Abstract
Background and Purpose
Dopamine and corticotrophin-releasing hormone (CRH; also known as corticotrophin-releasing factor) are key neurotransmitters in the interaction between stress and addiction. Repeated treatment with cocaine potentiates glutamatergic transmission in the rat basolateral amygdala/cortex pathway through a synergistic action of D1-like dopamine receptors and CRH type-2α receptors (CRF2α receptors). We hypothesized that this observed synergism could be instrumented by heteromers containing the dopamine D1 receptor and CRF2α receptor.
Experimental Approach
D1/CRF2α receptor heteromerization was demonstrated in HEK293T cells using co-immunoprecipitation, BRET and FRET assays, and by using the heteromer mobilization strategy. The ability of D1 receptors to signal through calcium, when singly expressed or co-expressed with CRF2α receptors, was evaluated by the calcium mobilization assay.
Key Results
D1/CRF2α receptor heteromers were observed in HEK293T cells. When singly expressed, D1 receptors were mostly located at the cell surface whereas CRF2α receptors accumulated intracellularly. Interestingly, co-expression of both receptors promoted D1 receptor intracellular and CRF2α receptor cell surface targeting. The heteromerization of D1/CRF2α receptors maintained the signalling through cAMP of both receptors but switched D1 receptor signalling properties, as the heteromeric D1 receptor was able to mobilize intracellular calcium upon stimulation with a D1 receptor agonist.
Conclusions and Implications
D1 and CRF2α receptors are capable of heterodimerization in living cells. D1/CRF2α receptor heteromerization might account, at least in part, for the complex physiological interactions established between dopamine and CRH in normal and pathological conditions such as addiction, representing a new potential pharmacological target.
Tables of Links
| TARGETS | LIGANDS |
|---|---|
| 5-HT2A/C receptor | cAMP |
| α1B-adrenoceptor | CRH (CRF) |
| μ opioid receptor | Dopamine |
| CB1 receptor | Glutamate |
| CRF1 receptor | SCH23390 |
| CRF2 receptor | Urocortin 1 |
| D1 receptor | |
| D2 receptor | |
| GABAB2 receptor |
This Table lists key protein targets and ligands in this document, which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2013/14 (Alexander et al., 2013b).
Introduction
Stress-induced relapse to drug seeking is one of the main problems in drug addiction treatment (Koob, 2008), in part because of the lack of suitable pharmacological targets. It has been shown that the exposure to drugs of abuse and to stressful stimuli induce similar neuronal plastic changes strengthening excitatory inputs to midbrain dopaminergic neurons (Saal et al., 2003). However, the mechanisms of this interaction are still not fully understood. The available evidence supports a key role for the neurotransmitters corticotrophin-releasing hormone (CRH also known as corticotrophin-releasing factor) and dopamine driving the interaction between stress and addiction (Corominas et al., 2010; George et al., 2012; Gysling, 2012; Zorrilla et al., 2014). CRH signalling occurs through the activation of two class B1 GPRC (Alexander et al., 2013a,2014), CRF1 and CRF2 receptors. Several studies have shown that the CRF1 receptor has a key role in plastic changes associated with stress and addiction (Shalev et al., 2010). However, more recently, the role of the CRF2 receptor in plastic changes induced by addictive drugs has also been recognized (Wang et al., 2007; Hahn et al., 2009; Cadet et al., 2014; Guan et al., 2014). Wise and his group (Wang et al., 2005) have shown that a stressful stimulus induces the release of CRH in the ventral tegmental area (VTA) of both naive and cocaine-experienced rats, and that glutamate release becomes sensitized to CRH only in cocaine-experienced rats. They have shown that this sensitization of VTA glutamate is mediated by CRF2 receptors (Wang et al., 2007). In addition, Orozco-Cabal et al. (2008), using an electrophysiological approach, showed a synergism between D1-like dopamine receptors and CRF2 receptors in basolateral amygdala to medial prefrontal cortex synaptic transmission in rats subjected to repeated cocaine treatment. Thus, the receptors involved in this synergism could be, in part, responsible for the link between addiction and stress.
Increasing evidence shows that the ability of GPCRs to form oligomers plays a key role in synaptic transmission (Ciruela et al., 2012). It has been documented that dopamine receptors are able to form heteromers between them and with other GPCRs (Perreault et al., 2014). Interestingly, it has been shown that the heteromerization of D1 and D2 dopamine receptors, a novel receptorial entity able to induce calcium mobilization through a switch to Gq-mediated activation of PLC (Lee et al., 2004; Rashid et al., 2007). More recent evidence suggests that the mechanisms of calcium mobilization induced by the heteromerization of D1 and D2 receptors are more complex, implying other intracellular mediators (Chun et al.,).
The possible heteromerization between D1 and CRF2 receptors could be responsible for the synergism between dopamine and CRH transmission (Orozco-Cabal et al., 2008). As the α-isoform of CRF2 receptors is the main isoform expressed in the brain (Chalmers et al., 1995; Lovenberg et al., 1995; Dautzenberg and Hauger, 2002), we hypothesized that D1 and CRF2α receptors assemble into functional interacting complexes (i.e. heteromers). The existence of a D1/CRF2α receptor heteromer would constitute a novel potential target for drug addiction pharmacotherapy.
The present study aimed to determine the molecular and functional interactions between D1 and CRF2α receptors in living cells. Overall, we provide evidence of the existence of a novel and genuine D1/CRF2α receptor heteromer that behaves as a different receptorial entity to their individual constituents.
Methods
Plasmid constructs
The constructs encoding human D1, CRF1 and CRF2α receptors were isolated by Touch-Down PCR using PfuUltra II Fusion HS DNA Polymerase (Agilent Technologies, Palo Alto, CA, USA). PCR products were digested with the respective restriction enzymes and cloned into the expression vector listed in Table 1. The D2 receptorYFP and GABAB2 receptorYFP constructs were prepared as previously described (Canals et al., 2003). The CRF2α receptorFlag and D1 receptor-nuclear localization sequence (NLS)YFP were generated by PCR overlapping extension as previously described (Heckman and Pease, 2007), using two coupled primers each. The primers used are listed in Table 1 and the restriction sites within the primers are underlined. All constructs were confirmed by DNA sequencing.
Table 1.
Constructs information
| Construct | Vector | Restriction enzyme | Primers |
|---|---|---|---|
| D1 receptor | pcDNA 3.1 | EcoRV and NotI | CCGGATATCGGCGCCAACATGAGGACTCTGAACACC |
| CGAGCGGCCGCTCAGGTTGGGTGCTGACCGTT | |||
| CRF2α receptor | pcDNA 3.1 | HindIII and XhoI | CCGAAGCTTGGCGCCACCATGGACGCGGCACTGCTC |
| CGACTCGAGTCACACAGCGGCCGTCTGCTT | |||
| D1 receptorYFP | pEYFP-N1 | EcoRI and BamHI | CTCGAATTCGCCACCATGAGGACTCTGAACACC |
| CGTCGCCGTCCAGCTCGACCAG | |||
| CRF2α receptorCFP | pECFP-N1 | EcoRI and KpnI | CCGGAATTCGCCACCATGGACGCGGCACTG |
| CGTCGCCGTCCAGCTCGACCAG | |||
| CRF1 receptorYFP | pEYFP-N1 | EcoRI and KpnI | GTGGAATTCACCATGGGAGGGCAC |
| CCGCGGTACCCAGACTGCTGTGGA | |||
| D1 receptormyc/His | pcDNA 3.1 myc/His | EcoRV and BamHI | CCGGATATCGGCGCCAACATGAGGACTCTGAACACC |
| CGAGGATCCGGTTGGGTGCTGACCGTT | |||
| CRF2α receptorRluc | pRluc-N1 | EcoRI and KpnI | CTCGAATTCGCCACCATGGACGCGGCACTGCTC CGCGGTACCGCCACAGCGGCCGTCTGCTTG |
| D1 receptor-NLSYFP | pEYFP-N1 | EcoRI and BamHI | CTCGAATTCGCCACCATGAGGACTCTGAACACC CCTAAGAGGGTTGAAAATCTTTTAAATTTTTTAGCATTAAAGGCATAAATG GCCTTTAATGCTAAAAAATTTAAAAGATTTTCAACCCTCTTAGGATGC CGTCGCCGTCCAGCTCGACCAG |
| CRF2α receptorFlag | pcDNA 3.1 | HindIII and NotI | CCGAAGCTTGGCGCCACCATGGACGCGGCACTGCTC GTCGTCATCCTTGTAGTCCACAGCGGCCGTCTGCTT |
| GACTACAAGGATGACGACGATAAGTGAGCG GAATAGAATGACACCTACTCAGACAATCGC |
Each construct was generated by PCR amplification and the primers used are listed. The restriction enzymes and vectors into which they were sub-cloned are also indicated.
Cell culture and transfection
HEK293T cells were grown in 100 mm plates with DMEM (Gibco, Gaithersburg, MD, USA) supplemented with 10% FBS (Hyclone Labs, Logan, UT, USA), 1% penicillin/streptomycin 100× (Gibco), 2 mM GlutaMax (Gibco) and 1% non-essential amino acids 100× (Gibco) at 37°C in a 5% CO2 humidified atmosphere. Transfection was performed 24 h post seeding, using either Lipofectamine 2000 reagent (Invitrogen, Carlsbad, CA, USA) or linear polyethylenimine (Poly Sciences, Inc., Warrington, PA, USA).
Immunofluorescence
HEK293T cells (7 × 105) were grown on 24-well plates on coverslips coated with poly-L-lysine (Sigma, Saint Louis, MO, USA). Between 0.4 and 0.5 μg of plasmid DNA was transfected. Forty-eight hours post-transfection, the cells were fixed with 4% paraformaldehyde (PFA). In some cases, cells transfected with fluorescent chimeras were incubated for 10 min at room temperature with 5 μg mL−1 of WGA647 (Invitrogen) in PBS (Winkler Ltda., Santiago, Chile), washed and mounted with Dako mounting media (Dako, Carpenteria, CA, USA).
For immunodetection of wild-type receptors, cells were washed and permeabilized with 0.2% Triton-X100 (Sigma) for 10 min at room temperature, washed twice and blocked with PBS containing 1% BSA (Rockland Immunochemical, Gilbertsville, PA, USA) for 1 h at room temperature. Cells were incubated for 1 h at room temperature with primary antibodies: goat anti-CRF2 receptor (dilution 1:200; sc-1826, Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) and rabbit anti-D1 receptor (dilution 1:200; sc-14001, Santa Cruz Biotechnology, Inc.) in PBS–BSA solution. Then, cells were washed and incubated for 1 h with the following secondary antibodies: donkey anti-goat AlexaFluor488 and donkey anti-rabbit AlexaFluor546. Cells were washed and mounted with mounting media (Dako). In the case of immunofluorescence for D1 receptormyc/His, a rabbit anti-mouse antibody was used (dilution 1:200; ab18185, Abcam, Cambridge, MA, USA). For calnexin, a rabbit anti-calnexin was used (dilution 1:200, Sigma).
Confocal microscopy
Fluorescence images were captured with a confocal microscope (Fluoview 1000, Olympus, New York, NY, USA) and Fluoview v6.0 software (Olympus). The images were digitally obtained with a 100× objective (N.A 1.4 oil) and using a sequential mode of laser scanning. All the images were obtained without Gain and with Offset Zero. Captured images were processed with ImageJ software (Rasband, WS, ImageJ, NIH, http://rsbweb.nih.gov/ij/). The deconvolution analysis was performed as previously described (Blanco et al., 2011) with ‘Iterative Deconvolve’ plugins. The co-localization analysis was made with ‘JaCoP’ plugins. Pearson's coefficient was used to measure the overlap of the pixels in dual-channel images, reflecting the degree of dependency between two variables or fluorescent labels (Bolte and Cordelieres, 2006).
cAMP measurement
HEK293T cells (2 × 106) were grown in 60 mm plates and transiently transfected with 2 μg of DNA. Forty-eight hours post-transfection, cells were rapidly washed twice in HBSS (14065-056, Gibco), detached and resuspended in the same buffer with 100 μM IBMX (Sigma). Four thousand cells per well were stimulated with SKF83959 (Tocris Bioscience, Ellisville, MO, USA) or urocortin I (Sigma) prepared in HBSS supplemented with 100 μM IBMX, 5 mM HEPES and 2% DMSO or 100 μM IBMX, 5 mM HEPES and 0.1% BSA respectively. cAMP measurement were performed on 384-well plate using a homogenous time-resolved fluorescence (HTRF) cAMP dynamic 2 Kit (Cisbio International, Bagnols-sur-Cèze, France), according to the manufacturer's recommendations.
Protein extraction and immunoprecipitation
HEK293T (8 × 106) cells growing in a 100 mm dish were transiently transfected with D1 receptormyc/His, CRF2α receptorFlag, D1 receptormyc/His plus CRF2α receptorFlag and pcDNA3.1 using Lipofectamine 2000 (Invitrogen). Protein extracts were obtained by using the modified protocol of Burgueño et al. (2003). In brief, 48 h post-transfection, the cells were collected in PBS and washed twice with PBS. Then, the cells were resuspended in lysis buffer (50 mM Tris-HCl, pH 8.0, 150 mM NaCl, 1 mM EDTA) with protease inhibitors Complete Mini (Roche Diagnostics, Indianapolis, IN, USA), incubated for 20 min on ice and centrifuged at 19 500× g for 30 min at 4°C. The pellet was resuspended in RIPA (50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 0.25% deoxycholate, 1% NP-40, 1 mM EDTA, Millipore, Temecula, CA, USA) containing protease inhibitors, and homogenized through a piston sonicator (Cell Ultrasonic Disrrupter, Kontes, Vineland, NJ, USA) with two pulses of 5–10 s and then stood for 30 min on ice. Finally, the homogenate was centrifuged at 19 500× g for 30 min at 4°C. The soluble-rich membrane extracts were collected and the protein concentration determined with the Micro BCA™ Protein Assay Kit (Thermo Scientific, Rockford, IL, USA).
For inmunoprecipitation, the soluble-rich membrane extracts were pre-cleared with ‘TrueBlot Anti-Rabbit Ig IP Beads’ (eBioscience, San Diego, CA, USA). The samples were incubated with 0.8 μg rabbit anti-myc antibody (Ab9106, Abcam) according to the manufacturer's recommendations. Loading buffer 2× (8 M urea, 2% SDS, 100 mM DTT, 375 mM Tris, pH 6.8) was added to each sample. Immune complexes were dissociated by addition of DTT (to 25 mM) and heating to 37°C for 2 h and resolved by 8% SDS-PAGE with 8% urea. Proteins were transferred to nitrocellulose membranes and immunoblotted with mouse anti-Flag antibody (Stratagene, La Jolla, CA, USA), and then HRP-conjugated donkey anti-mouse IgG (dilution 1:5000, Jackson ImmnunoResearch Laboratories, Inc., West Grove, PA, USA). The immunoreactive bands were developed using a chemiluminescent detection kit (‘SuperSignal West Pico Chemiluminescent Substrate’, Thermo Scientific) according to the manufacturer's recommendations.
BRET assays
For BRET experiments, HEK293T cells transiently transfected with a constant amount (1 μg) of plasmid encoding CRF2α receptorRluc and increasing amounts (0.25–3 μg) of plasmids encoding for D1 receptorYFP, D2 receptorYFP or GABAB2 receptorYFP, were rapidly washed twice in HBSS (137 mM NaCl, 5.4 mM KCl, 0.3 mM Na2HPO4, 0.4 mM KH2PO4, 4.2 mM NaHCO3, 1.3 mM CaCl2, 0.5 mM MgCl2, 0.6 mM MgSO4, 5.6 mM glucose, pH 7.4) containing 10 mM glucose, detached and resuspended in the same buffer. To control the cell number, protein concentration was determined using Micro BCA™ Protein Assay Kit (Thermo Scientific). Cell suspensions (20 μg protein) were distributed in triplicate into 96-well, black microplates with a clear bottom (Corning 3651, Corning, Stockholm, Sweden) for fluorescence measurement or white plates with a solid bottom (Corning 3600, Corning) for BRET determination. For BRET measurement, benzyl-coelenterazine (NanoLight Technology, Pinetop, AZ, USA) was added at a final concentration of 5 μM, and readings were performed 1 min after using the POLARstar Optima plate-reader (BMG Labtech, Durham, NC, USA) that allows the simultaneous integration of the signals detected with two filter settings (475 ± 30 and 535 ± 30 nm). The BRET ratio was defined as previously described (Canals et al., 2003; Ciruela et al., 2004). In brief, the BRET signal was determined by calculating the ratio of the light emitted by yellow fluorescent protein (YFP) (510–560 nm) over the light emitted by Rluc (440–500 nm). The net BRET values were obtained by subtracting the BRET background signal detected when the Rluc-tagged construct was expressed alone. Curves were fitted using a non-linear regression and one-phase exponential association fit equation using the GraphPad Prism software (GraphPad Prism, San Diego, CA, USA).
FRET experiments
FRET between CRF2α receptorCFP and D1 receptorYFP in living cells was determined by donor recovery after acceptor photobleaching. If FRET occurs, then the bleaching of the acceptor (i.e. YFP) yields a significant increase in fluorescence of the donor [i.e. cyan fluorescent protein (CFP)] (Vilardaga et al., 2008). In brief, cells expressing CFP plus D1 receptorYFP, YFP plus CRF2α receptorCFP, CRF2α receptorCFP alone, CRF2α receptorCFP plus D1 receptorYFP or CRF2α receptorCFP plus D2 receptorYFP were mounted in an Attofluor holder (Warner Instruments, Hamden, CT, USA) and placed on an inverted Axio Observer microscope (Zeiss Microimaging, Oberkochen, Germany) equipped with a 63× oil immersion objective and a dual-emission photometry system (TILL Photonics, Gräfelfing, Germany). A Polychrome V (Till Photonics) was used as the light source and signals detected by photodiodes were digitized using a Digidata 1440A analogue/digital converter (Molecular Devices, Sunnyvale, CA, USA). pCLAMP (Molecular Devices) and GraphPad Prism software were used for data collection and analysis respectively. Therefore, upon excitation at 436 ± 10 nm [beam splitter dichroic long-pass (DCLP) 460 nm] and an illumination time set to 10 ms at 10 Hz, the emission light intensities were determined at 535 ± 15 nm (YFP) and 480 ± 20 nm (CFPpre) with a beam splitter DCLP of 505 nm. Subsequently, acceptor photobleaching was performed by direct illumination of YFP at 500 nm for 10 min. Finally, the emission intensities of YFP and CFP (CFPpost) were recorded again. FRET efficiency was calculated according to the equation: FRETefficiency = 1 − (CFPpre/CFPpost).
Calcium measurement
D1 receptors alone or combined with CRF2α receptors were transfected into HEK293T cells. Forty-eight hours after transfection, cells were loaded with 5 μM FURA-2 AM (Invitrogen) and 5 μM pluronic acid in HBSS (120 mM NaCl, 5.4 mM KCl, 0.8 mM MgCl2, 15 mM glucose, 20 mM HEPES, pH 7.4) plus 1.8 mM CaCl2 for 45 min at 37°C. Then, the medium was changed and allowed to de-esterify for 30 min at 37°C. Finally, the medium was changed to HCSS without calcium and fluorescence of FURA-2 AM was measured at 340 and 380 nm in a Spinning Disk confocal microscope (Olympus) (Leyton et al., 2014).
Statistical analysis
Results are presented as means ± SEM and analyses were carried out with GraphPad Prism v5.0 (GraphPad Software). Accordingly, Mann–Whitney test or one-way anova followed by Bonferroni post hoc test was used to determine significance.
Materials
SKF83959 [6-chloro-2,3,4,5-tetrahydro-3-methyl-1-(3-methylphenyl)-1H-3-benzazepine-7,8-diol hydrobomide] and SCH23390 [(R)-(+)-7-chloro-8-hydroxy-3-methyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine hydrochloride] were purchased from Tocris Biosciences (Bristol, UK). Urocortin I was purchased from Phoenix Peptides (Burlingame, CA, USA).
Results
Differential subcellular distribution of D1 and CRF2α receptors
The subcellular distribution of D1 and CRF2α receptors was evaluated. Accordingly, HEK293T cells were transiently transfected with D1 or CRF2α receptors tagged at their C-terminus with YFP and CFP (D1 receptorYFP and CRF2α receptorCFP) respectively. The fluorescence labelling of the individual cells was classified into surface (bright ring surrounding the cell) or intracellular (dense intracellular fluorescence) as previously done for α1A/B- and α1D-adrenoceptors respectively (Hague et al., 2004b).
As shown in Figure 1A (upper), D1 receptorYFP presented a mainly surface phenotype such as previously reported (O'Dowd et al., 2005). Quantitative analysis indicated that 77.4 ± 2.8% of the cells presented the surface phenotype. In contrast, CRF2α receptorCFP presented an intracellular phenotype (Figure 1A, bottom). Quantitative analysis indicated that 85.3 ± 2.7% of cells presented an intracellular phenotype. To remove the influence of the tagged fluorescence proteins in the subcellular distribution pattern of these receptors, immunofluorescence was performed in HEK293T cells transiently transfected with D1 or CRF2α receptors. As shown in Figure 1B, wild-type receptors showed the same phenotype as tagged receptors.
Figure 1.

Subcellular distribution pattern of D1 and CRF2α receptors expressed in HEK293T cells. (A) Fluorescence of chimeric proteins. D1 receptorYFP and CRF2α receptorCFP expressed in HEK293T-cells, fluorescence images of YFP and CFP were obtained by confocal microscopy. The upper panel shows the surface fluorescence of D1 receptorYFP, and the bottom panel shows the intracellular fluorescence of CRF2α receptorCFP. (B) Immunofluorescence of wild-type receptors. D1 receptor (left) and CRF2α receptor (right) were detected by immnunofluorescence. Six fields of cells per independent experiment (n = 3) were examined. Individual cells were classified as having either fluorescence almost exclusively in a bright ring surrounding the cell (surface), or dense intracellular fluorescence (intracellular). Data are expressed as mean ± SEM and represent results from about 200 cells. *P ≤ 0.05, Mann–Whitney test. Scale bar: 10 μm. (C–D) Concentration–response curves showing cAMP levels. Cells expressing D1 receptors were stimulated with increasing concentration of SKF83959 (C); data are expressed as mean ± SEM (n = 3). Cells expressing CRF2α receptors were stimulated with increasing concentration of urocortin (UCN)I (D), data are expressed as mean ± SEM (n = 6).
It has been reported that both D1 and CRF2α receptors signal through Gαs in HEK293T cells, increasing cAMP levels (Dautzenberg and Hauger, 2002; Neve et al., 2004; Gutknecht et al., 2008). In order to determine if the transfected receptors were functional in the conditions of the present study, we measured cAMP levels induced by respective agonists in HEK293 cells transfected with each receptor alone. We used the HTRF technique to measure cAMP levels induced by the presence of increasing concentrations of SKF83959, D1R agonist and of urocortin I, a CRH2R agonist. The concentration–response curves showed that activation of both receptors induced cAMP accumulation in a ligand concentration-dependent manner with a Log(EC50) = −9.122 ± 0.15 for SKF83959 (Fig. 1C) and Log(EC50) = −10.11 ± 0.29 for urocortin I (Figure 1D). To discount the possibility that cAMP accumulation is due to the activation of endogenous GPCRs present in HEK293T cells, we tested the effect of each ligand on HEK293T cells transfected only with the empty vector.
D1 and CRF2α receptors mutually modify their subcellular distribution
It has been reported that receptor heteromerization could change the dynamics and subcellular localization of the receptors involved (Terrillon and Bouvier, 2004). Thus, we evaluated the possibility that these receptors could be co-distributed when they are co-expressed in HEK293T cells. The co-expression of D1 and CRF2α receptors drastically changed their subcellular distribution pattern (Figure 2). D1 receptorsYFP acquired a higher intracellular phenotype in cells co-expressing CRF2α receptorCFP showing a pattern similar to CRF2α receptorCFP yielding a high degree of superposition of both labels (Figure 2A). Consistently, a Pearson's coefficient of 0.86 ± 0.01 was obtained indicating that both receptors exhibited a high degree of co-localization. Interestingly, in some cells that expressed only D1 receptorYFP, in the same preparation, the surface phenotype of D1 receptorYFP was preserved (Figure 2A, asterisk). These data indicate that CRF2α receptorCFP drags D1 receptorYFP to the intracellular compartment, suggesting the formation of a heteromeric complex between D1 and CRF2α receptors. Figure 2B shows the quantification of the percentage of cells showing surface or intracellular D1 receptorYFP phenotype when expressed alone or with CRF2α receptorCFP. A significant decrease in the percentage of cells expressing D1 receptorYFP in the surface and a concomitant increase in the intracellular compartment was observed when D1 receptorYFP was co-expressed with CRF2α receptorCFP. In order to evaluate the specificity of the CRF2α receptorCFP induced-retention of D1 receptorYFP in the intracellular compartment, HEK293T cells, were co-transfected either with CRF1 receptorYFP or CRF1 receptorYFP plus CRF2α receptorCFP. CRF1 receptorYFP expressed alone presented a surface phenotype (data not shown) and its distribution was not changed when both CRF1 receptorYFP and CRF2α receptorCFP were co-expressed (Figure 2C), further proving that the effect of CRF2α receptor over D1 receptor localization was specific.
Figure 2.

D1 receptorYFP and CRF2α receptorCFP mutually modify their subcellular distribution when they are co-expressed. (A) Co-expression of chimeric protein D1 receptorYFP and CRF2α receptorCFP in HEK293T cells. Six fields of cells per independent experiment (n = 3) were examined. D1 receptorYFP acquires an intracellular phenotype in cells co-expressing CRF2α receptorCFP, with high degree of co-distribution in the merge. The cells that only expressed D1 receptorYFP maintained their surface phenotype (asterisk). (B) Quantification of surface and intracellular phenotypes when D1 receptorYFP is expressed alone or with CRF2α receptorCFP. The percentage of cells with receptors at the cell surface is greater when D1 receptorYFP was expressed alone, lower when CRF2α receptorCFP was expressed alone, and an intermediate level was obtained when the receptors where co-expressed. Data are expressed as mean ± SEM and represent results from about 200 cells of three independent experiments (n = 3) *P ≤ 0.05, Mann–Whitney test. (C) Co-expression of CRF1 receptorYFP and CRF2α receptorCFP. The subcellular localization of both receptors is maintained.
Heteromerization of D1R and CRH2αR
To confirm that D1 and CRF2α receptors physically interact, co-immuneprecipitation, BRET and FRET assays were performed in HEK293T cells. D1 receptors and CRF2α receptors were shown to physically interact.
Co-immunoprecipitation experiments were performed, as a classical biochemical approach. Interestingly, CRF2α receptorFlag (band of ∼70 kDa) was able to co-immunoprecipate together with D1 receptormyc/His using a rabbit anti-myc polyclonal antibody, only from extracts of cells transiently transfected with D1 receptormyc/His plus CRF2α receptorFlag (Figure 3A, IP: lane 3). In addition, a higher molecular weight band (∼130 kDa) was observed. Importantly, these bands did not appear in immunoprecipitates of proteins extracted from cells transfected with D1 receptormyc/His (lane 1), CRF2α receptorFlag (lane 2) or the empty pcDNA vector (lane 4). Finally, the presence of D1 receptors in the crude extracts was confirmed by Western blot using a rabbit polyclonal antibody against c-myc (Figure 3A, right). Overall, these results showed that D1 and CRF2α receptors form a protein complex.
Figure 3.
Heteromerization of D1 receptors and CRF2α receptors. (A) Co-immunoprecipitation of D1 receptormyc/His and CRF2α receptorFlag from HEK293T cells. Solubilized extracts from cells expressing D1 receptormyc/His (lane 1), CRF2α receptorFlag (lane 2), D1 receptormyc/His plus CRF2α receptorFlag (lane 3) or empty pcDNA 3.1 vector (lane 4) were immunoprecipitated with a rabbit anti-myc polyclonal antibody. Extracts (crude) and immunoprecipitates (IP) were analysed using a mouse anti-Flag and a rabbit anti c-myc antibodies. CRF2α receptorFlag immunoprecipitated with D1 receptormyc/His (IP, lane 3). The presence of D1 receptormyc/His was corroborated (lanes 1 and 3). (B) BRET saturation curve. BRET was measured in HEK293T cells co-expressing CRF2α receptorRluc plus D1 receptorYFP (blue squares), CRF2α receptorRluc plus D2 receptorYFP (red circles) or CRF2α receptorRluc plus GABAB2 receptorYFP (black circles). Co-transfections were performed with increasing amounts of the YFP-tagged vectors while the CRF2α receptorRluc was maintained constant. Plotted on the X-axis is the fluorescence value obtained from the YFP, normalized to the luminescence value of CRF2α receptorRluc 10 min after h-coelenterazine incubation. (C) Determination of the D1 and CRF2α receptor oligomerization by FRET experiments in living cells. D1 receptorYFP and CRF2α receptorCFP were expressed in HEK293T cells, and fluorescence images of CFP and YFP were recorded before (pre) and after (post) the YFP was photobleached by 5 min of exposure to light at 500 nm to corroborate the extent of acceptor photodestruction (left panel). Emission intensities of CRF2α receptorCFP (480 nm, blue) and D1 receptorYFP (535 nm, yellow) from single cells expressing both CRF2α receptorCFP and D1 receptorYFP were recorded before and after YFP photobleaching (right panel) to determine the FRET efficiency. Scale bar: 10 μm. (D) Quantification of the FRET efficiency of different FRET pairs: CFP plus D1 receptorYFP (n = 5), YFP plus CRH2αRCFP (n = 10), CRH2αRCFP (n = 5), D1RYFP plus CRF2α receptorCFP (n = 15) and D2 receptorYFP plus CRF2α receptorCFP (n = 10). The data indicate the mean ± SEM. An asterisk denotes the data that are significantly different from the control FRET pairs (i.e., CFP or YFP co-transfections). *P < 0.001, anova with a Bonferroni multiple comparison post hoc test.
Next, the ability of D1 and CRF2α receptors to heteromerize was evaluated using biophysical approaches. Accordingly, the degree of D1/CRF2α receptor interaction was analysed by means of BRET experiments (Figure 3B). Thus, a BRET saturation curve was constructed in HEK293T cells co-transfected with a constant amount of CRF2α receptorRluc and increasing concentrations of D1 receptorYFP, D2 receptorYFP or GABAB2 receptorYFP. Interestingly, a positive BRET signal was obtained between CRF2α receptorRluc and D1 receptorYFP. As shown in Figure 3B, the BRET ratio between CRF2α receptorRluc and D1 receptorYFP increased as a hyperbolic function of the concentration of the D1 receptorYFP. Importantly, the pairing of CRF2α receptorRluc with D2 receptorYFP led to a non-significant BRET signal, which was within the same range as that observed for the CRF2α receptorRluc/GABAB2 receptorYFP pair (i.e. non-specific), thus supporting the specificity of the CRF2α receptorRluc/D1 receptorYFP interaction.
Additionally, the formation of D1/CRF2α receptor heteromers was also assessed by means of FRET experiments. To this end, cells were transiently transfected with D1 receptorYFP and CRF2α receptorCFP, and their ability to heteromerize was determined by calculating the FRET efficiency using the donor recovery after acceptor bleaching approach (Fig. 3C). The formation of D1/CRF2α receptor heteromers is indicated by the resonance energy transfer between the fluorescent proteins, which was measured by the recovery of the CFP emission after photobleaching of YFP (Figure 3C). The application of the photobleaching protocol to cells expressing only CRF2α receptorCFP did not modify the emission intensity of CFP (Figure 3D). As additional controls, to ensure that the FRET signal was due to a specific interaction and not to a random proximity of the fluorophores, FRET efficiency was determined in cells co-expressing CRF2α receptorCFP plus YFP, D1 receptorYFP plus CFP or CRF2α receptorCFP plus D2 receptorYFP, at comparable fluorescence levels. Under these experimental conditions the FRET efficiency of the CRF2α receptorCFP/D1 receptorYFP pair was significantly higher (9.65 ± 1.07; P < 0.001) than that observed in the corresponding negative controls (i.e. CRF2α receptorCFP/YFP and CFP/D1 receptorYFP pairs) (Figure 3D). Interestingly, the FRET efficiency of the CRF2α receptorCFP and D2 receptorYFP pair was not significantly different from that observed for the other negative controls used (Figure 3D). Overall, these results strongly suggest that D1 and CRF2α receptors physically interact in living cells.
D1 receptor and CRF2α receptor form a stable heteromer
In order to further test the capacity of D1 and CRF2α receptors to physically interact and the stability of the heteromer, we used the heteromer mobilization strategy described by O'Dowd et al. (2005) associated with immunofluorescence for WGA647, a cell surface marker. Briefly, this strategy involves the incorporation of a NLS to a given GPCR such that the GPCR acquires the unique property of being retained in the intracellular compartment in the absence of antagonist, and in the cell surface in the presence of inverse agonist/antagonist. Thus, the heteromeric interaction and its stability are evaluated analysing the co-trafficking of both receptors to the different subcellular compartments. As expected, the incorporation of the NLS sequence reported by O'Dowd et al. (2005) to D1 receptorYFP (D1 receptor-NLSYFP) conferred a mainly intracellular phenotype for D1 receptor-NLSYFP (≈90% of the cells, data not shown).
The use of immunofluorescence with WGA647 as marker of the cell surface confirmed that D1 receptorYFP fluorescence is mainly present at the cell surface meanwhile CRF2α receptorCFP is present mainly intracellularly (Figure 4A). Figure 4B shows the results obtained with the strategy of O'Dowd et al. (2005) co-expressing D1 receptorYFP plus CRF2α receptorCFP (Fig. 4Ba–c), D1 receptor-NLSYFP plus CRF2α receptorCFP (Figure 4Bd–f) and of D1 receptor-NLSYFP plus CRF2α receptorCFP in the presence of 1 μM SCH23390 (Figure 4Bg–i). Colocalization analysis showed that the incorporation of NLS to D1 receptorYFP decreased Pearson's correlation coefficient between CFP and WGA647 from 0.37 ± 0.02 to 0.02 ± 0.01, and the presence of 1 μM SCH23390 restored the cell surface expression of CRF2α receptorCFP up to a Pearson's correlation coefficient of 0.27 ± 0.02 (Figure 4C).
Figure 4.

D1 and CRF2α receptors form a stable heteromeric complex in HEK293T cells. (A) Co-localization of CRF2α receptorCFP with the plasma membrane marker WGA647, when it was co-expressed with: D1 receptorYFP (a–c), D1 receptor-NLSYFP (d–f) or D1 receptor-NLSYFP treated with SCH23390 (1 μM) (g–i). The number of CRF2α receptors in the plasma membrane was augmented when it was co-expressed with D1 receptorYFP (a–c). When co-transfected with D1 receptor-NLSYFP, the CRF2α receptorCFP co-trafficked with the D1 receptor-NLSYFP to the intracellular compartment, being excluded from the plasma membrane (d–f). The treatment with SCH23390 (1 μM) to the cells co-transfected with D1 receptor-NLSYFP and CRF2α receptorCFP allowed both receptors to be observed at the cell surface (g–i). (B) Pearson's correlation coefficient between CFP and WGA647. In each condition, the Pearson's coefficient was calculated from the different transfection conditions. Data are expressed as mean ± SEM and represent results from 18 to 30 cells of two independent experiments. (C) Quantification of the percentage of cells showing a surface phenotype for CRF2α receptors. The percentage of cells was calculated in the different transfection conditions. Data are expressed as mean ± SEM and represent results from about 200 cells from three independent experiments (n = 3) *P ≤ 0.05, Mann–Whitney test.
The percentage of cells with CRF2α receptorCFP at the cell surface was estimated comparing the localization of CFP fluorescence with WGA647. When both receptors were co-expressed 41.8 ± 4.8% of cells presented CRF2α receptorCFP at the cell surface (Figure 4D). The incorporation of NLS to D1 receptorYFP (D1 receptor-NLSYFP) significantly decreased the percentage of cells with CRF2α receptorCFP at the cell surface indicating that D1 receptor-NLSYFP was physically interacting with CRF2α receptorCFP and was therefore able to drag CRF2α receptorCFP forcing the intracellular localization induced by NLS. The formation of a stable heteromer was further demonstrated in the presence of 1 μM SCH23390, a specific D1 receptor antagonist/inverse agonist that is able to retain D1 receptor-NLSYFP at the cell surface. Indeed, in the presence of 1 μM SCH23390 a significant recovery in the percentage of cells expressing CRF2α receptorCFP at the cell surface was observed (Figure 4D). These data show that D1 receptorYFP and CRF2α receptorCFP form a stable heteromer.
As the intracellular fluorescence given by the D1 /CRF2α receptor heteromer looks like the cell reticular compartment, we analysed the colocalization of the receptors with calnexin, a marker of the cellular reticular compartment. As can be seen in Figure 5A, cells transfected with D1 receptormyc/His showed a poor colocalization with calnexin that significantly increased when D1 receptormyc/His and CRF2α receptor were co-expressed (Figure 5B). In contrast, immunofluorescence against CRF2α receptor showed high colocalization with calnexin (Figure 5A) that did not significantly change when both receptors were co-expressed (Figure 5C).
Figure 5.

Colocalization of D1 and CRF2α receptors with calnexin, a marker of the cellular reticular compartment. (A) Immunofluoresence of D1 receptormyc/His and CRF2α receptors expressed alone or co-expressed in HEK293T cells and of calnexin. (B) Pearson's correlation coefficient between D1 receptormyc/His and calnexin in the presence or absence of CRF2α receptors (P < 0.001; Mann–Whitney test). (C) Pearson's correlation coefficient between CRF2α receptors and calnexin in the presence or absence of D1 receptormyc/His.
D1/CRF2α receptor heteromer and cAMP signalling
In order to test whether the heteromerization between D1 and CRF2α receptors switch their cAMP signalling mode, we measured cAMP induced by increasing concentrations of SKF83959 (D1 receptor agonist; Figure 6A) and urocortin I (CRF2 receptor agonist; Figure 6B) in HEK293T cells transfected with D1 receptors, CRF2α receptors and D1 plus CRF2α receptors. As expected, the presence of SKF83959 induced a concentration-dependent increase in cAMP in D1 receptor, but not CRF2α receptor, expressing cells (Figure 6A). The D1 receptor specificity of the response to SKF83959 was confirmed because HEK293T cells transfected with the empty vector did not show a response. Interestingly, SKF83959 induced the same level of response when both receptors were co-expressed. The presence of 10 μM SCH23390 displaced the SKF83959 concentration–response curve to the right obtained with both, D1 receptor and D1 plus CRF2α receptor expressing cells further confirming the specificity of the response (Figure 6A).
Figure 6.

Concentration–response curves for cAMP accumulation. HEK293T cells expressing D1 receptors and CRF2α receptor alone and cells co-expressing D1 plus CRF2α receptors were incubated in the presence of different concentrations of SKF83959 (A) or urocortin (UCN) I (B) with or without 10 μM SCH23390 (SCH). The results are the mean ± SEM of three independent experiments.
The presence of urocortin I induced a concentration-dependent increase in cAMP in CRF2α receptor, but not D1 receptor expressing cells (Figure 6B). The CRF2α receptor specificity of the response to urocortin I was confirmed because HEK293T cells transfected with the empty vector did not show a response. Similar to what was observed with SKF83959, the presence of urocortin I induced the same level of response when both receptors were co-expressed (Figure 6B). The presence of 10 μM SCH23390 displaced the SKF83959 concentration–response curve to the right obtained with both D1 receptor and D1 plus CRF2α receptor-expressing cells further confirming the specificity of the response (Figure 6A). To test a possible crosstalk between both receptors, we determined cAMP accumulation induced by urocortin I in the presence of SCH23390. As can be seen in Figure 6B, the presence of 10 μM SCH23390 did not modify the urocortin I concentration–response curve. These data indicate that the heteromerization between both receptors does not modify cAMP signalling from each individual receptor.
D1/CRF2α receptor heteromer activation by dopamine agonists triggers calcium mobilization
HEK293T cells transfected with only D1 receptors or D1 plus CRF2α receptors were loaded with FURA-2 AM and intracellular calcium mobilization was measured in a calcium-free medium. The addition of 10 μM SKF83959, a D1 receptor agonist, to cells expressing only D1 receptors triggered calcium mobilization in just one of the tested cells (n = 67), representing 1.5% of them. However, the addition of 10 μM SKF83959 to cells co-transfected with D1 plus CRF2α receptors, triggered calcium mobilization in 36.7% of the tested cells (n = 71) (Figure 7A–B). Mobilization of intracellular calcium in response to the ionophore ionomycin was measured in each tested cell to determine cell responsiveness (data not shown).
Figure 7.

Calcium mobilization induced by the activation of D1 receptors. HEK293T cells expressing D1 receptors alone or D1 plus CRF2α receptors were loaded with FURA-2 AM. Intracellular calcium mobilization was measured in living cells at 340 and 380 nm in a confocal microscope and time lapse of the 340/380 ratio was plotted. The selective D1 receptor agonist, SKF83959, generated calcium mobilization when the receptors were co-expressed (n = 71), but no effect was observed when D1 receptors were expressed alone (n = 67). The data were obtained from four independent experiments.
Discussion and conclusions
In the present study, we show that D1 receptors and CRF2α receptors are capable of forming a stable heteromer in living cells, modifying their subcellular localization and signalling. Each receptor has a distinct subcellular localization, the D1 receptor being mainly at the cell surface and CRF2α receptor mostly intracellular. Interestingly, when both receptors were co-expressed, a balanced subcellular localization for both receptors was observed, thus suggesting that D1 and CRF2α receptors heteromerize. We demonstrated the existence of D1/CRF2α receptor heteromers by co-immunoprecipitation, BRET and FRET assays. Furthermore, we show that D1 and CRF2α receptors form a stable heteromer using the heteromer mobilization strategy described by O'Dowd et al. (2005). In addition, we showed that the heteromerization of D1 receptors with CRF2α receptors switches the signalling of D1 receptors from a receptor that does not mobilize intracellular calcium to a receptor that mobilizes intracellular calcium. Thus, our results indicate that the heteromerization of D1 receptors and CRF2α receptors has at least two fundamental functional implications: (1) change in the subcellular localization of both receptors and (2) change in the signalling cascade associated with D1 receptor stimulation.
Our results show that transfected CRF2α receptors were mainly located intracellularly (≈85%); similar to what is has been observed in neurons of the rat dorsal raphe nucleus (Waselus et al., 2009). There are other GPCRs that are constitutively intracellular such as GABAB1 receptors (Margeta-Mitrovic et al., 2000), CB1 receptors (Andersson et al., 2003), α1D-adrenoceptors (Hague et al., 2004a) and 5-HT2A/C (Magalhaes et al., 2010) receptors. Interestingly, Markovic et al. (2008) have shown that the β isoform of the CRF2 receptor is constitutively present in the cell surface, in contrast to what we observed with the CRF2α receptor. The α and β isoforms of the CRF2 receptor differ in their N-terminal sequence (Grigoriadis et al., 1996). It has been shown that the rat CRF2α receptor has a non-cleavable pseudo signal peptide of 19-aminoacids in the N-terminal (Rutz et al., 2006), and that the lack of its cleavage affects the expression and traffic of CRF2α receptors to the plasma membrane (Schulz et al., 2010; Teichmann et al., 2012). Interestingly, Teichmann et al. (2012) have shown that the rat CRF2α receptor does not dimerize and have determined that this is due to its non-cleavable N-terminal pseudo signal peptide. Our data suggest that the presence of the pseudo signal peptide is not an impediment for the heteromerization of CRF2α receptors with a class A GPCR such as the D1 receptor. The 19-amino acid N-terminal segment of the CRF2α receptor is highly conserved among several species. Therefore, it is not surprising that the human CRF2α receptor studied herein is also found mainly intracellularly. Other receptors like α1D-adrenoceptors (Hague et al., 2004a) and CB1 receptors (Andersson et al., 2003) also have an intracellular localization that depends on their N-terminal. As stated before, the CRF2 receptor was found predominantly in the cytoplasm in neurons of the rat dorsal raphe nucleus (Waselus et al., 2009). Even though the CRF2 receptor antibody used by Waselus et al. (2009) might not discriminate between both CRF2 receptor isoforms (Lukkes et al., 2011), their study relates to the CRF2α receptor, as it has been shown the CRF2α isoform is present in neurons and the CRF2β receptor in non-neuronal cells in the CNS (Chalmers et al., 1995; Lovenberg et al., 1995; Dautzenberg and Hauger, 2002). In contrast to CRF2α receptor, and as has been previously shown (Vickery and von Zastrow, 1999; O'Dowd et al., 2005; Sun et al., 2009), the transfected D1 receptor presented a surface phenotype.
Our results show that the heteromerization between D1 and CRF2α receptors alters the subcellular localization of both receptors. The need for a GPCR partner to traffic to another subcellular compartment has been previously demonstrated for other GPCRs (Kaupmann et al., 1998; Margeta-Mitrovic et al., 2000). We observed that the CRF2α receptor decreases the surface phenotype of the D1 receptor to an intermediate surface/intracellular phenotype. There is evidence that when the D1 receptor heteromerizes with the D2 receptor, the D1 receptor also acquires an intermediate phenotype (So et al., 2005). In this case, the heteromerization increases the presence of D2 receptors and decreases the presence of D1 receptors in the cell surface. We also showed that heteromerization between D1 and CRF2α receptors promotes the trafficking of CRF2α receptors to the plasma membrane. Likewise, the D1 receptor improves the trafficking of μ-opioid receptors (Juhasz et al., 2008) and α1B-adrenoceptor improves the trafficking of α1D-adrenoceptors to the cell surface (Hague et al., 2004a). Thus, the aforementioned findings, as well as the present study, suggest that the increased trafficking of CRF2α receptors to the cell surface and of D1 receptors to the intracellular compartment depends on the interaction between them. It is tempting to speculate that the changes in subcellular localization of CRF2 receptors observed in the dorsal raphe after stress could be due to its heteromerization with another GPCR (Waselus et al., 2009).
The heteromer mobilization strategy described by O'Dowd et al. (2005) allowed us to further prove that D1 and CRF2α receptors are capable of forming a stable heteromer. In the presence of the NLS sequence in the D1 receptor, co-expressed D1 receptor-NLS and the CRF2α receptor function as a unit present at the cell surface and internalizes as such, in the presence or absence of the D1 receptor antagonist respectively.
Finally, we demonstrated that the heteromerization of D1 with CRF2α receptors conferred the property of mobilizing intracellular calcium. Cells expressing D1 receptors alone are not capable of mobilizing calcium. In contrast, when the D1 receptor is co-expressed with the CRF2α receptor, intracellular calcium mobilization is induced by the presence of a D1 receptor agonist. Interestingly, our results show a strong parallel between the heteromerization of D1 with D2 receptors and of D1 with CRF2α receptors, not only at the subcellular distribution level mentioned earlier, but also at the functional level. Similar to what we observed for D1 and CRF2α receptors, it has been shown that the capacity of D1 receptors to mobilize intracellular calcium depends on the formation of a stable D1/D2 receptor heteromer (Lee et al., 2004; Hasbi et al., 2009; 2010,). It has been shown that the change in signalling pathway of the D1 receptor in the D1/D2 receptor heteromer is due to a change in the GPCRs, from Gαs to Gαq/11 (Lee et al., 2004; Rashid et al., 2007; Hasbi et al., 2010; Verma et al., 2010). However, there is also evidence that the change in D1 receptor signalling may also be due to downstream signalling pathways that do not depend on the heteromerization (Chun et al., 2013). There is also evidence of other GPCRs in which the heteromerization switches the GPCRs (George et al., 2000; Mellado et al., 2001; Charles et al., 2003; Ferrada et al., 2009). Thus, it is possible that the calcium mobilization associated with the activation of D1 receptors when D1 receptors are co-expressed with CRF2α receptors is due to a change in the GPCRs. Further studies should address whether the change in intracellular calcium mobilization induced by the activation of D1 receptors co-expressed with CRF2α receptors is due to a change in its affinity for Gq (Hasbi et al., 2009) or is due to a downstream crosstalk (Chun et al., 2013), as has been shown for the D1/D2 receptor heteromer.
The functional implications of the existence of D1/ CRF2α receptor heteromers in vivo are presently unknown. However, there are several anatomical substrates such as the prefrontal cortex, amygdala, bed nucleus of the stria terminalis, lateral septum and VTA where these two receptors could be co-expressed in neurons and/or nerve terminals (Corominas et al., 2010; Gysling, 2012). It is tempting to speculate that the D1/CRF2α receptor heteromer could be responsible for the potentiation of glutamatergic transmission from the basolateral amygdala to medial prefrontal cortex (Orozco-Cabal et al., 2008) and for the switch from inhibition to facilitation of CRF2α receptors observed in the lateral septum of rats repeatedly treated with cocaine (Liu et al., 2005).
In summary, we showed that D1 and CRF2αR receptors are capable of forming a stable heteromer with functional implications, such as changes in the subcellular localization of both receptors and in the signalling of D1 receptors. We would like to suggest that the existence of a D1/CRF2α receptor heteromer might explain, at least in part, the complex physiological interactions established between dopamine and CRH in normal and pathological conditions such as addiction, representing a new potential pharmacological target.
Acknowledgments
Supported by FONDECYT (grants N° 1070340, 1110392, 7070246 and 7080124); and Millenium Science Initiative MSI (grants N° P06/008-F and P10/063-F). K. A. A., E. H. B. and P. G. S. were recipients of doctoral fellowships from CONICYT, Chile. K. A. A. was a recipient of a travel award from CONICYT. This work was also supported by grants SAF2011-24779 and Consolider-Ingenio CSD2008-00005 from MINECO and ICREA Academia-2010 from the Catalan Institution for Research and Advanced Studies (ICREA) to F. C. We thank the collaboration of Ms Cledi C Cerda for the cAMP accumulation studies. We acknowledge FONDECYT grant N° 1130079, which made it possible to perform the cAMP accumulation studies.
Glossary
- CFP
cyan fluorescent protein
- CRH
corticotrophin-releasing hormone
- CRF2α receptor
corticotrophin-releasing hormone type-2α receptor
- YFP
yellow fluorescent protein
List of author contributions
J. F., P. G., K. A. A. and P. G. S. designed and performed experiments and analysed data. E. H. B. designed experiments and advised on the quantitative analysis of protein expression studies. J. M. C. advised on the design and analysis of calcium signalling studies. J. F., P. G. and P. G. S. wrote the manuscript. F. C. directed and coordinated the FRET and BRET experiments. K. G. directed and coordinated the transfection and signalling studies. All authors contributed on the editing of the manuscript.
Conflicts of interest
The authors declare no conflict of interest.
References
- Alexander SP, Benson HE, Faccenda E, Pawson AJ, Sharman JL, McGrath JC, et al. The concise guide to PHARMACOLOGY 2013/14: overview. Br J Pharmacol. 2013a;170:1449–1458. [Google Scholar]
- Alexander SP, Benson HE, Faccenda E, Pawson AJ, Sharman JL, McGrath JC, et al. The concise guide to PHARMACOLOGY 2013/14: G-protein coupled receptors. Br J Pharmacol. 2013b;170:1459–1581. doi: 10.1111/bph.12445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Andersson H, D'Antona AM, Kendall DA, Von Heijne G, Chin CN. Membrane assembly of the cannabinoid receptor 1: impact of a long N-terminal tail. Mol Pharmacol. 2003;64:570–577. doi: 10.1124/mol.64.3.570. [DOI] [PubMed] [Google Scholar]
- Blanco EH, Zuñiga JP, Andres ME, Alvarez AR, Gysling K. Corticotropin-releasing factor binding protein enters the regulated secretory pathway in neuroendocrine cells and cortical neurons. Neuropeptides. 2011;45:273–279. doi: 10.1016/j.npep.2011.05.002. [DOI] [PubMed] [Google Scholar]
- Bolte S, Cordelieres FP. A guided tour into subcellular colocalization analysis in light microscopy. J Microsc. 2006;224:213–232. doi: 10.1111/j.1365-2818.2006.01706.x. [DOI] [PubMed] [Google Scholar]
- Burgueño J, Enrich C, Canela EI, Mallol J, Lluis C, Franco R, et al. Metabotropic glutamate type 1alpha receptor localizes in low-density caveolin-rich plasma membrane fractions. J Neurochem. 2003;86:785–791. doi: 10.1046/j.1471-4159.2003.01842.x. [DOI] [PubMed] [Google Scholar]
- Cadet JL, Brannock C, Ladenheim B, McCoy MT, Krasnova IN, Lehrmann E, et al. Enhanced upregulation of CRH mRNA expression in the nucleus accumbens of male rats after a second injection of methamphetamine given thirty days later. PLoS ONE. 2014;9:e84665. doi: 10.1371/journal.pone.0084665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Canals M, Marcellino D, Fanelli F, Ciruela F, de Benedetti P, Goldberg SR, et al. Adenosine A2A-dopamine D2 receptor-receptor heteromerization: qualitative and quantitative assessment by fluorescence and bioluminescence energy transfer. J Biol Chem. 2003;278:46741–46749. doi: 10.1074/jbc.M306451200. [DOI] [PubMed] [Google Scholar]
- Chalmers DT, Lovenberg TW, De Souza EB. Localization of novel corticotropin-releasing factor receptor (CRF2) mRNA expression to specific subcortical nuclei in rat brain: comparison with CRF1 receptor mRNA expression. J Neurosci. 1995;15:6340–6350. doi: 10.1523/JNEUROSCI.15-10-06340.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Charles AC, Mostovskaya N, Asas K, Evans CJ, Dankovich ML, Hales TG. Coexpression of delta-opioid receptors with micro receptors in GH3 cells changes the functional response to micro agonists from inhibitory to excitatory. Mol Pharmacol. 2003;63:89–95. doi: 10.1124/mol.63.1.89. [DOI] [PubMed] [Google Scholar]
- Chun LS, Free RB, Doyle TB, Huang XP, Rankin ML, Sibley DR. D1-D2 dopamine receptor synergy promotes calcium signaling via multiple mechanisms. Mol Pharmacol. 2013;84:190–200. doi: 10.1124/mol.113.085175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ciruela F, Burgueño J, Casadó V, Canals M, Marcellino D, Goldberg SR, et al. Combining mass spectrometry and pull-down techniques for the study of receptor heteromerization. Direct epitope-epitope electrostatic interactions between adenosine A2A and dopamine D2 receptors. Anal Chem. 2004;76:5354–5363. doi: 10.1021/ac049295f. [DOI] [PubMed] [Google Scholar]
- Ciruela F, Fernández-Dueñas V, Llorente J, Borroto-Escuela D, Cuffí ML, Carbonell L, et al. G protein-coupled receptor oligomerization and brain integration: focus on adenosinergic transmission. Brain Res. 2012;1476:86–95. doi: 10.1016/j.brainres.2012.04.056. [DOI] [PubMed] [Google Scholar]
- Corominas M, Roncero C, Casas M. Corticotropin releasing factor and neuroplasticity in cocaine addiction. Life Sci. 2010;86:1–9. doi: 10.1016/j.lfs.2009.11.005. [DOI] [PubMed] [Google Scholar]
- Dautzenberg FM, Hauger RL. The CRF peptide family and their receptors: yet more partners discovered. Trends Pharmacol Sci. 2002;23:71–77. doi: 10.1016/s0165-6147(02)01946-6. [DOI] [PubMed] [Google Scholar]
- Ferrada C, Moreno E, Casadó V, Bongers G, Cortés A, Mallol J, et al. Marked changes in signal transduction upon heteromerization of dopamine D1 and histamine H3 receptors. Br J Pharmacol. 2009;157:64–75. doi: 10.1111/j.1476-5381.2009.00152.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- George O, Le Moal M, Koob GF. Allostasis and addiction: role of the dopamine and corticotropin-releasing factor systems. Physiol Behav. 2012;106:58–64. doi: 10.1016/j.physbeh.2011.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- George SR, Fan T, Xie Z, Tse R, Tam V, Varghese G, et al. Oligomerization of mu- and delta-opioid receptors. Generation of novel functional properties. J Biol Chem. 2000;275:26128–26135. doi: 10.1074/jbc.M000345200. [DOI] [PubMed] [Google Scholar]
- Grigoriadis DE, Lovenberg TW, Chalmers DT, Liaw C, De Souze EB. Characterization of corticotropin-releasing factor receptor subtypes. Ann N Y Acad Sci. 1996;780:60–80. doi: 10.1111/j.1749-6632.1996.tb15112.x. [DOI] [PubMed] [Google Scholar]
- Guan X, Wan R, Zhu C, Li S. Corticotropin-releasing factor receptor type-2 is involved in the cocaine-primed reinstatement of cocaine conditioned place preference in rats. Behav Brain Res. 2014;258:90–96. doi: 10.1016/j.bbr.2013.10.019. [DOI] [PubMed] [Google Scholar]
- Gutknecht E, Hauger RL, Van der Linden I, Vauquelin G, Dautzenberg FM. Expression, binding, and signaling properties of CRF2(a) receptors endogenously expressed in human retinoblastoma Y79 cells: passage-dependent regulation of functional receptors. J Neurochem. 2008;104:926–936. doi: 10.1111/j.1471-4159.2007.05052.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gysling K. Relevance of both type-1 and type-2 corticotropin releasing factor receptors in stress-induced relapse to cocaine seeking behaviour. Biochem Pharmacol. 2012;83:1–5. doi: 10.1016/j.bcp.2011.07.101. [DOI] [PubMed] [Google Scholar]
- Hague C, Chen Z, Pupo AS, Schulte NA, Toews ML, Minneman KP. The N terminus of the human alpha1D-adrenergic receptor prevents cell surface expression. J Pharmacol Exp Ther. 2004a;309:388–397. doi: 10.1124/jpet.103.060509. [DOI] [PubMed] [Google Scholar]
- Hague C, Uberti MA, Chen Z, Hall RA, Minneman KP. Cell surface expression of alpha1D-adrenergic receptors is controlled by heterodimerization with alpha1B-adrenergic receptors. J Biol Chem. 2004b;279:15541–15549. doi: 10.1074/jbc.M314014200. [DOI] [PubMed] [Google Scholar]
- Hahn J, Hopf FW, Bonci A. Chronic cocaine enhances corticotropin-releasing factor-dependent potentiation of excitatory transmission in ventral tegmental area dopamine neurons. J Neurosci. 2009;29:6535–6544. doi: 10.1523/JNEUROSCI.4773-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hasbi A, Fan T, Alijaniaram M, Nguyen T, Perreault ML, O'Dowd BF, et al. Calcium signaling cascade links dopamine D1–D2 receptor heteromer to striatal BDNF production and neuronal growth. Proc Natl Acad Sci U S A. 2009;106:21377–21382. doi: 10.1073/pnas.0903676106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hasbi A, O'Dowd BF, George SR. Heteromerization of dopamine D2 receptors with dopamine D1 or D5 receptors generates intracellular calcium signaling by different mechanisms. Curr Opin Pharmacol. 2010;10:93–99. doi: 10.1016/j.coph.2009.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heckman KL, Pease LR. Gene splicing and mutagenesis by PCR-driven overlap extension. Nat Protoc. 2007;2:924–932. doi: 10.1038/nprot.2007.132. [DOI] [PubMed] [Google Scholar]
- Juhasz JR, Hasbi A, Rashid AJ, So CH, George SR, O'Dowd BF. Mu-opioid receptor heterooligomer formation with the dopamine D1 receptor as directly visualized in living cells. Eur J Pharmacol. 2008;581:235–243. doi: 10.1016/j.ejphar.2007.11.060. [DOI] [PubMed] [Google Scholar]
- Kaupmann K, Malitschek B, Schuler V, Heid J, Froestl W, Beck P, et al. GABA(B)-receptor subtypes assemble into functional heteromeric complexes. Nature. 1998;396:683–687. doi: 10.1038/25360. [DOI] [PubMed] [Google Scholar]
- Koob GF. A role for brain stress systems in addiction. Neuron. 2008;59:11–34. doi: 10.1016/j.neuron.2008.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee SP, So CH, Rashid AJ, Varghese G, Cheng R, Lanca AJ, et al. Dopamine D1 and D2 receptor Co-activation generates a novel phospholipase C-mediated calcium signal. J Biol Chem. 2004;279:35671–35678. doi: 10.1074/jbc.M401923200. [DOI] [PubMed] [Google Scholar]
- Leyton V, Goles NI, Fuenzalida-Uribe N, Campusano JM. Octopamine and dopamine differentially modulate the nicotine-induced calcium response in Drosophila mushroom body Kenyon cells. Neurosci Lett. 2014;560:16–20. doi: 10.1016/j.neulet.2013.12.006. [DOI] [PubMed] [Google Scholar]
- Liu J, Yu B, Orozco-Cabal L, Grigoriadis DE, Rivier J, Vale WW, et al. Chronic cocaine administration switches corticotropin-releasing factor2 receptor-mediated depression to facilitation of glutamatergic transmission in the lateral septum. J Neurosci. 2005;25:577–583. doi: 10.1523/JNEUROSCI.4196-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lovenberg TW, Chalmers DT, Liu C, De Souza EB. CRF2 alpha and CRF2 beta receptor mRNAs are differentially distributed between the rat central nervous system and peripheral tissues. Endocrinology. 1995;136:4139–4142. doi: 10.1210/endo.136.9.7544278. [DOI] [PubMed] [Google Scholar]
- Lukkes JL, Staub DR, Dietrich A, Truitt W, Neufeld-Cohen A, Chen A, et al. Topographical distribution of corticotropin-releasing factor type 2 receptor-like immunoreactivity in the rat dorsal raphe nucleus: co-localization with tryptophan hydroxylase. Neuroscience. 2011;183:47–63. doi: 10.1016/j.neuroscience.2011.03.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Magalhaes AC, Holmes KD, Dale LB, Comps-Agrar L, Lee D, Yadav PN, et al. CRF receptor 1 regulates anxiety behavior via sensitization of 5-HT2 receptor signaling. Nat Neurosci. 2010;13:622–629. doi: 10.1038/nn.2529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Margeta-Mitrovic M, Jan YN, Jan LY. A trafficking checkpoint controls GABA(B) receptor heterodimerization. Neuron. 2000;27:97–106. doi: 10.1016/s0896-6273(00)00012-x. [DOI] [PubMed] [Google Scholar]
- Markovic D, Punn A, Lehnert H, Grammatopoulos DK. Intracellular mechanisms regulating corticotropin-releasing hormone receptor-2beta endocytosis and interaction with extracellularly regulated kinase 1/2 and p38 mitogen-activated protein kinase signaling cascades. Mol Endocrinol. 2008;22:689–706. doi: 10.1210/me.2007-0136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mellado M, Rodriguez-Frade JM, Vila-Coro AJ, Fernandez S, Martin de Ana A, Jones DR, et al. Chemokine receptor homo- or heterodimerization activates distinct signaling pathways. EMBO J. 2001;20:2497–2507. doi: 10.1093/emboj/20.10.2497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neve KA, Seamans JK, Trantham-Davidson H. Dopamine receptor signaling. J Recept Signal Transduct Res. 2004;24:165–205. doi: 10.1081/rrs-200029981. [DOI] [PubMed] [Google Scholar]
- O'Dowd BF, Ji X, Alijaniaram M, Rajaram RD, Kong MM, Rashid A, et al. Dopamine receptor oligomerization visualized in living cells. J Biol Chem. 2005;280:37225–37235. doi: 10.1074/jbc.M504562200. [DOI] [PubMed] [Google Scholar]
- Orozco-Cabal L, Liu J, Pollandt S, Schmidt K, Shinnick-Gallagher P, Gallagher JP. Dopamine and corticotropin-releasing factor synergistically alter basolateral amygdala-to-medial prefrontal cortex synaptic transmission: functional switch after chronic cocaine administration. J Neurosci. 2008;28:529–542. doi: 10.1523/JNEUROSCI.2666-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SP, Buneman OP, et al. NC-IUPHAR. The IUPHAR/BPS Guide to PHARMACOLOGY: an expert-driven knowledgebase of drug targets and their ligands. Nucl. Acids Res. 42(Database Issue):D1098–1106. doi: 10.1093/nar/gkt1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perreault ML, Hasbi A, O'Dowd BF, George SR. Heteromeric dopamine receptor signaling complexes: emerging neurobiology and disease relevance. Neuropsychopharmacology. 2014;39:156–168. doi: 10.1038/npp.2013.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rashid AJ, So CH, Kong MM, Furtak T, El-Ghundi M, Cheng R, et al. D1-D2 dopamine receptor heterooligomers with unique pharmacology are coupled to rapid activation of Gq/11 in the striatum. Proc Natl Acad Sci U S A. 2007;104:654–659. doi: 10.1073/pnas.0604049104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rutz C, Renner A, Alken M, Schulz K, Beyermann M, Wiesner B, et al. The corticotropin-releasing factor receptor type 2a contains an N-terminal pseudo signal peptide. J Biol Chem. 2006;281:24910–24921. doi: 10.1074/jbc.M601554200. [DOI] [PubMed] [Google Scholar]
- Saal D, Dong Y, Bonci A, Malenka RC. Drugs of abuse and stress trigger a common synaptic adaptation in dopamine neurons. Neuron. 2003;37:577–582. doi: 10.1016/s0896-6273(03)00021-7. [DOI] [PubMed] [Google Scholar]
- Schulz K, Rutz C, Westendorf C, Ridelis I, Vogelbein S, Furkert J, et al. The pseudo signal peptide of the corticotropin-releasing factor receptor type 2a decreases receptor expression and prevents Gi-mediated inhibition of adenylyl cyclase activity. J Biol Chem. 2010;285:32878–32887. doi: 10.1074/jbc.M110.129627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shalev U, Erb S, Shaham Y. Role of CRF and other neuropeptides in stress-induced reinstatement of drug seeking. Brain Res. 2010;1314:15–28. doi: 10.1016/j.brainres.2009.07.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- So CH, Varghese G, Curley KJ, Kong MM, Alijaniaram M, Ji X, et al. D1 and D2 dopamine receptors form heterooligomers and cointernalize after selective activation of either receptor. Mol Pharmacol. 2005;68:568–578. doi: 10.1124/mol.105.012229. [DOI] [PubMed] [Google Scholar]
- Sun P, Wang J, Gu W, Cheng W, Jin GZ, Friedman E, et al. PSD-95 regulates D1 dopamine receptor resensitization, but not receptor-mediated Gs-protein activation. Cell Res. 2009;19:612–624. doi: 10.1038/cr.2009.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Teichmann A, Rutz C, Kreuchwig A, Krause G, Wiesner B, Schulein R. The Pseudo signal peptide of the corticotropin-releasing factor receptor type 2A prevents receptor oligomerization. J Biol Chem. 2012;287:27265–27274. doi: 10.1074/jbc.M112.360594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Terrillon S, Bouvier M. Roles of G-protein-coupled receptor dimerization. EMBO Rep. 2004;5:30–34. doi: 10.1038/sj.embor.7400052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verma V, Hasbi A, O'Dowd BF, George SR. Dopamine D1-D2 receptor heteromer-mediated calcium release is desensitized by D1 receptor occupancy with or without signal activation: dual functional regulation by G protein-coupled receptor kinase 2. J Biol Chem. 2010;285:35092–35103. doi: 10.1074/jbc.M109.088625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vickery RG, von Zastrow M. Distinct dynamin-dependent and -independent mechanisms target structurally homologous dopamine receptors to different endocytic membranes. J Cell Biol. 1999;144:31–43. doi: 10.1083/jcb.144.1.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vilardaga JP, Nikolaev VO, Lorenz K, Ferrandon S, Zhuang Z, Lohse MJ. Conformational cross-talk between alpha2A-adrenergic and mu-opioid receptors controls cell signaling. Nat Chem Biol. 2008;4:126–131. doi: 10.1038/nchembio.64. [DOI] [PubMed] [Google Scholar]
- Wang B, Shaham Y, Zitzman D, Azari S, Wise RA, You ZB. Cocaine experience establishes control of midbrain glutamate and dopamine by corticotropin-releasing factor: a role in stress-induced relapse to drug seeking. J Neurosci. 2005;25:5389–5396. doi: 10.1523/JNEUROSCI.0955-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang B, You ZB, Rice KC, Wise RA. Stress-induced relapse to cocaine seeking: roles for the CRF(2) receptor and CRF-binding protein in the ventral tegmental area of the rat. Psychopharmacology (Berl) 2007;193:283–294. doi: 10.1007/s00213-007-0782-3. [DOI] [PubMed] [Google Scholar]
- Waselus M, Nazzaro C, Valentino RJ, Van Bockstaele EJ. Stress-induced redistribution of corticotropin-releasing factor receptor subtypes in the dorsal raphe nucleus. Biol Psychiatry. 2009;66:76–83. doi: 10.1016/j.biopsych.2009.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zorrilla EP, Logrip ML, Koob GF. Corticotropin releasing factor: a key role in the neurobiology of addiction. Front Neuroendocrinol. 2014;35:234–244. doi: 10.1016/j.yfrne.2014.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]

