Hahntow I, and Michel MC (2007). Impact of GPCRs in clinical medicine: monogenic diseases, genetic variants and drug targets. Biochim Biophys Acta 1768, 994–1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Isberg V, de Graaf C, Bortolato A, Cherezov V, Katritch V, Marshall FH, Mordalski S, Pin JP, Stevens RC, Vriend G, et al. (2015). Generic GPCR residue numbers - aligning topology maps while minding the gaps. Trends Pharmacol Sci 36, 22–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang M, and Bajpayee NS (2009). Molecular mechanisms of go signaling. Neurosignals 17, 23–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karczewski KJ, Francioli LC, Tiao G, Cummings BB, Alfoldi J, Wang Q, Collins RL, Laricchia KM, Ganna A, Birnbaum DP, et al. (2020). The mutational constraint spectrum quantified from variation in 141,456 humans. Nature 581, 434–443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kenakin T (2011). Functional selectivity and biased receptor signaling. J Pharmacol Exp Ther 336, 296–302. [DOI] [PubMed] [Google Scholar]
- Kenakin T (2019). Biased Receptor Signaling in Drug Discovery. Pharmacol Rev 71, 267–315. [DOI] [PubMed] [Google Scholar]
- Kenakin T, and Christopoulos A (2013). Signalling bias in new drug discovery: detection, quantification and therapeutic impact. Nat Rev Drug Discov 12, 205–216. [DOI] [PubMed] [Google Scholar]
- Kenakin T, and Miller LJ (2010). Seven transmembrane receptors as shapeshifting proteins: the impact of allosteric modulation and functional selectivity on new drug discovery. Pharmacol Rev 62, 265–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kenakin T, Watson C, Muniz-Medina V, Christopoulos A, and Novick S (2012). A simple method for quantifying functional selectivity and agonist bias. ACS Chem Neurosci 3, 193–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim S, Chen J, Cheng T, Gindulyte A, He J, He S, Li Q, Shoemaker BA, Thiessen PA, Yu B, et al. (2019). PubChem 2019 update: improved access to chemical data. Nucleic Acids Res 47, D1102–D1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klein C, Brin MF, Kramer PL, Sena-Esteves M, de Leon D, Doheny D, Bressman S, Fahn S, Breakefield XO, and Ozelius LJ (1999). Association of a missense change in the D2 dopamine receptor with myoclonus dystonia. Proc Natl Acad Sci U S A 96, 5173–5176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klein C, Gurvich N, Sena-Esteves M, Bressman S, Brin MF, Ebersole BJ, Fink S, Forsgren L, Friedman J, Grimes D, et al. (2000). Evaluation of the role of the D2 dopamine receptor in myoclonus dystonia. Ann Neurol 47, 369–373. [PubMed] [Google Scholar]
- Klein Herenbrink C, Sykes DA, Donthamsetti P, Canals M, Coudrat T, Shonberg J, Scammells PJ, Capuano B, Sexton PM, Charlton SJ, et al. (2016). The role of kinetic context in apparent biased agonism at GPCRs. Nat Commun 7, 10842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klein Herenbrink C, Verma R, Lim HD, Kopinathan A, Keen A, Shonberg J, Draper-Joyce CJ, Scammells PJ, Christopoulos A, Javitch JA, et al. (2019). Molecular Determinants of the Intrinsic Efficacy of the Antipsychotic Aripiprazole. ACS Chem Biol 14, 1780–1792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kling RC, Tschammer N, Lanig H, Clark T, and Gmeiner P (2014). Active-state model of a dopamine D2 receptor-Galphai complex stabilized by aripiprazole-type partial agonists. PLoS One 9, e100069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leck KJ, Blaha CD, Matthaei KI, Forster GL, Holgate J, and Hendry IA (2006). Gz proteins are functionally coupled to dopamine D2-like receptors in vivo. Neuropharmacology 51, 597–605. [DOI] [PubMed] [Google Scholar]
- Madras BK (2013). History of the discovery of the antipsychotic dopamine D2 receptor: a basis for the dopamine hypothesis of schizophrenia. J Hist Neurosci 22, 62–78. [DOI] [PubMed] [Google Scholar]
- Mailman RB, and Murthy V (2010). Third generation antipsychotic drugs: partial agonism or receptor functional selectivity? Curr Pharm Des 16, 488–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Masri B, Salahpour A, Didriksen M, Ghisi V, Beaulieu JM, Gainetdinov RR, and Caron MG (2008). Antagonism of dopamine D2 receptor/beta-arrestin 2 interaction is a common property of clinically effective antipsychotics. Proc Natl Acad Sci U S A 105, 13656–13661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Masuho I, Martemyanov KA, and Lambert NA (2015a). Monitoring G Protein Activation in Cells with BRET. Methods Mol Biol 1335, 107–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Masuho I, Ostrovskaya O, Kramer GM, Jones CD, Xie K, and Martemyanov KA (2015b). Distinct profiles of functional discrimination among G proteins determine the actions of G protein-coupled receptors. Sci Signal 8, ra123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mauri MC, Paletta S, Maffini M, Colassanti A, Dragogna F, Di Pace C, and Altamura AC (2014). Clinical pharmacology of atypical antipsychotics: an update. EXCLI J 13, 1163–1191. [PMC free article] [PubMed] [Google Scholar]
- Michel MC, and Charlton SJ (2018). Biased Agonism in Drug Discovery-Is It Too Soon to Choose a Path? Mol Pharmacol 93, 259–265. [DOI] [PubMed] [Google Scholar]
- Milligan G, and Kostenis E (2006). Heterotrimeric G-proteins: a short history. Br J Pharmacol 147 Suppl 1, S46–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miyamoto S, Miyake N, Jarskog LF, Fleischhacker WW, and Lieberman JA (2012). Pharmacological treatment of schizophrenia: a critical review of the pharmacology and clinical effects of current and future therapeutic agents. Mol Psychiatry 17, 1206–1227. [DOI] [PubMed] [Google Scholar]
- Neve KA, Seamans JK, and Trantham-Davidson H (2004). Dopamine Receptor Signaling. Journal of Receptor and Signal Transduction Research 24, 165–205. [DOI] [PubMed] [Google Scholar]
- Olsen RHJ, DiBerto JF, English JG, Glaudin AM, Krumm BE, Slocum ST, Che T, Gavin AC, McCorvy JD, Roth BL, et al. (2020). TRUPATH, an open-source biosensor platform for interrogating the GPCR transducerome. Nat Chem Biol. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pandy-Szekeres G, Munk C, Tsonkov TM, Mordalski S, Harpsoe K, Hauser AS, Bojarski AJ, and Gloriam DE (2018). GPCRdb in 2018: adding GPCR structure models and ligands. Nucleic Acids Res 46, D440–D446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peng Y, McCorvy JD, Harpsoe K, Lansu K, Yuan S, Popov P, Qu L, Pu M, Che T, Nikolajsen LF, et al. (2018). 5-HT2C Receptor Structures Reveal the Structural Basis of GPCR Polypharmacology. Cell 172, 719–730 e714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rajagopal S, Rajagopal K, and Lefkowitz RJ (2010). Teaching old receptors new tricks: biasing seven-transmembrane receptors. Nat Rev Drug Discov 9, 373–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Randáková A, Nelic D, Ungerová D, Nwokeye P, Su Q, Doležal V, El-Fakahany EE, Boulos J, and Jakubík J (2020). Novel M2-Selective, Gi-Biased Agonists of Muscarinic Acetylcholine Receptors. Br J Pharmacol 177, 2073–2089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raveh A, Cooper A, Guy-David L, and Reuveny E (2010). Nonenzymatic rapid control of GIRK channel function by a G protein-coupled receptor kinase. Cell 143, 750–760. [DOI] [PubMed] [Google Scholar]
- Reiter E, Ahn S, Shukla AK, and Lefkowitz RJ (2012). Molecular mechanism of beta-arrestin-biased agonism at seven-transmembrane receptors. Annu Rev Pharmacol Toxicol 52, 179–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ryba DM, Li J, Cowan CL, Russell B, Wolska BM, and Solaro RJ (2017). Long-Term Biased β-Arrestin Signaling Improves Cardiac Structure and Function in Dilated Cardiomyopathy. Circulation 135, 1056–1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanchez-Soto M, Bonifazi A, Cai NS, Ellenberger MP, Newman AH, Ferre S, and Yano H (2016). Evidence for Noncanonical Neurotransmitter Activation: Norepinephrine as a Dopamine D2-Like Receptor Agonist. Mol Pharmacol 89, 457–466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santos R, Ursu O, Gaulton A, Bento AP, Donadi RS, Bologa CG, Karlsson A, Al-Lazikani B, Hersey A, Oprea TI, et al. (2017). A comprehensive map of molecular drug targets. Nat Rev Drug Discov 16, 19–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sartania N, and Strange PG (1999). Role of Conserved Serine Residues in the Interaction of Agonists with D3 Dopamine Receptors. J Neurochem 72. [DOI] [PubMed] [Google Scholar]
- Shenoy SK, and Lefkowitz RJ (2011). beta-Arrestin-mediated receptor trafficking and signal transduction. Trends Pharmacol Sci 32, 521–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shukla AK, Xiao K, and Lefkowitz RJ (2011). Emerging paradigms of beta-arrestin-dependent seven transmembrane receptor signaling. Trends Biochem Sci 36, 457–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singla N, Minkowitz HS, Soergel DG, Burt DA, Subach RA, Salamea MY, Fossler MJ, and Skobieranda F (2017). A randomized, Phase IIb study investigating oliceridine (TRV130), a novel micro-receptor G-protein pathway selective (mu-GPS) modulator, for the management of moderate to severe acute pain following abdominoplasty. J Pain Res 10, 2413–2424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith JS, Lefkowitz RJ, and Rajagopal S (2018). Biased signalling: from simple switches to allosteric microprocessors. Nat Rev Drug Discov 17, 243–260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spongier D, Waeber C, Pantaloni C, Holsboer F, Bockaert J, Seeburgt PH, and Journot L (1993). Differential signal transduction by five splice variants of the PACAP receptor. Nature 365, 170–175. [DOI] [PubMed] [Google Scholar]
- Sunahara R, Dessauer CW, and Gilman AG (1996). Complexity and diversity of mammalian adenylyl cyclases. Annu Rev Pharmacol Toxicol 36, 461–480. [DOI] [PubMed] [Google Scholar]
- Syrovatkina V, Alegre KO, Dey R, and Huang XY (2016). Regulation, Signaling, and Physiological Functions of G-Proteins. J Mol Biol 428, 3850–3868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Szabo M, Klein Herenbrink C, Christopoulos A, Lane JR, and Capuano B (2014). Structure-activity relationships of privileged structures lead to the discovery of novel biased ligands at the dopamine D(2) receptor. J Med Chem 57, 4924–4939. [DOI] [PubMed] [Google Scholar]
- Tschammer N, Bollinger S, Kenakin T, and Gmeiner P (2011). Histidine 6.55 is a major determinant of ligand-biased signaling in dopamine D2L receptor. Mol Pharmacol 79, 575–585. [DOI] [PubMed] [Google Scholar]
- Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson A, Kampf C, Sjostedt E, Asplund A, et al. (2015). Proteomics. Tissue-based map of the human proteome. Science 347, Human Protein Atlas available from http://www.proteinatlas.org. [DOI] [PubMed] [Google Scholar]
- Urban JD, Clarke WP, von Zastrow M, Nichols DE, Kobilka B, Weinstein H, Javitch JA, Roth BL, Christopoulos A, Sexton PM, et al. (2007). Functional selectivity and classical concepts of quantitative pharmacology. J Pharmacol Exp Ther 320, 1–13. [DOI] [PubMed] [Google Scholar]
- Urs NM, Peterson SM, and Caron MG (2017). New Concepts in Dopamine D2 Receptor Biased Signaling and Implications for Schizophrenia Therapy. Biol Psychiatry 81, 78–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vass M, Podlewska S, de Esch IJP, Bojarski AJ, Leurs R, Kooistra AJ, and de Graaf C (2019). Aminergic GPCR-Ligand Interactions: A Chemical and Structural Map of Receptor Mutation Data. J Med Chem 62, 3784–3839. [DOI] [PubMed] [Google Scholar]
- Violin JD, Crombie AL, Soergel DG, and Lark MW (2014). Biased ligands at G-protein-coupled receptors: promise and progress. Trends Pharmacol Sci 35, 308–316. [DOI] [PubMed] [Google Scholar]
- Viscusi ER, Webster L, Kuss M, Daniels S, Bolognese JA, Zuckerman S, Soergel DG, Subach RA, Cook E, and Skobieranda F (2016). A randomized, phase 2 study investigating TRV130, a biased ligand of the mu-opioid receptor, for the intravenous treatment of acute pain. Pain 157, 264–272. [DOI] [PubMed] [Google Scholar]
- Wacker D, Stevens RC, and Roth BL (2017). How Ligands Illuminate GPCR Molecular Pharmacology. Cell 170, 414–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang S, Che T, Levit A, Shoichet BK, Wacker D, and Roth BL (2018). Structure of the D2 dopamine receptor bound to the atypical antipsychotic drug risperidone. Nature 555, 269–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang S, Wacker D, Levit A, Che T, Betz RM, McCorvy JD, Venkatakrishnan AJ, Huang XP, Dror RO, Shoichet BK, et al. (2017). D4 dopamine receptor high-resolution structures enable the discovery of selective agonists. Science 358, 381–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Warne T, Edwards PC, Dóre AS, Leslie AGW, and Tate CG (2019). Molecular basis for high-affinity agonist binding in GPCRs. Science 364, 775–778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weichert D, Banerjee A, Hiller C, Kling RC, Hubner H, and Gmeiner P (2015). Molecular determinants of biased agonism at the dopamine D(2) receptor. J Med Chem 58, 2703–2717. [DOI] [PubMed] [Google Scholar]
- Wettschureck N, and Offermanns S (2005). Mammalian G proteins and their cell type specific functions. Physiol Rev 85, 1159–1204. [DOI] [PubMed] [Google Scholar]
- Wootten D, Christopoulos A, Marti-Solano M, Babu MM, and Sexton PM (2018). Mechanisms of signalling and biased agonism in G protein-coupled receptors. Nat Rev Mol Cell Biol 19, 638–653. [DOI] [PubMed] [Google Scholar]
- Yao J, Pan YQ, Ding M, Pang H, and Wang BJ (2015). Association between DRD2 (rs1799732 and rs1801028) and ANKK1 (rs1800497) polymorphisms and schizophrenia: a meta-analysis. Am J Med Genet B Neuropsychiatr Genet 168B, 1–13. [DOI] [PubMed] [Google Scholar]
- Yin J, Chen KM, Clark MJ, Hijazi M, Kumari P, Bai XC, Sunahara RK, Barth P, and Rosenbaum DM (2020). Structure of a D2 dopamine receptor-G-protein complex in a lipid membrane. Nature 584, 125–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhuang Y, Xu P, Mao C, Wang L, Krumm B, Zhou XE, Huang S, Liu H, Cheng X, Huang XP, et al. (2021). Structural insights into the human D1 and D2 dopamine receptor signaling complexes. Cell 184, 931–942 e918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gulati S, Jin H, Masuho I, Orban T, Cai Y, Pardon E, Martemyanov KA, Kiser PD, Stewart PL, Ford CP, et al. (2018). Targeting G protein-coupled receptor signaling at the G protein level with a selective nanobody inhibitor. Nat Commun 9, 1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hollins B, Kuravi S, Digby GJ, and Lambert NA (2009). The c-terminus of GRK3 indicates rapid dissociation of G protein heterotrimers. Cell Signal 21, 1015–1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim S, Chen J, Cheng T, Gindulyte A, He J, He S, Li Q, Shoemaker BA, Thiessen PA, Yu B, et al. (2019). PubChem 2019 update: improved access to chemical data. Nucleic Acids Res 47, D1102–D1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olsen RHJ, DiBerto JF, English JG, Glaudin AM, Krumm BE, Slocum ST, Che T, Gavin AC, McCorvy JD, Roth BL, et al. (2020). TRUPATH, an open-source biosensor platform for interrogating the GPCR transducerome. Nat Chem Biol. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raveh A, Cooper A, Guy-David L, and Reuveny E (2010). Nonenzymatic rapid control of GIRK channel function by a G protein-coupled receptor kinase. Cell 143, 750–760. [DOI] [PubMed] [Google Scholar]
- Warne T, Edwards PC, Dóre AS, Leslie AGW, and Tate CG (2019). Molecular basis for high-affinity agonist binding in GPCRs. Science 364, 775–778. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The source data for raw traces of GPCR responses used for quantitative analysis reported in this study have not been deposited in a publicly available repository because of their trivial nature and custom format they are generated in. They have been archived locally on the institutional cloud service. To request access, please contact the Lead Contact. The paper does not report any original code. Any additional information to reanalyse the data reported in this paper is available from the lead contact upon request.
