Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Feb 1.
Published in final edited form as: Neurochem Res. 2015 Dec 31;41(0):328–339. doi: 10.1007/s11064-015-1808-6

Efficacy of hybrid tetrahydrobenzo[d]thiazole based aryl piperazines D-264 and D-301 at D2 and D3 receptors

Juan Zhen 1, Tamara Antonio 1, Joanna C Jacob 2, David K Grandy 3, Maarten EA Reith 1,4,#, Aloke K Dutta 5,#, Dana E Selley 2,#
PMCID: PMC4775387  NIHMSID: NIHMS748592  PMID: 26718829

Abstract

In elucidating the role of pharmacodynamic efficacy at D3 receptors in therapeutic effectiveness of dopamine receptor agonists, the influence of study system must be understood. Here two compounds with D3 over D2 selectivity developed in our earlier work, D-264 and D-301, are compared in dopamine receptor-mediated G-protein activation in striatal regions of wild-type and D2 receptor knockout mice and in CHO cells expressing D2 or D3 receptors. In caudate-putamen of D2 knockout mice, D-301 was ~ 3-fold more efficacious than D-264 in activating G-proteins as assessed by [35S]GTP S binding; in nucleus accumbens, D-301 stimulated G-protein activation whereas D-264 did not. In contrast, the two ligands exerted similar efficacy in both regions of wild-type mice, suggesting both ligands activate D2 receptors with similar efficacy. In D2 and D3 receptor-expressing CHO cells, D-264 and D-301 appeared to act in the [35S]GTP S assay as full agonists because they produced maximal stimulation equal to dopamine. Competition for [3H]spiperone binding was then performed to determine Ki/EC50 ratios as an index of receptor reserve for each ligand. Action of D-301, but not D-264, showed receptor reserve in D3 but not in D2 receptor-expressing cells, whereas dopamine showed receptor reserve in both cell lines. G o1 is highly expressed in brain and is important in D2 -like receptor-G protein coupling. Transfection of G o1 in D3- but not D2-expressing CHO cells led to receptor reserve for D-264 without altering receptor expression levels. D-301 and dopamine exhibited receptor reserve in D3-expressing cells both with and without transfection of G o1. Altogether, these results indicate that D-301 has greater intrinsic efficacy to activate D3 receptors than D-264, whereas the two compounds act on D2 receptors with similar intrinsic efficacy. These findings also suggest caution in interpreting Emax values from functional assays in receptor-transfected cell models without accounting for receptor reserve.

Keywords: dopamine receptors, D2, D3, intrinsic efficacy, G Protein, rat tissue, [35S]GTP S binding assay, G o1

Introduction

The D2-like family of dopamine receptors is a major drug target for treatment of motor and neuropsychiatric disorders[1,2]. The D3 member of this family in particular, is associated with regulation of motivation/reward, mood, cognition and motor function, and is expressed throughout the limbic forebrain and basal ganglia, albeit at lower levels than D2 receptors [3-5]. D3 receptors are promising targets for treatment of drug abuse, schizophrenia, attention deficit hyperactivity disorders and motor diseases such as Parkinson's disease [1,6-8]. Despite the promise of therapeutic targeting of this receptor, development of ligands with high selectivity for D3 over other D2-like receptors and which possess a range of intrinsic efficacies, has been challenging. In order to elucidate the role of pharmacodynamic efficacy at D3 receptors in therapeutic effectiveness, the influence of study system, such as cell lines heterologously expressing the receptor versus native systems (e.g., striatum), must be understood.

D3 receptors are coupled to the activation of mainly Gi/o family G proteins [9,10]. G protein activation by receptors promotes exchange of GDP for GTP, which can be quantified in the GTP 35S binding assay [11,9]. It is well known that G protein-coupled receptor (GPCR) ligands can display partial agonism in this assay with brain tissue when the receptor number is low compared with available G proteins, whereas the same ligands can become full agonists in cells overexpressing GPCRs such that the receptor number is high compared with available G protein [11-17]. Even though most high efficacy agonists maximally stimulate GTP 35S binding in these cell lines, their intrinsic efficacy may vary as shown by the varying levels of receptor reserve depending on the activating ligand [12,17].

The work described here examined the intrinsic efficacy of two synthetic agonists at D2 and D3 dopamine receptors. These compounds are two related hybrid tetrahydrobenzo[d]thiazole based arylpiperazines developed in our earlier work [18,19]: D-264 and D-301. Both compounds contain the tetrahydrobenzo[d]thiazole moiety, connected through a (propyl substituted)N-ethyl linker with a piperazine biphenyl moiety (D-264) or a piperazine isoquinolin moiety (D-301) (Fig. 1). The tetrahydrobenzo[d]thiazole portion is thought to bind to the primary binding pocket of the dopamine receptor, whereas the aryl piperazine moiety likely binds an accessory binding site(s) [20]. Both compounds appeared to be full agonists relative to DA. We chose D-264 as one of the compounds for study since it not only displayed preferential D3 potency but also exhibited efficacious in vivo activity including potent neuroprotection [19,21]. D-301 was selected because the compound exhibited high potency and very high selectivity for D3 receptors [18]. Another reason for selecting these two compounds is their structural diversity in the substitution of piperazine ring of the compounds such that D-301 has an isoquinoline moiety as opposed to presence of biphenyl substitution in the case of D-264. This might potentially impact interaction at the accessory binding sites of the receptors which might lead to production of differential receptors activation.

Fig. 1.

Fig. 1

Chemical structures of D264 and D301.

Two approaches were taken in this work to study potential differences in intrinsic efficacy of D-264 and D-301. First, the ability of the compounds to stimulate GTP 35S binding was determined in assays with brain tissue from either wild-type (WT) or D2 dopamine receptor knock-out (KO) mice [22]. In WT mice, both D2 and D3 receptors can contribute to G protein activation, whereas in D2 KO mice, the D2 component has been removed. In this scenario, a difference in maximal activation between the two compounds suggests a difference in intrinsic efficacy at the level of D3 receptor-mediated G protein activation. In a second approach, cell lines expressing only D2 or D3 receptors were used to determine intrinsic efficacy by assessing ligand-stimulated GTP 35S binding and competition for [3H]spiperone binding under identical conditions. In this system, the ratio of binding Ki to G protein activation EC50 values, combined with maximal activation values (Emax), was used to evaluate the intrinsic efficacy of ligands [12,23]. Because of the reportedly greater efficiency of D3 receptor coupling to Go relative to Gi [24] and the fact that Go is highly expressed in the brain [25], experiments were conducted in dopamine receptor-expressing cells both in the absence and presence of transiently expressed G o1. The results from the two approaches combined indicate that D-301 has a greater intrinsic efficacy at D3 receptors than D-264, whereas the two compounds acted on D2 receptors with similar intrinsic efficacy.

Materials and Methods

Materials and animals

[3H]spiperone (15 Ci/mmole) and [35S]GTP S (1250 Ci/mmole) were purchased from PerkinElmer (Waltham, Massachusetts). Compounds D-264 and D301 were synthesized by us as described previously ([18], in which compound 24c is D-301, and [19], in which compound (−)-34 is D-264). Plasmid DNA (pcDNA3.1) expressing human G o1 was obtained from the cDNA Resource Center (www.cdna.org). Other chemicals were obtained from commercial sources such as Fisher (Pittsburg, PA, USA) or Sigma (St. Louis, MO, USA). Female D2R WT and KO mice on a C57/Bl6 background were bred at the Oregon Health and Science University.

Cell culture and transient transfection of G o

CHO cells stably expressing D2 or D3 human DA receptors (CHO-hD2 and CHO-hD3 cells) were the same as used in our previous work [26,27]. CHO-hD2 was cultured in Dulbecco's Modified Eagle Medium/ nutrient mixture F12 and CHO-hD3 in Dulbecco's Modified Eagle Medium, in both cases supplemented with 10% bovine calf serum, and 2 mM glutamine, at 37°C and 5% CO2. Geneticin (200 μg/mL G418) was always present in order to maintain selection pressure. Where indicated in the text, transient transfection with G o1 was achieved in both cell lines with Lipofectamine® 2000 (Qiagen, Germantown, MD, USA). In order to assess expression of G o1, 36 hr after transfection cells were scraped and resuspended in NP40 lysis buffer (50 mM Tris-HCl, 150 mM NaCl, 1% Triton, pH 8.5, containing protease inhibitor cocktail [Sigma-Aldrich] and incubated on ice for a total of 30 min with 10-sec vortexing every 10 min. After centrifugation at 20, 000 g for 15 min at 4°C, the supernatant was collected and protein concentration was determined by a Bio-Rad DC protein assay kit. For western blot analysis, equal amounts of protein (30-60 μg per lane) were loaded on 10% Tris-glycine polyacrylamide gel. Membranes were probed with rabbit anti- G o1 antibody (GNAO1, Proteintech Group, Chicago, IL) and rabbit anti-β-actin (Sigma-Aldrich). The target protein bands were visualized by enhanced chemiluminescence (ECL, Amersham Pharmacia Biotech, Buckinghamshire, England) and quantified using Image-J (downloaded from website of National Institute of Heath). The integrated density value (IDV) for the protein G o1 was normalized with that for loading control β-actin. In each experiment the ratio of normalized IDV for G o1 protein with transient transfection over that without transient transfection was calculated.

Cell-free membrane preparation

Confluent cells were gently washed with ice-cold Dulbecco's Phosphate-Buffered Saline (DPBS) (without calcium and magnesium salts) and then lysed with 3 mL ice-cold lysis buffer (50 mM Tris-HCl and 1 mM EDTA, PH =7.4). Cell lysate was transferred to a Beckman polycarbonate thick-wall centrifuge tube and centrifuged at 35,000 g for 15 minutes at 4°C. The resultant pellet was homogenized with a Brinkmann Polytron (Brinkmann Instruments, Westbury, NY) at setting 6 for 15 seconds in the above lysis buffer and centrifuged again at 35,000 g for 15 minutes at 4°C. The final pellet containing crude cell-free membranes was suspended in the following assay buffer, for CHO-hD2: 20 mM HEPES (pH = 7.4), 10 mM MgCl2, 150 mM NaCL, 0.2 mM EGTA, 0.001% BSA, 3 μM GDP; and for CHO-hD3: 20 mM HEPES (pH = 7.4), 3 mM MgCl2, 100 mM NaCL, 0.2 mM EGTA, 0.001% BSA, 6 μM GDP. The resuspended membrane preparations were kept in ice.

For mouse tissues, mice were euthanized at Oregon Health and Science University, and brains were frozen at −30°C in 2-methylbutane as previously described. Brains were shipped on dry ice by overnight express to Virginia Commonwealth University, where they were stored at −80°C until use. On the day of each assay, one brain each from D2 KO and wild-type mice were thawed, and the caudate-putamen (dorsal striatum) and nucleus accumbens (ventral striatum) were dissected on ice. The tissue was homogenized in 50 mM Tris-HCl (pH 7.4), 3 mM MgCl2 and 1 mM EGTA, pH 7.4, and centrifuged at 48,000 × g for 10 min. The supernatant was discarded, and the pellet resuspended in 50 mM Tris-HCl (pH 7.4), 3 mM MgCl2, 0.1 mM EGTA and 100 mM NaCl, the volume was normalized to obtain equal protein concentrations between samples, and membranes were pre-incubated for 10 min at 30°C with 30 mU/ml adenosine deaminase to inactivate endogenous adenosine prior to assay.

[35S]GTPγS binding to D2 and D3 DA receptors in CHO cells

Because GDP is sensitive to temperature and light, GDP was only retrieved and added to the cell suspension (3 μM unlabeled GDP for CHO-D2 and 6 μM for CHO-D3 in the assay) just before initiating the binding experiment. Different concentrations of test compounds were first pre-incubated with cell suspension for 15 minutes at 30°C. After addition of [35S]GTP S at 0.1 nM (final concentration), the binding assay was continued for another 45 minutes at 30°C. Nonspecific binding was defined with 10 μM unlabeled GTP S. Assays were terminated by filtration with ice-cold lysis buffer through a glass fiber filtermat B in the Brandel-96 cell harvester. The filtermat was dried under hot air and, after addition of 10 ml of Betaplate Scint (PerkinElmer), counted for radioactivity in a Microbeta liquid scintillation counter (PerkinElmer).

[35S]GTP S binding in mouse striatum

Membranes were prepared from dissected caudate-putamen (dorsal striatum) and nucleus accumbens (ventral striatum), and ligand-stimulated [35S]GTP S binding was conducted essentially as described[28]. Briefly, membranes (3-4 μg protein) was incubated with 30 μM GDP, 0.1 nM [35S]GTP S and varying ligand concentrations in 50 mM Tris-HCl (pH 7.4), 3 mM MgCl2, 0.1 mM EGTA, 100 mM NaCl and 3 mU/ml adenosine deaminase, 0.5 ml final volume, for 90 min at 30°C. The incubation was terminated by rapid filtration through GF/B glass fiber filters and washed three times with ice-cold 50 mM Tris-HCl (pH 7.4). Bound radioactivity was determined by liquid scintillation spectrophotometry after overnight extraction of the filters in scintillation fluid. Non-specific binding was determined in the presence of 20 μM unlabeled GTP S, and basal binding was determined in the absence of agonist. All assays were performed in triplicate, and data were analyzed as specific binding.

[3H]spiperone binding to D2 and D3 DA receptors in CHO cells

In order to have conditions identical to those used in the above functional [35S]GTP S assay, we had the same final concentrations of GDP, 3 μM for D2 and 6 μM for D3, and 0.1 nM GTP S, present in the [3H]spiperone binding assays. Various concentrations of test compounds, cell-free membranes, and [3H]spiperone (2 nM final) were incubated together for 1 h at 30°C in the assay buffer. Two μM (+)-butaclamol was used to define nonspecific binding. Assays were terminated by filtration with ice-cold 0.9% saline through a glass fiber filtermat B in the Brandel-96 cell harvester. Avoiding ligand depletion is especially critical for ultra-high affinity ligands such as [3H]spiperone in the case of D2/3 receptors. Thus, receptor concentration should be lower than the Kd of radioligand or Ki of unlabeled compound; otherwise the affinity of ligand binding to receptors would be under-estimated [29,30]. Here we use the optimized protocol of [3H]spiperone binding to DA receptors, consisting of raising the concentration of [3H]spiperone (2 nM in the current study) instead of the assay volume [30], allowing the determination of the affinity of test ligand as low as 0.01 nM.

D4 DA receptor affinity

Broad-spectrum in vitro receptor binding data for analogue D-264 were kindly provided by Dr. Bryan L. Roth (University of North Carolina at Chapel Hill, NC) as part of the National Institute of Mental Health's (NIMH) Psychoactive Drug Screening Program (PDSP), and these data included the binding affinity of D-264 for D4 DA receptors. In brief, the PDSP assesses binding of novel compounds at a wide array of cloned human G-protein coupled receptors, membrane transporters and other proteins known to be psychoactive drug targets. For detailed description of the PDSP experimental protocols, please refer to the PDSP Web site at http://pdsp.med.unc.edu/ and select the “Binding Assay” link.

Data calculations and statistical analysis

[35S]GTP S data were transformed to % stimulation (where net stimulated [35S]GTP S binding (fmol/mg) = ligand-stimulated – basal [35S]GTP S binding (fmol/mg) and basal [35S]GTP S binding was measured in the absence of agonist. Results from the mouse striatum experiments were analyzed with GraphPad Prism and fit to the 3-parameter logistic equation: E = (Emax / (1 + 10[(log EC50 − log L) × nH)), where [log L] = the log10 of the ligand concentration and nH = the Hill coefficient (slope). IC50 values in [3H]spiperone binding were fit to the one-site competitive binding model (logistic equation) with the computer program Origin (Microcal Software, Northampton, MA, USA) and converted to inhibition constants (Ki) by the Cheng–Prusoff equation [31]. The logistic equation in Origin was also used for fitting the activation of [35S]GTP S binding to CHO cells by dopamine, and is the same as the approach used above. The intrinsic efficacy was calculated from relative Emax (Emax of test ligand/Emax of full agonist, i.e., dopamine in the current study) and EC50 obtained from concentration-effect curves in the [35S]GTP S binding assay, and Ki obtained in the [3H]spiperone binding assay, as follows:

IntrinsicEfficacy=(Emax,drugEmax,dopamine)×(KiEC50+1)×12

[12,23].

The error in the Ki/EC50 ratio was computed from the propagation of the errors in the Ki and EC50 ([32], equation 1a therein); likewise these errors along with that in Emax, drug were propagated for calculating the error in intrinsic efficacy Statistical tests involving values for the Ki/EC50 ratio and intrinsic efficacy were performed by inputting the mean, SEM, and smallest group size (conservative approach) in the given data set, into the Instat program which is part of the Graphpad software; the smallest n varied between 3 and 7 depending on the data set. The accepted level of significance was 0.05. Tests were two-tailed except in the case of determining whether the Ki/EC50 ratio or intrinsic efficacy was different from unity, because receptor reserve would drive such values only in the direction of > 1 (i.e., representing a one-tailed case).

Results

D2 KO mice

To determine the contribution of D3 receptors to agonist-stimulated G-protein activation in mouse striatum by D-264 and D-301, stimulation of [35S]GTP S binding was examined in membranes prepared from caudate-putamen (Figure 2A, C) and nucleus accumbens (Figure 1B, D) of D2 receptor KO compared to WT mice. The concentration-effect curves were analyzed by non-linear regression to generate maximal stimulation (Emax) and EC50 values, as well as Hill coefficients (Table 1). Both agonists produced similar maximal stimulation of [35S]GTP S binding in caudate-putamen (58-64% stimulation) and nucleus accumbens (31-41% stimulation) of WT mice (Table 1). In D2 KO mice, however, the D-301 Emax value was significantly greater than that of D-264 (approximately 17% versus 6% stimulation, respectively) in caudate-putamen. In nucleus accumbens, only D-301 significantly stimulated [35S]GTP S binding in D2 KO mice (Emax = 11.5%), whereas D-264 did not stimulate [35S]GTP S binding above basal levels. The EC50 values of D-301 were significantly greater than those of D-264 in both regions of WT mice, although the EC50 value of D-301 was not significantly different from that of D-264 in the caudate-putamen of D2 KO mice. These results suggest that despite its generally lower potency, D-301 activates D3 receptors more efficaciously than D-264. In agreement with this interpretation, the Hill coefficients of the D-301 concentration-effect curves were significantly less than unity in both regions of WT mice, whereas those of D-264 were not, suggesting that two different binding sites (presumably D2 and D3 receptors) were contributing to the stimulation by this ligand. Nonetheless, Hill coefficients for both D-264 and D-301 were significantly greater in the caudate-putamen of D2 KO compared to WT mice, suggesting that D3 receptors also contribute slightly to the stimulation by D-264 in this region. Moreover, the Hill coefficients of D-301 in both regions of D2 KO mice were not significantly lower than unity, in accordance with the response being purely D3-mediated. Altogether, these results suggest that in WT mice, D3 receptor-mediated G-protein activation had a greater influence on the concentration-effect curves of the higher efficacy D3 ligand, D-301, than on those of D-264, which appeared to be a lower efficacy ligand at D3 receptors.

Fig. 2.

Fig. 2

Stimulation by D264 and D301 of [35S]GTPγS binding to membranes prepared from mouse -WT(■) and D2R-KO (□) -caudate-putamen and Nucleus accumbens. Points are shown as means and standard error.

Table 1.

Emax and EC50 values from agonist-stimulated [35S]GTP S binding in caudate-putamen (CPu) and nucleus accumbens (NAc) of D2R WT and KO mice

D-264 D-301
CPu Emax (%) EC50 (nM) nH Emax (%) EC50 (nM) nH
D2R WT 58.2 ± 4.6 11.9 ± 4.9 0.73 ± 0.12 63.6 ± 6.0 39.1 ± 9.9$ 0.56 ± 0.09#
D2R KO 5.7 ± 0.2** 3.8 ± 1.8 1.23 ± 0.14* 16.8 ± 1.4**$ 50.5 ± 19.9 1.36 ± 0.29*
NAc Emax (%) EC50 (nM) nH Emax (%) EC50 (nM) nH
D2R WT 30.7 ± 5.1 39.4 ± 22.3 0.94 ± 0.12 41.0 ± 5.8 369.5 ± 157.6$ 0.66 ± 0.04#
D2R KO N.D. N.D. N.D. 11.5 ± 2.8* 42.6 ± 25.8 0.85 ± 0.10

Data are mean values ± SEM (n = 3-4). N.D., not determined because there was no significant stimulation by this agonist.

*

p < 0.05

**

p < 0.01 different from the corresponding value in WT mice.

$

p < 0.05 different from the D-264 value within the same genotype.

#

p < 0.05 different from unity.

Cells heterologously expressing D2 or D3 receptors

In the GTP 35S binding assay, D-264 and D-301 appeared to act as full agonists at D2 and D3 receptors expressed in CHO-cells with relative Emax values close to 100% of the maximal activation by DA (top half of Tables 2a and 2b). As observed previously [18,19,27], both compounds were D3-selective, with EC50 values more than an order of magnitude lower at D3 than D2 receptors (please note that in above [18] D-301 = compound 24c). Such selectivity was also observed for the competition of these compounds for [3H]spiperone binding (see Ki in top half of Tables 2a and 2b), as observed previously under somewhat different assay conditions [18,27]. Hill coefficients for binding and concentration-effect curves with all three agonists, in cells expressing either D2 or D3 receptors, were generally not different from unity (not shown). The one exception was the D-264 competition-binding curve in D2 receptor-expressing cells, which had a Hill coefficient of 0.66 ± 0.12.

Table 2.

Functional parameters (Emax and EC50) obtained in [35S] GTP S assay and binding parameter (Ki) in [3H]spiperone assay in CHO-D2 (a) and CHO-D3 (b) without and with Go present from transient transfection. Results are shown as mean ± S.E. averaged from 3-8 independent experiments. Ratios of Ki over EC50 and intrinsic efficacies were computed as described in Materials and Methods.

Table 2a
CHO-D2
Compound Emax (% DA) EC50 (nM) Ki (nM) Ki/EC50 Intr. Effic.
D-264 104±1.5 39.0±4.6 44.7±6.4 1.14±0.21 1.11±0.11
D-301 97.7±1.6 62.6±7.8 108±13 1.73±0.30 1.33±0.15
DA 100 363±32 3,074±340 8.47±1.20$ 4.73±0.60$
CHO-D2 with exogenous G o1 transient transfection
D-264 92.4±9.8 56±14.5 125±27.6* 2.23±0.76 1.49±0.39
D-301 101±6 31.9±3.6* 52.9±11.8* 1.66±0.41 1.34±0.22
DA 100 199±53 1922±439 9.66±3.39 5.33±1.69
Table 2b
CHO-D3
Compound Emax (% DA) EC50 (nM) Ki (nM) Ki/EC50 Intr. Effic.
D-264 93.0±4.2 2.34±0.34 2.82±0.62 1.21±0.32 1.03±0.15
D-301 107±4.4 1.01±0.23 2.19±0.33 2.17±0.59$ 1.70±0.32$
DA 100 7.51±0.29 235±20 31.3±2.9$ 16.2±1.5$
CHO-D3 with exogenous G o1 transient transfectio
D-264 75.7±5.83* 1.31±0.08* 4.00±0.61 3.05±0.50*$ 1.53±0.22
D-301 102±2.3 1.22±0.11 4.72±0.77* 3.87±0.72$ 2.48±0.37$
DA 100 11.6±1.78 70.4±3.41* 6.07±0.98*$ 3.53±0.49*$
*

P< 0.05 compared with absence of G o1 in same cell line (Student's t-test);

$

P<0.05 compared with unity (one-sample Student's t-test).;

Because the conditions for the GTP 35S and [3H]spiperone binding assays were identical in the current experiments, the ratio Ki/EC50 can be used as an estimate of receptor reserve, with ratios > 1 indicating receptor reserve [12,23]. The Ki/EC50 ratio is proportional to intrinsic efficacy when Emax of a ligand / Emax full agonist is ~ 1, as seen in formula (1) (Methods). For the agonists D-264 and D-301 studied here, only D-301 showed a Ki/EC50 ratio statistically greater than unity (as did DA itself) at D3 but not at D2 receptors, with intrinsic efficacies statistically greater than unity in the same cases (top halves of Tables 2a and 2b).

Because G o is highly expressed in the brain [25], but not CHO cells [33], and previous reports emphasize the importance of G o in D3 receptor-G-protein coupling[9,34-37], G o1 was transiently co-expressed in CHO cells expressing either D2 or D3. Although our CHO cell lines were found to express endogenous G o1, detectable with GNAO1 antibody (Fig. 3 bottom panel), transient transfection with exogenous G o1 approximately doubled the total G o1 expression (Fig. 3 top panel). The presence of exogenous G o1 did not affect the observed KD or Bmax values of [3H]spiperone binding (Table 3). In the case of D2, exogenous G o1 affected some values of functional and binding parameters, but this did not result in significant changes in intrinsic efficacy as compared with the absence of exogenous G o (Table 2a). Although the intrinsic efficacy values for both D-264 and D-301 with G o1 appeared slightly elevated above unity (~1.4-fold), there was no statistically significant elevation over unity at D2. In contrast, the changes observed at D3 with G o transfection led to an intrinsic efficacy significantly higher than unity for D-301 (2.48) but not D-264 (1.53) (Table 2b). Dopamine itself had a higher than unity intrinsic activity both in the absence and presence of exogenous G o1 at D3. Hill coefficients (not shown) were not different from unity in any of the binding or concentration-effect curves with all three ligands in either D2 or D3 receptor-expressing cells after co-transfection of G o.

Fig. 3.

Fig. 3

Expression of G o1 protein after transient transfection with exogenous G o1 of CHO cells stably expressing D2 and D3. Cell lystates of CHO-D2 and CHO-D3 with and without transient transfection were resolved on reducing 10% SDS-PAGE gels and subjected to western analysis. Data were averaged from 3 independent experiments and variation is shown as SEM. Increases upon transfection were 2.31 ± 0.32-fold and 1.80 ± 0.23-fold for D2 and D3 cells, respectively. **, P<0.005, *, P< 0.01 (compared with unity, one sample Student's t-test).

Table 3.

Saturation analysis of [3H]spiperone binding (Kd and Bmax) in CHO-D2/CHO-D3 with and without transient transfection of Gαo1. Results are shown as mean ± SE from 4 independent experiments.

CHO-D2 CHO-D3
Without exogenous G o1 with exogenous G o1 Without exogenous G o1 with exogenous G o1
KD, nM Bmax, pmoles/mg KD, nM Bmax, pmoles/mg KD, nM Bmax, pmoles/mg KD, nM Bmax, pmoles/mg
0.038±0.004 0.640±0.13 0.040±0.005 0.930±0.26 0.277±0.03 6.76±1.32 0.321±0.02 7.33±0.55

Discussion

The present study revealed intrinsic efficacy differences for D3 receptor-mediated G protein activation by two synthetic D3-selective ligands, D-264 and D-301, that were both previously reported to be full agonists relative to the endogenous ligand dopamine in cell lines heterologously expressing D2 and D3 receptors [18,19]. Results in D2 KO mice revealed differences in Emax values of ligand-stimulated [35S]GTP S binding in caudate-putamen, such that D-301 was nearly three-fold more efficacious to activate G proteins in this region than D-264. In nucleus accumbens of D2 KO mice, D-301 produced significant G protein activation with an Emax value that was similar to the maximal stimulation by this ligand in caudate-putamen, whereas no significant stimulation of [35S]GTP S binding was detected with D-264 in nucleus accumbens. In contrast, Emax values of the two ligands did not differ significantly in either region of WT mice. These results indicate that D-301 possesses greater intrinsic efficacy to activate D3 receptors than D-264 in mouse striatum, whereas the two ligands have similar efficacy to activate D2 receptors. In agreement, Hill coefficients of the D-301 concentration-effect curves were statistically less than unity in both striatal regions of WT but not D2 KO mice, suggesting the contribution of both D2 and D3 receptors to the overall D2-like receptor-mediated G protein signal with this ligand. In contrast, Hill coefficients for D-264 were not statistically different from unity in either region, suggesting that G protein activation by this ligand was mediated predominantly by a single receptor type, presumably the D2 receptor. In the case of D-264 in caudate-putamen, the contribution of the D3 receptor was apparently too low to reduce the Hill number significantly below unity.

The finding that D-264 produced small but significant stimulation of [35S]GTP S binding in caudate-putamen but not in nucleus accumbens was somewhat unexpected, because D3 receptor levels are reportedly greater in the latter region in rats [4]. It is possible that our dissections also included portions of the Islands of Calleja, where D3 receptors are also highly expressed [4]. Moreover, autoradiographic studies in post-mortem human brain also showed relatively high D3 receptor levels in the more ventral aspects of both the caudate and putamen [5]. It is also possible that D3 receptors are more efficiently coupled to G protein activation in the caudate-putamen than nucleus accumbens, despite their lower expression level in the former region. Finally, it is possible that Gi/o protein-coupled D4 receptors contributed to the non-D2 mediated [35S]GTP S signal in both caudate-putamen and nucleus accumbens, as D4 receptor expression has been reported in the range of 20-25% of that of D2 receptors in these striatal regions, with even distribution between caudate-putamen and nucleus accumbens [38,39]. D-264 has a Ki value of 49 nM at D4 receptors (National Institute of Mental Health's Psychoactive Drug Screening Program [PDSP], data not shown), well within the concentration range examined, but D264 had very minimal effect on G-protein activity in the striatum of D2R KO mice. D4 affinity of D-301 is unknown, however, we have observed no significant effect of the D4-selective antagonist LY745870 on D-301-stimulated [35S]GTP S binding in ICR mouse striatum at antagonist concentrations up to 1 μM (J.R. Secor McVoy and D.E. Selley, unpublished data), which is approximately 1,000 times greater than its Ki value at D4 receptors [40]. Future studies could be conducted to determine the relative efficacy of these ligands to stimulate D3 receptor-mediated G protein activation in striatal regions of D2 and D4 double KO mice.

Our findings with D2/3 receptor-mediated G protein activity in mouse striatum have implications for the exclusive use of heterologously transfected cell lines to determine dopaminergic ligand efficacy, because both D-301 and D-264 produced similar Emax values that were not different from DA in CHO cells expressing either D2 or D3 receptors. Similar results have been obtained with several mu opiate ligands that appeared to be full agonists in cell lines heterologously expressing the mu opioid receptor but partial agonists in rat thalamus, based on Emax values of ligand-stimulated [35S]GTP S binding [11,12,14,41]. However, heterologous receptor expression is generally driven by efficient viral promoters and is therefore often higher than in native tissues. The relationship between receptor expression levels and ligand efficacy determinations are well established; in the mu opioid receptor example we previously observed that MOR expression levels were approximately five-fold greater in transfected CHO cells than in rat thalamus [11,12]. In the current study, D2 and D3 receptor Bmax values were approximately 2 to 4-fold and 135-fold, respectively, greater than those reported in rodent striatum [22,42,43]. Although receptor Bmax comparisons between brain and cell lines must be viewed with caution because presumably not every cell type in a dissected brain region expresses the receptor of interest, it seems likely that these receptors, especially D3, are over-expressed in our CHO cell models relative to naturally occurring levels in the striatum. This is particularly true when taking into account the ratio of receptors to activated Gi/o proteins, because activated Gi/o protein levels tend to be much greater in the brain [11]. One should, however, keep in mind that compensatory changes in dopamine receptor expression – especially ‘D2 high’ receptor expression- are well documented in dopamine receptor knockout animals [44]. All considerations taken together, one expects receptor reserve for G protein activation by high efficacy ligands to be more likely present in transfected cell models. Therefore, we determined the intrinsic efficacy of ligand-stimulated G protein activation by determining receptor binding Ki to G protein activation EC50 values [12,23], which revealed significantly greater intrinsic efficacy of DA than either D-264 or D-301 in cells expressing either D2 or D3 receptors. Strikingly, the intrinsic efficacy of the synthetic ligands relative to DA was only approximately 23-28% at D2 and 6.4% (D-264) to 10.5% (D-301) at D3 receptors. Accordingly, DA produced Ki/EC50 ratios greater than unity in both D2 and D3 receptor-expressing cells, indicative of receptor reserve, whereas only D-301 in D3 receptor-expressing cells and neither synthetic ligand in D2 receptor-expressing cells had a Ki/EC50 ratio significantly greater than unity. Moreover, the intrinsic efficacies of D-264 and D-301 were approximately equal at D2 receptors, whereas that of D-301 was 1.65-fold greater than D-264 at D3 receptors. Although these results are in general agreement with those obtained in mouse striatum, the efficacy of D-301 relative to D-264 for D3 receptor-mediated G-protein activation seemed to be lower in the cell model compared to striatum.

Another major difference between the CHO cell models and mouse striatum, besides D3 receptor expression levels, is the high expression of G o1 in striatum [25] compared to its reported absence in CHO cells [33]. We therefore examined the relative intrinsic efficacies of D-264 and D-301 in the same cell lines with co-expression of G o1. Unexpectedly, endogenous G o1 protein was detectable in our CHO cell lines, and transfection with exogenous G o1 only led to an approximate doubling of G o1 (Fig. 3). It is therefore not surprising that in most cases the effects of transfected exogenous G o1 were quite moderate, as were the observed DA-induced increases in GTPγS binding (exogenous G o1 increased the % baseline response to standard, maximally effective concentration of DA from 223% to 244% in D2 cells and from 134% to 160% in D3 cells as measured for each receptor subtype in two independent transfection experiments assayed in sextuplicate). Expression of exogenous G o1 did not affect receptor expression levels of either receptor type, and minimally affected the intrinsic efficacy of DA at D2 receptors. Perhaps surprisingly, the intrinsic efficacy of DA at D3 receptors was significantly reduced by exogenous G o1 expression, and this reduction appeared to mainly result from a significant decrease in binding Ki value, suggesting that DA might form more stable high affinity D3 receptor complexes with G o1 than with the G i isoforms present in CHO cells. However, the lack of a corresponding decrease in EC50 value suggests that this enhanced high affinity complex did not result in increased efficiency to induce guanine nucleotide exchange.

Studies suggest that both D2 and D3 receptors are more efficiently coupled to G o than G i2 or 3 [24,45], which are the major G i isoforms in CHO cells[46]. Moreover, differential activation of G o versus G i isoforms by D2-like receptors can also be ligand-dependent [47,48]. In the present study, the intrinsic efficacies of both D-264 and D-301 were not significantly affected by exogenous G o1 expression in D2-expressing cells. Exogenous G o1-expression appeared to enhance the intrinsic efficacy of D-264 and D-301 (by ~50%) in D3-expressing cells, such that D-264 gained receptor reserve indicated by a Ki/EC50 value greater than unity. However, there was not a statistically significant increase in the calculated intrinsic efficacy of either ligand and only D-301 exhibited an intrinsic efficacy value greater than unity. Nonetheless, only in the presence of exogenous G o1 did the intrinsic efficacy of D-301 at D3 receptors approach that of DA, whereas this ligand retained very low intrinsic efficacy in D2-expressing cells regardless of the presence or absence of exogenous G o1. Examination of the receptor-binding data suggests that the major explanation for this convergence of intrinsic efficacy values between D-301 and DA was that the binding Ki values were differentially affected by exogenous G o1 expression. The D-301 affinity decreased in exogenous G o1 expressing cells whereas the DA affinity increased, and the EC50 values of both ligands were unaffected. Although the potency of D-264 to activate G-proteins was significantly increased by exogenous G o1 expression in D3-expressing cells, the Emax value was moderately decreased so there was no significant effect of exogenous G o1 expression on intrinsic efficacy of this ligand. Thus, the major effect of exogenous G o1 expression in D3-expressing cells was enhancement of the binding affinity of DA but not of the synthetic ligands, such that the intrinsic efficacy values became more similar between DA and the synthetic ligands (especially D-301) in exogenous G o1-expressing cells.

While the precise mechanisms underlying the relative intrinsic efficacy differences between D-301 and D-264 at D3 receptors remains to be elucidated, one relevant conclusion from these CHO cell studies is that exogenous G o1 expression did not affect the ratio of intrinsic efficacies of D-301:D-264 at D3 receptors, which was approximately 1.65 regardless of exogenous G o1 expression. Thus, the major difference between the D-301:D-264 efficacy ratio in D3-CHO cells compared to mouse striatum appears to be D3 receptor expression level, which could be addressed in future studies by progressively reducing D3 receptor expression in the CHO cell model using an irreversible antagonist. This treatment would be expected to disproportionately suppress the efficacy of D-264 relative to that of D-301. It is known that decreases in receptor expression levels disproportionally suppress the relative efficacy of low compared to high efficacy ligands, as has been seen with mu opioid [12] and other GPCRs [49-51]. Nonetheless, because results in both the striatum of D3 KO mice and transfected CHO cells (when taking into account the intrinsic efficacy calculation) indicated that D301 is more efficacious than D264 to activate G-proteins via D3 receptors, we can therefore reject the null hypothesis that D301 and D264 are equally efficacious as D3 agonists.

Considering the important role of receptor expression density in the determination of ligand efficacy, it is interesting that maximal stimulation of [35S]GTP S binding in absolute terms was similar between cells expressing D2 and D3 receptors with Bmax values of <1 and >7 pmol/mg, respectively. This discrepancy between receptor density and G protein activation magnitude probably derives from the low efficiency of D3 receptor-G-protein coupling, as previously reported [9,34,52] The low efficiency of D3-mediated G protein activation could be related to its reportedly rigid conformation, which also imparts lesser sensitivity of agonist binding affinity to guanine nucleotides [53-55]. Interestingly, despite the reported low efficiency of D3 receptor-mediated activation of G i/o proteins, this receptor seems to be more promiscuously coupled to activation of multiple G families, including G s [56,57] and G q [9], relative to other D2-like receptor family members. It will be of interest in future studies to determine whether the efficacy relationship between D301 and D264 is similar for activation of these other G types.

In conclusion, the present study revealed differential intrinsic efficacies for D3 receptor-mediated G protein activation between two chemically similar synthetic ligands, with D-301 showing greater efficacy than D-264. In contrast, these two ligands showed similar efficacy to activate D2 receptors. This conclusion is supported by data showing greater Emax values of D-301 than D-264 in striatal regions from D2 KO mice, and greater intrinsic efficacy of D-301 than D-264 in D3-expressing CHO cells based on a calculation that accounts for receptor reserve in this model. The higher intrinsic efficacy of D-301 relative to D-264 was not significantly altered by co-expression of exogenous G o1, suggesting that higher D3 receptor expression and not differential expression of G subtypes is the predominant factor underlying the differential relative Emax values for G protein activation between the CHO cell model and mouse striatum. These findings suggest caution in interpretation of ligand efficacy determinations from a single model system, particularly when potential receptor reserve for the response is not directly determined.

Acknowledgements

This work is supported by National Institute of Neurological Disorders and Stroke/ National Institute of Health (NS047198, AKD) and National Institute of Health (R01-NS070715, DES).

Footnotes

Compliance with Ethical Standards

Conflict of interest: The authors declare no conflict of interest.

References

  • 1.Sokoloff P, Diaz J, Le Foll B, Guillin O, Leriche L, Bezard E, Gross C. The dopamine D3 receptor: a therapeutic target for the treatment of neuropsychiatric disorders. CNS & neurological disorders drug targets. 2006;5(1):25–43. doi: 10.2174/187152706784111551. [DOI] [PubMed] [Google Scholar]
  • 2.Brisch R, Saniotis A, Wolf R, Bielau H, Bernstein HG, Steiner J, Bogerts B, Braun K, Jankowski Z, Kumaratilake J, Henneberg M, Gos T. The role of dopamine in schizophrenia from a neurobiological and evolutionary perspective: old fashioned, but still in vogue. Frontiers in psychiatry. 2014;5:47. doi: 10.3389/fpsyt.2014.00047. doi:10.3389/fpsyt.2014.00047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lahti RA, Roberts RC, Tamminga CA. D2-family receptor distribution in human postmortem tissue: an autoradiographic study. Neuroreport. 1995;6(18):2505–2512. doi: 10.1097/00001756-199512150-00015. [DOI] [PubMed] [Google Scholar]
  • 4.Stanwood GD, Lucki I, McGonigle P. Differential regulation of dopamine D2 and D3 receptors by chronic drug treatments. The Journal of pharmacology and experimental therapeutics. 2000;295(3):1232–1240. [PubMed] [Google Scholar]
  • 5.Hall H, Halldin C, Dijkstra D, Wikstrom H, Wise LD, Pugsley TA, Sokoloff P, Pauli S, Farde L, Sedvall G. Autoradiographic localisation of D3-dopamine receptors in the human brain using the selective D3-dopamine receptor agonist (+)-[3H]PD 128907. Psychopharmacology. 1996;128(3):240–247. doi: 10.1007/s002130050131. [DOI] [PubMed] [Google Scholar]
  • 6.Barth V, Need AB, Tzavara ET, Giros B, Overshiner C, Gleason SD, Wade M, Johansson AM, Perry K, Nomikos GG, Witkin JM. In vivo occupancy of dopamine D3 receptors by antagonists produces neurochemical and behavioral effects of potential relevance to attention-deficit-hyperactivity disorder. The Journal of pharmacology and experimental therapeutics. 2013;344(2):501–510. doi: 10.1124/jpet.112.198895. doi:10.1124/jpet.112.198895. [DOI] [PubMed] [Google Scholar]
  • 7.Le Foll B, Di Ciano P. Neuronal circuitry underlying the impact of D3 receptor ligands in drug addiction. European neuropsychopharmacology : the journal of the European College of Neuropsychopharmacology. 2014 doi: 10.1016/j.euroneuro.2014.08.017. doi:10.1016/j.euroneuro.2014.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Keck TM, John WS, Czoty PW, Nader MA, Newman AH. Identifying Medication Targets for Psychostimulant Addiction: Unraveling the Dopamine D3 Receptor Hypothesis. Journal of medicinal chemistry. 2015;58(14):5361–5380. doi: 10.1021/jm501512b. doi:10.1021/jm501512b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.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. Molecular pharmacology. 1999;55(3):564–574. [PubMed] [Google Scholar]
  • 10.Neve KA, Seamans JK, Trantham-Davidson H. Dopamine receptor signaling. Journal of receptor and signal transduction research. 2004;24(3):165–205. doi: 10.1081/rrs-200029981. [DOI] [PubMed] [Google Scholar]
  • 11.Selley DE, Sim LJ, Xiao R, Liu Q, Childers SR. mu-Opioid receptor-stimulated guanosine-5′-O-(gamma-thio)-triphosphate binding in rat thalamus and cultured cell lines: signal transduction mechanisms underlying agonist efficacy. Molecular pharmacology. 1997;51(1):87–96. doi: 10.1124/mol.51.1.87. [DOI] [PubMed] [Google Scholar]
  • 12.Selley DE, Liu Q, Childers SR. Signal transduction correlates of mu opioid agonist intrinsic efficacy: receptor-stimulated [35S]GTP gamma S binding in mMOR-CHO cells and rat thalamus. The Journal of pharmacology and experimental therapeutics. 1998;285(2):496–505. [PubMed] [Google Scholar]
  • 13.Sally EJ, Xu H, Dersch CM, Hsin LW, Chang LT, Prisinzano TE, Simpson DS, Giuvelis D, Rice KC, Jacobson AE, Cheng K, Bilsky EJ, Rothman RB. Identification of a novel “almost neutral” micro-opioid receptor antagonist in CHO cells expressing the cloned human mu-opioid receptor. Synapse (New York, NY) 2010;64(4):280–288. doi: 10.1002/syn.20723. doi:10.1002/syn.20723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Antonio T, Childers SR, Rothman RB, Dersch CM, King C, Kuehne M, Bornmann WG, Eshleman AJ, Janowsky A, Simon ER, Reith ME, Alper K. Effect of Iboga alkaloids on micro-opioid receptor-coupled G protein activation. PloS one. 2013;8(10):e77262. doi: 10.1371/journal.pone.0077262. doi:10.1371/journal.pone.0077262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Meller E, Bohmaker K, Namba Y, Friedhoff AJ, Goldstein M. Relationship between receptor occupancy and response at striatal dopamine autoreceptors. Molecular pharmacology. 1987;31(6):592–598. [PubMed] [Google Scholar]
  • 16.Burris KD, Molski TF, Xu C, Ryan E, Tottori K, Kikuchi T, Yocca FD, Molinoff PB. Aripiprazole, a novel antipsychotic, is a high-affinity partial agonist at human dopamine D2 receptors. The Journal of pharmacology and experimental therapeutics. 2002;302(1):381–389. doi: 10.1124/jpet.102.033175. [DOI] [PubMed] [Google Scholar]
  • 17.Tadori Y, Forbes RA, McQuade RD, Kikuchi T. Receptor reserve-dependent properties of antipsychotics at human dopamine D2 receptors. European journal of pharmacology. 2009;607(1-3):35–40. doi: 10.1016/j.ejphar.2009.02.007. doi:10.1016/j.ejphar.2009.02.007. [DOI] [PubMed] [Google Scholar]
  • 18.Ghosh B, Antonio T, Zhen J, Kharkar P, Reith ME, Dutta AK. Development of (S)-N6-(2-(4-(isoquinolin-1-yl)piperazin-1-yl)ethyl)-N6-propyl-4,5,6,7-tetrahydro benzo[d]-thiazole-2,6-diamine and its analogue as a D3 receptor preferring agonist: potent in vivo activity in Parkinson's disease animal models. Journal of medicinal chemistry. 2010;53(3):1023–1037. doi: 10.1021/jm901184n. doi:10.1021/jm901184n. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Biswas S, Hazeldine S, Ghosh B, Parrington I, Kuzhikandathil E, Reith ME, Dutta AK. Bioisosteric heterocyclic versions of 7-{[2-(4-phenyl-piperazin-1-yl)ethyl]propylamino}-5,6,7,8-tetrahydronaphthalen-2- ol: identification of highly potent and selective agonists for dopamine D3 receptor with potent in vivo activity. J Med Chem. 2008;51(10):3005–3019. doi: 10.1021/jm701524h. doi:10.1021/jm701524h. [DOI] [PubMed] [Google Scholar]
  • 20.Kortagere S, Cheng SY, Antonio T, Zhen J, Reith ME, Dutta AK. Interaction of novel hybrid compounds with the D3 dopamine receptor: Site-directed mutagenesis and homology modeling studies. Biochemical pharmacology. 2011;81(1):157–163. doi: 10.1016/j.bcp.2010.08.026. doi:10.1016/j.bcp.2010.08.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Li C, Biswas S, Li X, Dutta AK, Le W. Novel D3 dopamine receptor-preferring agonist D-264: Evidence of neuroprotective property in Parkinson's disease animal models induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and lactacystin. Journal of neuroscience research. 2010;88(11):2513–2523. doi: 10.1002/jnr.22405. doi:10.1002/jnr.22405. [DOI] [PubMed] [Google Scholar]
  • 22.Kelly MA, Rubinstein M, Asa SL, Zhang G, Saez C, Bunzow JR, Allen RG, Hnasko R, Ben-Jonathan N, Grandy DK, Low MJ. Pituitary lactotroph hyperplasia and chronic hyperprolactinemia in dopamine D2 receptor-deficient mice. Neuron. 1997;19(1):103–113. doi: 10.1016/s0896-6273(00)80351-7. [DOI] [PubMed] [Google Scholar]
  • 23.Ehlert FJ. The relationship between muscarinic receptor occupancy and adenylate cyclase inhibition in the rabbit myocardium. Molecular pharmacology. 1985;28(5):410–421. [PubMed] [Google Scholar]
  • 24.Lane EL, Handley OJ, Rosser AE, Dunnett SB. Potential cellular and regenerative approaches for the treatment of Parkinson's disease. Neuropsychiatric disease and treatment. 2008;4(5):835–845. doi: 10.2147/ndt.s2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hildebrandt JD. Role of subunit diversity in signaling by heterotrimeric G proteins. Biochemical pharmacology. 1997;54(3):325–339. doi: 10.1016/s0006-2952(97)00269-4. [DOI] [PubMed] [Google Scholar]
  • 26.Gogoi S, Antonio T, Rajagopalan S, Reith M, Andersen J, Dutta AK. Dopamine D(2)/D(3) agonists with potent iron chelation, antioxidant and neuroprotective properties: potential implication in symptomatic and neuroprotective treatment of Parkinson's disease. ChemMedChem. 2011;6(6):991–995. doi: 10.1002/cmdc.201100140. doi:10.1002/cmdc.201100140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Johnson M, Antonio T, Reith ME, Dutta AK. Structure-activity relationship study of N(6)-(2-(4-(1HIndol-5-yl)piperazin-1-yl)ethyl)-N(6)-propyl-4,5,6,7-tetrahydroben zo[d]thiazole-2,6-diamine analogues: development of highly selective D3 dopamine receptor agonists along with a highly potent D2/D3 agonist and their pharmacological characterization. Journal of medicinal chemistry. 2012;55(12):5826–5840. doi: 10.1021/jm300268s. doi:10.1021/jm300268s. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Sim-Selley LJ, Cassidy MP, Sparta A, Zachariou V, Nestler EJ, Selley DE. Effect of DeltaFosB overexpression on opioid and cannabinoid receptor-mediated signaling in the nucleus accumbens. Neuropharmacology. 2011;61(8):1470–1476. doi: 10.1016/j.neuropharm.2011.08.046. doi:10.1016/j.neuropharm.2011.08.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Munson PJ, Rodbard D. An exact correction to the “Cheng-Prusoff” correction. J Recept Res. 1988;8(1-4):533–546. doi: 10.3109/10799898809049010. [DOI] [PubMed] [Google Scholar]
  • 30.Zhen J, Antonio T, Dutta AK, Reith ME. Concentration of receptor and ligand revisited in a modified receptor binding protocol for high-affinity radioligands: [3H]Spiperone binding to D2 and D3 dopamine receptors. J Neurosci Methods. 2010;188(1):32–38. doi: 10.1016/j.jneumeth.2010.01.031. doi:10.1016/j.jneumeth.2010.01.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Cheng Y, Prusoff WH. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochemical pharmacology. 1973;22(23):3099–3108. doi: 10.1016/0006-2952(73)90196-2. [DOI] [PubMed] [Google Scholar]
  • 32.Silverman MP, Strange WLTC. The distribution of composite measurements: How to be certain of the uncertainties in what we measure. American Journal of Physics. 2004;72(8):13. [Google Scholar]
  • 33.Raymond JR, Olsen CL, Gettys TW. Cell-specific physical and functional coupling of human 5-HT1A receptors to inhibitory G protein alpha-subunits and lack of coupling to Gs alpha. Biochemistry. 1993;32(41):11064–11073. doi: 10.1021/bi00092a016. [DOI] [PubMed] [Google Scholar]
  • 34.Zaworski PG, Alberts GL, Pregenzer JF, Im WB, Slightom JL, Gill GS. Efficient functional coupling of the human D3 dopamine receptor to G(o) subtype of G proteins in SH-SY5Y cells. British journal of pharmacology. 1999;128(6):1181–1188. doi: 10.1038/sj.bjp.0702905. doi:10.1038/sj.bjp.0702905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gainetdinov RR, Bohn LM, Sotnikova TD, Cyr M, Laakso A, Macrae AD, Torres GE, Kim KM, Lefkowitz RJ, Caron MG, Premont RT. Dopaminergic supersensitivity in G protein-coupled receptor kinase 6-deficient mice. Neuron. 2003;38(2):291–303. doi: 10.1016/s0896-6273(03)00192-2. [DOI] [PubMed] [Google Scholar]
  • 36.Kim KM, Gainetdinov RR, Laporte SA, Caron MG, Barak LS. G protein-coupled receptor kinase regulates dopamine D3 receptor signaling by modulating the stability of a receptor-filamin-beta-arrestin complex. A case of autoreceptor regulation. The Journal of biological chemistry. 2005;280(13):12774–12780. doi: 10.1074/jbc.M408901200. doi:10.1074/jbc.M408901200. [DOI] [PubMed] [Google Scholar]
  • 37.Barak LS, Gilchrist J, Becker JM, Kim KM. Relationship between the G protein signaling and homologous desensitization of G protein-coupled receptors. Biochemical and biophysical research communications. 2006;339(2):695–700. doi: 10.1016/j.bbrc.2005.11.070. doi:10.1016/j.bbrc.2005.11.070. [DOI] [PubMed] [Google Scholar]
  • 38.Defagot MC, Malchiodi EL, Villar MJ, Antonelli MC. Distribution of D4 dopamine receptor in rat brain with sequence-specific antibodies. Brain research Molecular brain research. 1997;45(1):1–12. doi: 10.1016/s0169-328x(96)00235-5. [DOI] [PubMed] [Google Scholar]
  • 39.Tarazi FI, Campbell A, Yeghiayan SK, Baldessarini RJ. Localization of dopamine receptor subtypes in corpus striatum and nucleus accumbens septi of rat brain: comparison of D1-, D2-, and D4-like receptors. Neuroscience. 1998;83(1):169–176. doi: 10.1016/s0306-4522(97)00386-2. [DOI] [PubMed] [Google Scholar]
  • 40.Patel S, Freedman S, Chapman KL, Emms F, Fletcher AE, Knowles M, Marwood R, McAllister G, Myers J, Curtis N, Kulagowski JJ, Leeson PD, Ridgill M, Graham M, Matheson S, Rathbone D, Watt AP, Bristow LJ, Rupniak NM, Baskin E, Lynch JJ, Ragan CI. Biological profile of L-745,870, a selective antagonist with high affinity for the dopamine D4 receptor. The Journal of pharmacology and experimental therapeutics. 1997;283(2):636–647. [PubMed] [Google Scholar]
  • 41.Selley DE, Cao CC, Liu Q, Childers SR. Effects of sodium on agonist efficacy for G-protein activation in mu-opioid receptor-transfected CHO cells and rat thalamus. British journal of pharmacology. 2000;130(5):987–996. doi: 10.1038/sj.bjp.0703382. doi:10.1038/sj.bjp.0703382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Skinbjerg M, Seneca N, Liow JS, Hong J, Weinshenker D, Pike VW, Halldin C, Sibley DR, Innis RB. Dopamine beta-hydroxylase-deficient mice have normal densities of D(2) dopamine receptors in the high-affinity state based on in vivo PET imaging and in vitro radioligand binding. Synapse (New York, NY) 2010;64(9):699–703. doi: 10.1002/syn.20781. doi:10.1002/syn.20781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Piercey MF, Walker EL, Feldpausch DL, Camacho-Ochoa M. High affinity binding for pramipexole, a dopamine D3 receptor ligand, in rat striatum. Neuroscience letters. 1996;219(2):138–140. [PubMed] [Google Scholar]
  • 44.Seeman P, Schwarz J, Chen JF, Szechtman H, Perreault M, McKnight GS, Roder JC, Quirion R, Boksa P, Srivastava LK, Yanai K, Weinshenker D, Sumiyoshi T. Psychosis pathways converge via D2high dopamine receptors. Synapse (New York, NY) 2006;60(4):319–346. doi: 10.1002/syn.20303. doi:10.1002/syn.20303. [DOI] [PubMed] [Google Scholar]
  • 45.Nickolls SA, Strange PG. Interaction of the D2short dopamine receptor with G proteins: analysis of receptor/G protein selectivity. Biochemical pharmacology. 2003;65(7):1139–1150. doi: 10.1016/s0006-2952(03)00040-6. [DOI] [PubMed] [Google Scholar]
  • 46.Gettys TW, Sheriff-Carter K, Moomaw J, Taylor IL, Raymond JR. Characterization and use of crude alpha-subunit preparations for quantitative immunoblotting of G proteins. Analytical biochemistry. 1994;220(1):82–91. doi: 10.1006/abio.1994.1302. [DOI] [PubMed] [Google Scholar]
  • 47.Lane JR, Powney B, Wise A, Rees S, Milligan G. Protean agonism at the dopamine D2 receptor: (S)-3-(3-hydroxyphenyl)-N-propylpiperidine is an agonist for activation of Go1 but an antagonist/inverse agonist for Gi1,Gi2, and Gi3. Molecular pharmacology. 2007;71(5):1349–1359. doi: 10.1124/mol.106.032722. doi:10.1124/mol.106.032722. [DOI] [PubMed] [Google Scholar]
  • 48.Moller D, Kling RC, Skultety M, Leuner K, Hubner H, Gmeiner P. Functionally selective dopamine D(2), D(3) receptor partial agonists. Journal of medicinal chemistry. 2014;57(11):4861–4875. doi: 10.1021/jm5004039. doi:10.1021/jm5004039. [DOI] [PubMed] [Google Scholar]
  • 49.Boddeke HW, Fargin A, Raymond JR, Schoeffter P, Hoyer D. Agonist/antagonist interactions with cloned human 5-HT1A receptors: variations in intrinsic activity studied in transfected HeLa cells. Naunyn-Schmiedeberg's archives of pharmacology. 1992;345(3):257–263. doi: 10.1007/BF00168684. [DOI] [PubMed] [Google Scholar]
  • 50.MacEwan DJ, Kim GD, Milligan G. Analysis of the role of receptor number in defining the intrinsic activity and potency of partial agonists in neuroblastoma x glioma hybrid NG108-15 cells transfected to express differing levels of the human beta 2-adrenoceptor. Molecular pharmacology. 1995;48(2):316–325. [PubMed] [Google Scholar]
  • 51.Varrault A, Journot L, Audigier Y, Bockaert J. Transfection of human 5-hydroxytryptamine1A receptors in NIH-3T3 fibroblasts: effects of increasing receptor density on the coupling of 5-hydroxytryptamine1A receptors to adenylyl cyclase. Molecular pharmacology. 1992;41(6):999–1007. [PubMed] [Google Scholar]
  • 52.Freedman SB, Patel S, Marwood R, Emms F, Seabrook GR, Knowles MR, McAllister G. Expression and pharmacological characterization of the human D3 dopamine receptor. The Journal of pharmacology and experimental therapeutics. 1994;268(1):417–426. [PubMed] [Google Scholar]
  • 53.Vanhauwe JF, Fraeyman N, Francken BJ, Luyten WH, Leysen JE. Comparison of the ligand binding and signaling properties of human dopamine D(2) and D(3) receptors in Chinese hamster ovary cells. The Journal of pharmacology and experimental therapeutics. 1999;290(2):908–916. [PubMed] [Google Scholar]
  • 54.Vanhauwe JF, Josson K, Luyten WH, Driessen AJ, Leysen JE. G-protein sensitivity of ligand binding to human dopamine D(2) and D(3) receptors expressed in Escherichia coli: clues for a constrained D(3) receptor structure. The Journal of pharmacology and experimental therapeutics. 2000;295(1):274–283. [PubMed] [Google Scholar]
  • 55.Platania CB, Salomone S, Leggio GM, Drago F, Bucolo C. Homology modeling of dopamine D2 and D3 receptors: molecular dynamics refinement and docking evaluation. PloS one. 2012;7(9):e44316. doi: 10.1371/journal.pone.0044316. doi:10.1371/journal.pone.0044316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.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. Cellular and molecular neurobiology. 1999;19(5):653–664. doi: 10.1023/A:1006988603199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Ilani T, Fishburn CS, Levavi-Sivan B, Carmon S, Raveh L, Fuchs S. Coupling of dopamine receptors to G proteins: studies with chimeric D2/D3 dopamine receptors. Cellular and molecular neurobiology. 2002;22(1):47–56. doi: 10.1023/A:1015341712166. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES