• Nie Znaleziono Wyników

2A 1A andserotonin5-HT receptorsrevealthespecificityofreceptorinteractionsintheplasmamembrane FluorescencestudiesofhomooligomerizationofadenosineA

N/A
N/A
Protected

Academic year: 2022

Share "2A 1A andserotonin5-HT receptorsrevealthespecificityofreceptorinteractionsintheplasmamembrane FluorescencestudiesofhomooligomerizationofadenosineA"

Copied!
14
0
0

Pełen tekst

(1)

Fluorescence studies of homooligomerization of adenosine A 2A and serotonin 5-HT 1A receptors reveal the specificity of receptor interactions in the plasma membrane

Sylwia £ukasiewicz1*, Ewa B³asiak1*, Agata Faron-Górecka2, Agnieszka Polit1, Magdalena Tworzyd³o1, Andrzej Górecki1, Zygmunt Wasylewski1, Marta Dziedzicka-Wasylewska1,2

Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, PL 30-387 Kraków, Poland

Institute of Pharmacology, Polish Academy of Sciences, Smêtna 12, PL 31-343 Kraków, Poland

*Both authors contributed equally to this work

Correspondence: Marta Dziedzicka-Wasylewska, e-mail: wasyl@mol.uj.edu.pl

Abstract:

The concept that G protein-coupled receptors (GPCRs) function as oligomers has been widely accepted, however, different methodologies often used to study the phenomenon of GPCR interactions do not allow, as yet, for any generalization as to whether di- or oligomers are formed constitutively or are ligand-promoted. Here, we report on the use of three independent biophysical ap- proaches based on the Förster resonance energy transfer to study the adenosine A2Aand serotonin 5-HT1Areceptor (tagged with de- rivatives of green fluorescence protein, CFP – fluorescence donor and YFP – fluorescence acceptor) homodimerization in the plasma membrane of transiently transfected HEK 293 cell line. Homodimers of A2Aand 5-HT1Areceptors are formed constitutively, how- ever, specific ligands regulate the degree of these interactions: agonists (CGS 21680 and 8-OH-DPAT, respectively) further en- hanced while antagonists (SCH 58216 and methysergide) diminished the dimer formation. Although the acceptor photobleaching with the use of confocal microscopy as well as the fluorescence lifetime microscopy gave similar results, we strongly recommend the latter technique as a highly sensitive and quantitative approach to that kind of the study. The additional proof of specificity of the ob- served results is provided by the studies of interaction of adenosine A2Aand serotonin 5-HT1Areceptors with thea subunits of G pro- teins. The A2Areceptor interacted with Gas and 5-HT1Areceptor – with Gai, while physical interaction of these receptors with no appropriatea subunits partners (A2A-Gai and 5HT1A-Gas) has not been observed, despite the identical level of overexpression of proteins in all studied combinations.

Key words:

adenosine A )receptor, serotonin 5-HT)receptor, oligomerization of GPCRs, fluorescence lifetime microscopy, confocal microscopy, FRET

Abbreviations: BRET – bioluminescence resonance energy transfer, CFP – cyan fluorescent protein, FRET – fluorescence resonance energy transfer, Gai3 – G protein alpha inhibitory 3 subunit, GasL – G protein alpha stimulatory long subunit,

GPCRs – G protein-coupled receptors, HEK 293 – human em- bryonic kidney 293 cell line, TCSPC – time correlated single photon counting, YFP – yellow fluorescent protein

(2)

Introduction

The concept that G-protein-coupled receptors (GPCRs) can function as dimers or oligomers of higher order has been widely accepted [4]. The methodology of co- immunoprecipitation of receptors differentially tagged with epitopes and expressed in various cell types in vitro has been frequently used in the studies of GPCR dimerization [26]. However, some criticism concern- ing this methodology has been often raised. For ex- ample, the well-appreciated artifacts may result from the fact that stringent solubilization conditions often prevent the detection of constitutive dimers by pro- moting their partial or total dissociation. On the other hand, the hydrophobicity of the transmembrane do- mains of GPCRs might be responsible for non- specific aggregation following detergent extraction of proteins from cell membranes. Nevertheless, using the different techniques, it has been shown that many GPCRs can exist as dimers or higher order oligomers in living cells [12]. However, it has been shown for example that serotonin 5-HT1Areceptor was capable of forming homo- as well as heterooligomers with all GPCRs tested, including 5-HT1B, 5-HT1D, GABAB2 and EDG1, EDG3, GPR26 receptors, and even the authors of these studies have considered these find- ings as surprising [33]. Another member of the GPCRs family A, the adenosine A2A receptor can form both homodimers as well as heterooligomers with dopamine D2receptors [5, 6].

The question whether GPCR dimers or oligomers are constitutive or ligand-promoted has not been une- quivocally answered as yet [25]. The findings cannot currently be generalized, as different methodological approaches might lead to the erroneous conclusions.

Again, a matter of appropriate methodology em- ployed in that kind of studies might be of crucial im- portance. For example, while using coimmunopre- cipitation to study the influence of specific ligands on the GPCRs dimerization phenomenon, the conforma- tional changes resulting from ligand binding might change the stabilization of pre-existing dimers [46], which become partially resistant to the solubilization conditions, what may lead to the conclusion that ligand binding induces dimerization which is not the case.

Bearing in mind these uncertainties in coimmuno- precipitation methodology, we have used biophysical

approach to study the adenosine A2Aas well as sero- tonin 5-HT1A receptor homodimerization in the plasma membrane.

Both receptors belong to the GPCR family, play important role in neurotransmission and are the tar- gets of various pharmacological compounds used both in the basic research as well as in the clinical practice [11, 13, 20, 21, 27]. In most cases, serotonin 5-HT1Areceptor is coupled with Gi protein and nega- tively regulates the adenylate cyclase [31], whereas adenosine A2A receptor stimulates that enzyme mainlyvia the interaction with Gs protein [22, 42].

The development of fluorescent protein tags has made Förster resonance energy transfer very powerful and more frequently used tool for detecting changes in the distance between the fluorophores, therefore, between the proteins of interest which the probes la- bel. The use of cyan fluorescent protein (CFP) and yellow fluorescent protein (YFP) as a donor-acceptor pair seems the most adequate, since their emission (CFP) and the absorption (YFP) spectra overlap suffi- ciently [30]. Fluorescence lifetime microscopy, which spatially maps the donor lifetime, can overcome the problems of intensity-based methods (i.e. the acceptor photobleaching and steady-state spectrofluorimetry) because the lifetime is independent of the number of fluorescing molecules, so changes in lifetime reflect mainly changes in the distance between probes. In this study, we applied three different methods based on the fluorescence resonance energy transfer (FRET) phenomenon to investigate the receptors oligomeriza- tion. Regardless of the method used, our results show that A2Aand 5-HT1Areceptors can indeed form ho- modimers, and ligand binding regulates the degree of that interaction.

Materials and Methods

Materials

All molecular biology reagents were from Fermentas (Vilnius, Lithuania). Oligonucleotides were synthesiz- ed by IBB PAN (Warszawa, Poland). The pECFP-N1 and pEYFP-N1 vectors were from BD Biosciences, Clontech (Palo Alto, CA, USA). The pcDNA3.1(+) plasmids encoding human adenosine A2A receptor, human serotonin 5-HT1Areceptor, stimulatory GasL

(3)

and inhibitory Gai3 protein were from UMRcDNA Resource Center (University of Missouri-Rolla, MO, USA). Bacterial cell line Escherichia coli DH5a (Dam+) was from Novagen.

Human embryonic kidney 293 (HEK 293) cells were obtained from American Type Culture Collec- tion (Manassas, VA, USA). All materials for cell cul- ture were supplied by GIBCO and Sigma. Adenosine A2A receptor ligands: CGS 21680 (4-[2-[[6-amino- 9-(N-ethyl-b-D-ribofuranuronamidosyl)-9H-purin-2- -yl]amino]ethyl]benzenepropanoic acid hydrochlori- de), SCH 58261 (2-(2-furanyl)-7-(2-phenylethyl)-7H- -pyrazolo[4,3-e]1,2,4-triazolo[1,5-c]pyrimidin-5-ami- ne) were supplied by Tocris (Bristol, United King- dom) and caffeine (1,3,7-trimethylxanthine) was from Sigma. Serotonin 5-HT1A receptor agonist, 8-OH- DPAT ((±)-8-hydroxy-2-dipropylaminotetralin hydro- bromide) and antagonist, methysergide maleate ([8b(S)]- 9,10-didehydro-N-[1-(hydroxymethyl)propyl]-1,6-di- methylergoline-8-carboxamide maleate) were from Research Biochemicals International (Natick, MA, USA).

Construction of fusion proteins

The human adenosine A2Aand serotonin 5-HT1Are- ceptors as well as stimulatory and inhibitory G protein subunit genes cloned in pcDNA3.1(+) plasmid were used as starting point for the construction of fusion proteins. Molecules were tagged with cDNA coding enhanced cyan or yellow fluorescent proteins (ECFP or EYFP) and used after expression as fluorescence donor or acceptor, respectively. Henceforth the cyan (ECFP) and yellow (EYFP) variants are called CFP and YFP.

The full length cDNAs encoding 4 above-men- tioned proteins were PCR-amplified. Forward primer was universal for pcDNA3.1(+), reverse primers re- moved the STOP codons and introduced unique re- striction sites:EcoRI for A2A,XmaI for 5-HT1A,SacI for both kind of Ga subunits. Resulting fragments were inserted in-frame into NheI/EcoRI (A2A), NheI/XmaI (5-HT1A) or NheI/SacI (Ga) sites of the pECFP-N1 and pEYFP-N1 vectors.

The CFP-YFP fusion protein construct was ob- tained after PCR amplification of ECFP cDNA coding fragment on the pECFP-N1 matrix. The forward primer introduced in NheI restriction site and the re- verse primer excluded the STOP codon and intro- duced anEcoRI restriction site. The resulting product

was cloned into pEYFP-N1 plasmid usingEcoRI and NheI restriction endonucleases.

Cell culture and transfection

HEK 293 cells were grown in Dulbecco’s modified essential medium supplemented with 1% L-glutamine and 10% heat-inactivated fetal bovine serum (FBS), at 37°C, in the atmosphere of 5% CO2. Transient transfections of HEK 293 cells were performed by calcium phosphate precipitation method, as described by Sambrook et al. [34]. Cells were transfected with plasmids encoding either receptor (A2Aor 5-HT1A)- CFP or receptor (A2Aor 5-HT1A)-YFP fusion protein alone or cotransfected with both plasmids. The sepa- rate experiments included transfection with pECFP- EYFP vector and cotransfection with one of the recep- tors and the G-protein subunit fusion proteins. One day before transfection, cells were seeded into 100 mm dishes (3 × 106 cells/dish) – for fluorescent spectra measurements, and on glass coverslips in 30 mm dishes (1 × 106/dish) – for fluorescent lifetime meas- urements and confocal imaging. They were trans- fected with 12mg of DNA/100 mm dish and 2 mg of DNA/30 mm dish. The ratio of DNA coding donor and acceptor was 1:1.

Membrane preparation and radioligand binding assay

For binding experiments, HEK 293 cells transfected with plasmid coding for A2Aor 5HT1Areceptor were washed with phosphate-buffered saline (PBS), scraped from the dish, and centrifuged at 1000 rpm for 5 min.

The pellet was frozen at –30°C until it was used.

In A2Abinding experiments, frozen pellets were re- suspended in the binding buffer [50 mM Tris-HCl (pH 7.4)] containing 10 mM MgCl2and 2U/ml adenosine deaminase Calbiochem, Darmstadt, Germany), using an Ultra Turrax homogenizer. The homogenates were centrifuged at 30,000 × g for 10 min. That step was repeated twice. [3H]CGS21680 (specific activity of 40.5 Ci/mmol, PerkinElmer) was used as the adeno- sine A2A receptor-specific radioligand. Binding as- says were performed in a total volume of 700 ml.

Saturation studies were carried out on a fresh mem- brane preparation (final protein concentration of 200 mg/tube) using [3H]CGS21680 concentrations ranging from 0.05 to 17 nM (nonspecific binding was assessed by addition of 20 mM 2-chloroadenosine

(4)

(2-CADO, Sigma). Tubes were incubated for 120 min at 25°C and then binding was terminated with rapid filtration through glass fiber filters (GF/B, Whatman).

The filters were washed four times with 5 ml of ice- cold washing buffer [50 mM Tris-HCl (pH 7.4)] and bound radioactivity was determined by liquid scintil- lation counting (Beckman LS 650).

Binding experiments for 5HT1Areceptor were per- formed in analogous way: frozen pellets were resus- pended in the binding buffer [50 mM Tris-HCl (pH 7.4)], homogenized twice and resuspended in binding buffer containing 10mM pargyline and 5.7 mM CaCl2. [3H]8-OH-DPAT (specific activity of 106 Ci/mmol, PerkinElmer) was used as the serotonin 5HT1A receptor-specific radioligand. Assays were performed in a total volume of 600 ml. Saturation studies were carried out on a fresh membrane preparation (final protein concentration of 100 mg/tube) using [3H]8- OH-DPAT concentrations ranging from 0.12 to 3 nM (nonspecific binding was assessed by addition of 50mM serotonin HCl, RBI). After 15-min incubation at 37°C binding was terminated by rapid filtration through glass fiber filters then filters were washed and bound radioactivity was determined by liquid scintil- lation counting.

Fluorescence spectroscopy measurements

Spectrofluorimetric measurements of cell suspensions were recorded on Fluorolog 3 (Horiba, Jobin Yvon S.A.S., Longjumeau, France) at 37°C, 48 h after transfection. Cell culture from a single 100 mm dish was washed and detached from a plate using PBS. Af- terwards, the suspension was centrifuged at 1000 rpm and resuspended in 1 ml of isotonic buffer (137.5 mM NaCl, 1.25 mM MgCl2, 1.25 mM CaCl2, 6 mM KCl, 5.6 mM glucose, 10 mM HEPES, 0.4 mM NaH2PO4, pH 7.4). CFP was excited at 434 nm, YFP at 475 nm, fluorescence was detected at 450–550 nm through a double monochromator. The excitation and emission slits were 2 and 10 nm, respectively. All fluorescence spectra were measured in a 10 mm quartz cuvette (Hellma, Müllheim Germany). The spectral contribu- tions arising from light scattering and non-specific fluorescence of cells and buffer were eliminated by substracting the emission spectra of mocked- transfected cells from the fluorescence spectra of cells expressing the receptors-CFP and -YFP constructs [38].

Measurements of FRET by fluorescence life- time microscopy

Time-correlated single photon counting (TCSPC) measurements were performed on the inverted fluo- rescence microscope, Olympus platform IX 71. Spe- cimens were excited with the diode pulse laser (Ho- riba, Jobin Yvon IBH S.A.S., Longjumeau, France) at 434 nm with 1 MHz repetition. The fluorescence emission was recorded by a picosecond detector TBX-04 (Horiba, Jobin Yvon IBH S.A.S., Longju- meau, France). The Jobin Yvon IBH data station and the DAS 6 software were used for the data acquisition and the decay analysis.

Cells appropriated for TCSPC experiments were grown on the cover slips. The fluorescence decay from single cells transfected with fusion protein con- structs were measured using 60× objective and di- chroic beam splitter at 455 nm combined with emitter cut off filter > 475 nm. The excitations pulse diode la- ser profile, required for the deconvolution analysis, was measured on the diluted glycogen using fluorine cube with 400 nm dichroic beam splitter only. All measurements were performed at 37°C. Cells were in- cubated in the same isotonic buffer as for fluores- cence spectra measurements. In order to investigate the influence of specific ligands, the cells were incu- bated in the presence of agonists or antagonists at 37°C for 15 min before the experiment. During each experiment at least 10 cells on the cover slip were measured.

Each fluorescence decay was analyzed with the multiexponential model given by the equation:

I(t) ie-t/ i

i n

=

=1

α τ

(1)

where I(t) stands for fluorescence intensity in time t, aiare pre-exponential factors representing amplitudes of the components att = 0, tiare the decay times and n is the number of decay components. Best fit pa- rameters were obtained by minimization of reduced c2 value and residual distribution. The average fluo- rescence lifetimeátñ was calculated from the equation:

τ

α τ

= α τ

i i i i i i

2

(2)

The average efficiency of energy transferáEñ was calculated from the average fluorescence lifetime of

(5)

donor in the absence átDñ or in the presence átDAñ of acceptor from the equation:

E DA

D

= −1 τ

τ (3)

Confocal microscopy analysis

HEK 293 cells grown on cover slips were transiently transfected with the cDNA encoding the indicated pro- teins and were incubated with 0.2 mM CGS 21680 (A2Aagonist) and 0.1mM caffeine (A2Aantagonist), 100 mM methysergide (5-HT1A antagonist), 1 mM 8-OH-DPAT (5-HT1A agonist) for 15 min. Analysis was performed on confocal laser scanning microscope Leica TCS SP2 (Leica Microsystem, Mannheim, Ger- many) with acousto-optical beamsplitter and 100 mW argon laser. At first, a pre-photobleach image of fluo- rescence acceptor (YFP) was made by scanning while exciting with the 514 nm laser line. An image of pre- photobleach of fluorescence donor was acquired by scanning while exciting with 458 nm laser line. After- wards one region of the cell was selected, and accep- tor was again photobleached, until the signal of fluo- rescence emission was at the level of 50% of the ini- tial value. A post-photobleach images of donor and acceptor were made as described above. The FRET efficiency was calculated from the equation (4):

FRET D D

Eff postD pre post

= −

(4)

The statistical significance was assessed using Stu- dent’st-test.

The representative set of images accumulated dur- ing single photobleaching measurement is presented in Figure 1.

Results

Analysis of localization of adenosine A

2Aand serotonin 5-HT1Areceptor fusion proteins

Confocal microscopy observations indicate that fu- sion proteins of both adenosine A2A and serotonin 5-HT1A receptors were localized in plasma mem- brane. Figures 1 and 2 show HEK 293 cells tran- siently cotransfected with fusion protein constructs

Fig. 1. Colocalization of fluorescently tagged serotonin 5-HT)re- ceptors in the plasma membrane of transiently transfected HEK 293 cells. The proteins were subjected to FRET measurements with the use of acceptor photobleaching, as described in Experimental pro- cedures. The analyzed part of cell is delineated by green rectangle.

(A) Donor fluorescence intensity (5-HT)-CFP) before acceptor (5-HT)-YFP) photobleaching. (B) Acceptor fluorescence intensity (5-HT)-YFP) before acceptor photobleaching. (C) Increase in donor fluorescence intensity (5-HT)-CFP) after acceptor photobleaching.

(D) Decrease in acceptor fluorescence intensity after acceptor pho- tobleaching

Fig. 2. Localization of fluorescently tagged adenosine A )receptors in the plasma membrane of transiently transfected HEK 293 cells.

(A) Adenosine receptor tagged with YFP – fluorescence acceptor;

(B) Adenosine receptor tagged with CFP – fluorescence donor

(6)

encoding 5-HT1A and adenosine A2A receptors, re- spectively. Sharpness of cell’s edges confirms the sug- gestion that adenosine A2Aand serotonin 5-HT1Are- ceptor fusion proteins were localized in plasma mem- brane, in contrast to Ga proteins, that are additionally localized in cytosol (data not shown).

Fluorescence spectroscopy measurements of receptor dimerization

Fluorescence emission profiles for HEK 293 cells ex- pressing fusion proteins in different combinations (expression of one fusion protein: A2A-CFP, 5-HT1A- CFP, A2A-YFP, 5-HT1A-YFP or CFP-YFP; or coex- pression of two fusion proteins with CFP as a donor and YFP as an acceptor of energy: A2A-CFP with A2A-YFP, 5-HT1A-CFP with 5-HT1A-YFP and A2A-CFP with 5-HT1A-YFP) were compared using an excitation wavelength of 434 nm (donor absorption).

The fluorescence emission spectra differed between experiments. Figure 3A shows representative spectra obtained after transient transfection of HEK 293 cells with the construct of two fluorescent proteins CFP and YFP linked together by a short 15 amino acid chain. Significant energy transfer was observed when the cells were excited at donor absorption wavelength (434 nm). Small difference in YFP fluorescence emis- sion at 527 nm after excitation at 434 and 475 nm sug- gests that all conditions necessary for energy transfer were fulfilled and FRET was nearly maximal. The ef- ficiency of FRET for CFP-YFP mutant protein should be considered as the highest in comparison to other derivatives of fluorescent proteins such as receptor- CFP and receptor-YFP fusion protein. Although the spectroscopic measurements enable only quantitative estimation of FRET phenomenon, it is demonstrative, since one can compare the alterations in the ratio be- tween donor and acceptor emission peaks, what indi- cates whether the energy transfer takes place or not.

We used spectrofluorimetry to investigate the inter- actions between adenosine A2Aand serotonin 5-HT1A receptors. A representative spectrum from the experi- ment is presented in Figure 3B. It shows the emission spectrum of the cell suspension cotransfected with A2A-CFP and 5-HT1A-YFP fusion proteins after exci- tation at 434 nm. The obtained results indicate that these receptors (A2A and 5-HT1A) can form hetero- oligomers in the cell membrane.

Figure 3C shows spectra obtained after transfection of two HEK 293 cell populations with adenosine re-

ceptor hybrid mutants (A2A-CFP and A2A-YFP) sepa- rately, followed by mixing them before the measure- ment. There was no visible energy transfer after exci- tation at 434 nm, although both fluorophores were present in the sample.

Representative spectra of cells cotransfected with both A2Afusion proteins (A2A-CFP and A2A-YFP) is shown in Figure 3D. Relatively high fluorescence in- tensity at 525 nm in comparison with spectra shown in Figure 3B suggests that adenosine A2Areceptors were in the close vicinity, allowing the formation of homooligomeric structures.

Figure 3E shows typical spectra when two cell populations, one expressing 5-HT1A-CFP fusion pro- tein and the second one expressing 5-HT1A-YFP fu- sion protein, were mixed. There was no visible energy transfer, despite the presence of both fluorophores in the sample. The shape of the recorded spectra was the same as in case of HEK 293 cells transfected sepa- rately with the plasmids containing receptor-CFP or receptor-YFP fusion protein.

On the other hand, the FRET phenomenon was ob- served in HEK 293 cells cotransfected with the plas- mids encoding both serotonin receptor fusion proteins (5-HT1A-CFP and 5-HT1A-YFP). Spectra presented in Figure 3F confirm the homodimerization of the sero- tonin 5-HT1Areceptors.

Confocal microscopy studies of adenosine A2A and serotonin 5-HT1Areceptor interaction

Confocal microscopy experiments showed that adenosine A2Aand serotonin 5-HT1Areceptors exist as homooligomeric complexes in the cell membrane.

The observed FRET efficiency was influenced by spe- cific ligands (Fig. 4). 0.2mM CGS21680 – adenosine A2A agonist and 1 mM 8-OH-DPAT – serotonin 5-HT1Aagonist increased FRET efficiency, however, only results for 8-OH-DPAT are statistically signifi- cant. In case of receptor antagonists: 0.1mM caffeine and 100 mM methysergide the efficiency of FRET was lowered, and these effects were statistically sig- nificant.

Fluorescence lifetime microscopy studies of re- ceptor interaction

The time correlated single photon counting experi- ments were performed on the inverted fluorescence microscope. FRET phenomenon was observed in

(7)

Fig. 3. Representative fluorescence emission spectra of HEK 293 cells transfected with CFP and YFP derivatives. (A) Positive FRET control, spectra from cells transfected with the CFP-YFP hybrid mutant. Black – excitation at 434 nm, grey – excitation at 475 nm. (B) Representative fluorescence emission spectra of HEK 293 cells cotransfected with adenosine A )-CFP and serotonin 5-HT)-YFP receptor fusion proteins, excitation at 434 nm. (C) Negative FRET control, spectra from 1:1 mixture of cells individually expressing the A )-CFP fusion protein (black) excited at 434 nm and A )-YFP fusion protein (grey) excited at 475 nm. (D) Spectra of cells coexpressing the A )-CFP and A )-YFP fusion proteins excited at 434 nm in the absence of ligand (black line). (E) Negative FRET control, spectra from 1:1 mixture of cells individually expressing the 5-HT)-CFP fusion protein (black) excited at 434 nm and 5-HT)-YFP fusion protein (grey) excited at 475 nm. (F) Spectra from cells coexpressing the 5-HT)-CFP and 5-HT)-YFP fusion proteins excited at 434 nm in the absence of ligand (black line)

s

(8)

a single living cell. This kind of measurements is in- dependent of any change in fluorophore concentration or excitation intensity and allows to obtain the highly quantifiable data. For the detection of FRET effi- ciency, the precise measurement of the donor fluores- cence lifetime (CFP), in the presence and absence of the acceptor (YFP), is required. Fluorescence decays were analyzed as both mono- or multiexponentials.

Analysis of reducedc2value and residual distribution has led to the conclusion that best fit parameters were obtained with two exponentials. Adding the third one did not significantly influence the parameters and the fractional contribution of the additional lifetime was close to zero.

Average CFP fluorescence lifetime designated dur- ing TCSPC experiments was 2.37 ns. The average do- nor lifetime decreased in the presence of acceptor.

Fig. 4. Confocal microscopy. FRET efficiency of receptor-CFP and receptor-YFP pairs in the absence and presence of appropriate ago- nists and antagonists measured by acceptor photobleaching. Data are the mean ± SEM of four independent experiments performed in duplicate. Statistical significance was evaluated using Student’s t-test; (A) A )-CFP and A )-YFP; * p < 0.05 caffeine 0.1 µM vs. A ) A ). (B) 5-HT)-CFP and 5-HT)-YFP; * p < 0.05 8-OH-DPAT 1 µM and methysergide 100 µM vs. 5-HT)– 5-HT)

Fig. 5. Fluorescence lifetime microscopy measurements. (A) FRET positive control: HEK 293 cells expressing the donor molecule – CFP (D) and the donor connected through a short amino acid chain with acceptor molecule – CFP-YFP (DA). Dotted curves show the intensity decays of the donor alone (black) or the donor linked with the accep- tor (dark grey). Black solid lines are double exponential fits. The exci- tation pulse diode laser profile, set up at 434 nm, is shown by grey dotted curve. In the lower panels, the solid lines present weighted re- siduals for two exponential fits. (B) Representative fluorescence life- time measurements of HEK 293 cells expressing the fusion receptor 5-HT)-CFP (D) and coexpressing the donor and acceptor mole- cules fused with receptor (DA). Black dotted curves show the inten- sity decays of donor alone. Dark grey dotted curves show the inten- sity decays of donor in the presence of acceptor. Black solid lines are double exponential fits. The excitation pulse diode laser profile is shown by grey dotted curve. Solid lines on the lower panels present weighted residuals for two exponential fits

(9)

The highest FRET phenomenon observed in cells transfected with CFP-YFP hybrid was characterized by average donor lifetime of 1.52 ns that gives FRET efficiency of about 36% (Fig. 5A). When we com- pared the fluorescence decays measured for CFP bound to serotonin 5-HT1Areceptor in the presence or ab- sence 5-HT1A-YFP, the shortening of donor fluores- cence lifetime was observed, what indicates the con- stitutive 5-HT1Ahomooligomerization (Fig. 5B). The value of energy transfer efficiency was estimated as 7%. This value changed after incubation of cells with specific 5-HT1Areceptor ligands: 1mM 8-OH-DPAT (agonist), and 100 mM methysergide (antagonist).

Methysergide significantly decreased the FRET effi- ciency (2.5%) whilst 8-OH-DPAT caused slight but not statistically significant increase in energy transfer (9%).

Measurements of the cells cotransfected with two adenosine A2Areceptor fusion proteins indicate more than 10% energy transfer efficiency with average fluorescence lifetime 2.05 ns. This value changed af- ter addition of selective ligands (Tab. 1). The agonist, 0.2mM CGS 21680 caused further increase in transfer

efficiency to the level of 12.8%, however, the effect was not statistically significant. A selective antago- nist, 1mM SCH 58261 caused a significant decrease in FRET to the value of 6.8%.

We also observed heterooligomerization between adenosine A2A and serotonin 5HT1A receptors. The cells cotransfected with A2A-CFP and 5HT1A-YFP show 8.3% efficiency of energy transfer.

Fluorescence spectroscopy measurements of receptor and Ga subunits interactions

Separate experiments demonstrating the selectivity of protein interactions in fluorescence spectroscopy measurements were carried out with GasL or Gai3 fusion proteins used as fluorescence donors of energy and adenosine A2A or serotonin 5-HT1A receptors used as fluorescence acceptors. Figure 6 presents the spectra of HEK 293 cells cotransfected with one of the receptors (either A2A-YFP or 5-HT1A-YFP) and G protein subunits connected to CFP. In case of adeno- sine A2Areceptor (Fig. 6A), there was a visible en- ergy transfer when GasL was expressed in cells. This interaction seems to be specific since there was no

Tab. 1. Summary of energy transfer measurements by fluorescence lifetime microscopy in HEK 293 cells

Species Average lifetime [ns] Transfer efficiency

áEñ (%)

á tDñ átDAñ

=CFP 2.36 (± 0.02)

>CFP-YFP 1.52 (± 0.03) 35.8

?A )-CFP 2.30 (± 0.02)

@A )-CFP + A )-YFP 2.05 (± 0.02) 10.6

AA )-CFP + A )-YFP + CGS 21680 2.00 (± 0.03) 12.8

AA )-CFP + A )-YFP + SCH 58261 2.14 (± 0.02)** 6.8

B5-HT)-CFP 2.30 (± 0.02)

C5-HT)-CFP + 5-HT)-YFP 2.14 (± 0.02) 7.0

D5-HT)-CFP + 5-HT)-YFP + 8-OH-DPAT 2.10 (± 0.03) 9.0

D5-HT)-CFP + 5-HT)-YFP + methysergide 2.24 (± 0.03)** 2.5

EA )-CFP+ 5-HT)-YFP 2.11 (± 0.02) 8.3

Excitation was set up at 434 nm and emission was observed through appropriate interference filters, as described in experimental procedures.

The standard errors of means are presented in parentheses n = 15. Statistical significance was evaluated using Student’s t-test; **p < 0.02 SCH 58261 vs. A )– A ), ** p < 0.02 methysergide vs. 5-HT)– 5-HT).=measured in cells expressing CFP protein,>measured in cells expressing CFP coupled to YFP through a short amino acid chain,?measured in cells expressing adenosine A )receptor coupled to CFP,

@measured in cells coexpressing A )-CFP and A )-YFP,Ameasured in cells coexpressing A )-CFP and A )-YFP in the presence of agonist:

0.2 µM CGS 21680 and antagonist 1 µM SCH 58261,Bmeasured in cells expressing 5-HT)receptor coupled to CFP,Cmeasured in cells coex- pressing 5-HT)-CFP and 5-HT)-YFP,Dmeasured in cells coexpressing 5-HT)-CFP and 5-HT)-YFP in the presence of 1 µM agonist 8-OH-DPAT or 100 µM antagonist methysergide,Emeasured in cells coexpressing A )-CFP and 5-HT)-YFP

(10)

FRET after cotransfection of adenosine A2Areceptor withai3 subunit of G protein.

The opposite effect was observed with serotonin 5-HT1Areceptor anda subunits of G protein, which,

however, also demonstrates the specificity of protein interactions. There was a visible energy transfer be- tween fluorophores, when Gai3 subunit was present.

GasL subunit did not interact specifically with 5-HT1Areceptor. Results are shown in Figure 6B.

Figure 6C serves as negative control. It presents the spectra of two different populations of cells, mixed together, among which one expressed A2A-YFP and the second – GasL-CFP fusion protein. There was no FRET phenomenon between proteins which interact with each other only if expressed together in one cell.

Discussion

The aim of the present study was to introduce three independent approaches using the Förster resonance energy transfer (FRET) technology for studies of physical interactions between GPCRs, often referred to as GPCR oligomerization.

In most of intact cell population, studies using both bioluminescence resonance energy transfer (BRET) and FRET, in which fluorescent proteins are tagged to the C-terminal tail of GPCRs, it is not possible to de- fine the cellular location from which the signal de- rives, since the large fraction of the GPCRs expressed in a heterologous systems can be located intracellu- larly. The present experiments show that serotonin 5HT1Aand adenosine A2Areceptors tagged with fluo- rescent proteins CFP and YFP, expressed in the HEK 293 cells, are located in the plasma membrane, as can be judged by laser confocal microscopy, in contrast to a subunits of the G proteins which are not located ex- clusively in the sub-membrane area. The application of FRET microscopy, particularly when combined with photobleaching protocols [28, 29] has provided the means to monitor oligomerization of GPCRs in the plasma membrane of a single cell.

GPCRs oligomerization is difficult to analyze in native cells, therefore, human embryonic kidney cell line has been widely used in resonance energy trans- fer studies of membrane receptors, since these cells provide an accepted model, in which fluorescently tagged receptor proteins can be efficiently expressed.

As has been reported by Mercier et al. [24], the extent of dimerization ofb2-adrenergic receptors (shown by BRET) was unchanged over 20-fold range of expres- sion levels (from 1.4 to 26.3 pmol/mg protein). While

Fig. 6. Representative fluorescence emission spectra of HEK 293 cells cotransfected with A )-YFP and 5-HT)-YFP with Ga-CFP fusion proteins. (A) Spectra of cells coexpressing A )-YFP and GasL-CFP (solid line) or Gai3-CFP (dotted line) fusion proteins, excited at 434 nm; (B) Spectra of cells coexpressing 5-HT)-YFP and Gai3-CYFP (solid line) or GasL-CFP (dotted line) fusion proteins, excited at 434 nm; (C) Negative FRET control, spectra from 1:1 mixture of cells indi- vidually expressing A )-YFP (black) fusion proteins excited at 434 nm and GasL-CFP (grey) fusion protein excited at 475 nm

(11)

studying the homodimerization of neuropeptide Y re- ceptors, Dinger et al. [9] have also demonstrated that FRET effect was independent of the level of receptor expression. Similarly, BRET signal was stable for ex- pression levels ranging from 0.58 to 6.5 pmol/mg pro- tein for b2/b3-adrenergic receptor heterooligomers [3]. Our present studies were performed with the rea- sonable A2Aand 5HT1Areceptor levels, ranging from ca. 1.6 to 2.0 pmol/mg protein. These findings imply that examples of GPCR dimerization are not merely artifacts derived from the high levels of expression that are often achieved in heterologous cell system.

Qualitative fluorescence spectroscopy in cell sus- pension, performed by stationary fluorescence emis- sion spectra measurements, as well as quantitative FRET analysis, performed by confocal microscopy of cells grown on cover slips, indicate that both recep- tors which are the subject of the present study can form homodimers. Similar results were obtained with the use of fluorescence lifetime microscopy. The fluo- rescence lifetime refers to the time the fluorophore stays in its excited state before emitting a photon (a few ns). It is strongly affected by microenviron- mental factors and any energy transfer between an ex- cited molecule and its environment changes the fluo- rescence lifetime in a predictable way [17], independ- ent of chromophore concentration or excitation intensity. Therefore, fluorescence lifetime microscopy is a direct approach quantifying effects that involve energy transfer.

From the results presented here (obtained with all three methods used in the present study), it may be concluded that both receptors can form homodimers constitutively (also the formation of constitutive hete- rooligomers of A2A–5-HT1Areceptors has been ob- served), since the relatively high FRET efficiency has been observed even without any specific ligand. How- ever, upon the presence of a 5-HT1Areceptor agonist, 8-OH-DPAT, the transfer efficiency slightly in- creased, indicating the enhanced physical interaction of the receptor molecules, while the presence of methysergide, an antagonist, decreased the FRET effi- ciency. Similar effects were observed using independ- ent approaches, i.e. FRET efficiency values deter- mined by lifetime microscopy and by confocal mi- croscopy, both in the presence and absence of specific ligands, were in agreement. We believe that these complementary observation validate the acceptor photobleaching methodology used in the present study, despite the often raised criticism concerning

this approach [2], and further justify the use of each approach separately.

Adenosine A2A receptor homodimerization was also constitutive, with the transfer efficiency of ca.

10% (fluorescence lifetime microscopy result), how- ever, it was further influenced by specific ligands in a manner similar to that observed with 5-HT1Arecep- tor: an agonist, CGS 21680, significantly increased the adenosine A2Areceptor homodimerization, while an antagonist, SCH 58261, decreased it, although not completely, since the transfer efficiency of ca. 6.82%

was still observed upon the presence of this com- pound. Another A2Areceptor antagonist, caffeine, has also been shown to decrease the degree of receptor homodimerization.

In the previously published study, activation of the A2Areceptor by an agonist did not influence the de- gree of the receptor homodimerization [5], although both monomers and dimers were detected by im- munoblotting in cell extracts. On the other hand, cell surface biotinylation of proteins has shown that al- most all of the cell surface receptors were in the di- meric form. These authors have postulated that the homodimers are the most probably constitutively formed [5], similarly as has been shown in other stud- ies [14, 32, 40]. Additionally, it has been postulated that adenosine A2Areceptor agonists may lead to the reorganization of receptors within the plasma mem- brane while not affecting their degree of homodimeri- zation. Similar conclusion has been drawn by Ayoub et al. [1] from the studies of melatonin MT1 and MT2 receptors, and their ligands. It has been suggested that no correlation could be inferred between the receptor activation state and the dimerization state of the re- ceptor.

However, our results indicate that the ligands do in- fluence the degree of homodimerization, and the apparent discrepancy might result from different methodology used in both studies. Such discrepancies of the results concerning various receptors have been observed by others too, especially when the different experimental approach has been used. For example, Cvejic and Devi, using biochemical cross-linking, have reported that delta opioid receptor dimerization is reduced by agonist ligands in a concentration- and efficacy-dependent manner [7], whereas, using both BRET and time-resolved FRET approaches, McVey et al. [23] found that agonist and antagonist ligands had no effect on delta opioid receptor oligomeriza- tion. Another example is the chemokine receptor

(12)

CCR5, which was found to form an agonist-depen- dent dimer using biochemical cross-linking method [45] while the same receptor was shown by BRET to form a constitutive dimer and no change in the inten- sity of BRET was observed in the presence of an ago- nist [14].

On the other hand, the significant influence of spe- cific ligands on the receptor oligomerization has been frequently observed. For example, in the studies by Patel et al. [27] in CHO-K1 cells stably expressing somatostatin receptors, it has been shown that the re- ceptors are monomeric in their basal state and oligo- merize upon agonist activation. Also Wurch et al. [48]

have shown that there is an agonist-mediated oligo- merization of a splice variant of dopamine D2recep- tor (D2S) which has been postulated to exist presynap- tically and act as an autoreceptor. Additionally, in our recently published studies, we were able to show that dopamine D1 and D2 receptors oligomerize in the plasma membrane and that that phenomenon was sig- nificantly increased by the concomitant stimulation of both receptors by their specific agonists [10]. These results remain in line with the data provided by Lee et al. [18], who have shown that agonist stimulation of coexpressed D1and D2receptors resulted in stimula- tion of different signaling pathway, not activated by either receptor alone.

Indeed, the most fascinating feature of GPCR oligomerization, somewhat intuitively anticipated, is a distinct pharmacology and/or function of such oli- gomers as compared to the individual GPCRs ex- pressed alone which the most probably exist as homo- dimers. This has been already shown for opioid recep- tors [8, 19] but also for dopamine D2/D3 receptor heterooligomers [35]. One has also to take into ac- count that interactions between receptors and G pro- teins contribute to the apparent cooperativity, how- ever, as has been shown by Wreggett and Wells, ago- nists can bind with multiple affinities to preparations of muscarinic M2 receptors even in the absence of G proteins [47]. Similar view may well apply for recep- tor homodimers. Monomer-dimer selectivity has been suggested to explain discrepancies between the densi- ties of raclopride and spiperone binding to dopamine D2receptors [36, 37, 39],viz. raclopride has been sup- posed to recognize both binding sites within the ho- modimer, whereas spiperone – only a single binding unit.

The interpretation of FRET data is a complex task because energy transfer depends on several factors in-

cluding the distance between donor and acceptor, ex- pression level, protein affinity and stoichiometry.

FRET efficiency depends on the inverse of sixth power of the mean separation of the donor and accep- tor. This very high proximity-dependence means that for donors and acceptors forming constitutive oligo- meric structures FRET efficiency will, therefore, usu- ally be high. A potential complication of all FRET ex- periments is that background signals may arise from random interactions if donor and acceptor level is suf- ficiently high. FRET efficiency in this case will gen- erally be lower than that for oligomers as a result of the large average separation of donors and acceptors.

The problem often discussed while using biophysi- cal techniques to study receptor oligomerization con- cerns the fact that they predominantly involve heter- ologous expression systems, in most cases they have been performed on cell lines transfected with the re- ceptors of interest. Receptors are usually epitope tagged and in most cases are overexpressed. There- fore, it has been often suggested that biophysical tech- niques characterize artifactual interactions occurring due to high non-physiological protein expression [16]. However, results obtained in the present study, concerning serotonin 5-HT1Aand adenosine A2Are- ceptors and their interactions with the appropriate a subunits of G protein further confirm that the use of advanced fluorescence methodology indeed allows us to observe the true interactions. Adenosine A2Arecep- tor did not interact with Gai3, and serotonin 5-HT1A

did not interact with GasL, while the physical contact of these receptors with their appropriate a subunit partners could indeed have been observed, despite the identical level of overexpression of proteins in all studied combinations.

The experiments described above can also serve as a control which must be always performed when us- ing FRET to determine if two proteins interact. A con- trol that should be carried out is to express (preferen- tially using the same expression construct in all ex- periments) two non-interacting fusion proteins in the same cells that carry CFP and YFP and show that there was no FRET fluorescence after normalizing and making corrections for the cross-talk [15]. In the experiments concerning the interactions of the recep- tors under investigation that was exactly the case:

FRET was only observed while the receptor was co- expressed with the appropriatea subunit of G protein, and not in other cases. Although there is a discussion in the literature concerning the possibilities of photo-

(13)

conversion of YFP into a CFP-like species during ac- ceptor photobleaching FRET experiments [43], how- ever, we, as well as others, [41, 44] can exclude that, under standard conditions used, such photoconversion interferes with FRET measurements.

In conclusion, the present experiments further vali- date the use of advanced fluorescence techniques in the studies of oligomerization of membrane proteins.

We find the fluorescence lifetime measurements to be an especially useful and very sensitive tool to quanti- tatively monitor physical interactions between the re- ceptor proteins, both constitutive as well as in the re- sponse to specific ligands. The use of that approach would make a complete understanding of the mecha- nisms of receptor oligomerization easier, which in turn may have a great functional implications and im- pact on GPCRs pharmacology.

Acknowledgments:

We would like to dedicate this work to the memory of the late Professor Zygmunt Wasylewski, whose profound knowledge of fluorescence spectroscopy has made these studies possible.

The authors thank Olympus Polska for the possibility to use the fluorescence microscope, Olympus IX 71. This work was supported by grants from the Ministry of Science (2P05A 071 29 and P04A 070 29).

References:

1. Ayoub MA, Couturier C, Lucas-Meunier E, Angers S, Fossier P, Bouvier M, Jockers R: Monitoring of ligand- independent dimerization and ligand-induced conforma- tional changes of melatonin receptors in living cells by bioluminescence resonance energy transfer. J Biol Chem, 2002, 277, 21522–21528.

2. Barney C, Danuser G: FRET or no FRET: a quantitative comparison. Biophys J, 2003, 84, 3992–4010.

3. Breit A, Lagace M, Bouvier M: Hetero-oligomerization between beta2- and beta3-adrenergic receptors generates a beta-adrenergic signaling unit with distinct functional properties. J Biol Chem, 2004, 279, 28756–28765.

4. Bulenger S, Marullo S, Bouvier M: Emerging role of homo- and heterodimerization in G-protein-coupled re- ceptor biosynthesis and maturation. Trends Pharmacol Sci, 2005, 26, 131–137.

5. Canals M, Burgueno J, Marcellino D, Cabello N, Canela EI, Mallol J, Agnati L et al.: Homodimerization of adenosine A2A receptors: qualitative and quantitative assessment by fluorescence and bioluminescence energy transfer. J Neurochem, 2004, 88, 726–734.

6. Canals M, Marcellino D, Fanelli F, Ciruela F, de Bene- detti P, Goldberg SR, Neve K et al.: Adenosine A2A- dopamine D2 receptor-receptor heteromerization: quali- tative and quantitative assessment by fluorescence and

bioluminescence energy transfer. J Biol Chem, 2003, 278, 46741–46749.

7. Cvejic S, Devi LA: Dimerization of the delta opioid re- ceptor: implication for a role in receptor internalization.

J Biol Chem, 1997, 272, 26959–26964.

8. Devi LA: Heterodimerization of G-protein-coupled re- ceptors: pharmacology, signaling and trafficking. Trends Pharmacol Sci, 2001, 22, 532–537.

9. Dinger MC, Bader JE, Kobor AD, Kretzschmar AK, Beck-Sickinger AG: Homodimerization of neuropeptide Y receptors by fluorescence resonance energy transfer in living cells. J Biol Chem, 2003, 278, 10562–10571.

10. Dziedzicka-Wasylewska M, Faron-Górecka A, Andrecka J, Polit A, Kuœmider M, Wasylewski Z: Fluo- rescence studies reveal heterodimerization of dopamine D1and D2receptors in the plasma membrane. Biochem- istry, 2006, 45, 8751–8759.

11. Fredholm BB, Cunha RA, Svenningsson P: Pharmacolo- gy of adenosine A2A receptors and therapeutic applica- tions. Curr Top Med Chem, 2002, 3, 1349–1364.

12. Hansen JL, Sheikh SP: Functional consequences of 7TM receptor dimerization. Eur J Pharm Sci, 2004, 23, 301–317.

13. Hensler JG: Regulation of 5-HT1A receptor function in brain following agonist or antidepressant administration.

Life Sci, 2003, 72, 1665–1682.

14. Issafras H, Angers S, Bulenger S, Blanpain C, Parmen- tier M, Labbe-Jullie C, Bouvier M et al.: Constitutive agonist-independent CCR5 oligomerization and anti- body-mediated clustering occurring at physiological lev- els of receptors. J Biol Chem, 2002, 277, 34666–34673.

15. Janetopoulos C, Devreotes P: Monitoring receptor-me- diated activation of heterotrimeric G-proteins by fluores- cence resonance energy transfer. Methods, 2002, 27, 366–373.

16. Kroemer KM, Pfleger KD, Eidne KA: G-protein coupled receptor oligomerization in neuroendocrine pathways.

Front Neuroendocrinol, 2003, 24, 254–278.

17. Lakowicz JR: Principles of Fluorescence Spectroscopy.

Kluwer Academic/Plenum Publishers, New York, 1999.

18. Lee SP, So CH, Rashid AJ, Varghese G, Cheng R, Lanca AJ, O’Dowd et al.: Dopamine D1 and D2 receptor co- activation generates a novel phospholipase C-mediated calcium signal. J Biol Chem, 2004, 279, 5671–35678.

19. Levac BA, O’Dowd BF, George SR: Oligomerization of opioid receptors: generation of novel signaling units.

Curr Opin Pharmacol, 2002, 2, 76–81.

20. Lucki I, Singh A, Kreiss DS: Antidepressant-like beha- vioral effects of serotonin receptor agonists. Neurosci Biobehav Rev, 1994, 18, 85–95.

21. Malec D, Poleszak E: Involvement of adenosine recep- tors in dizocilpine-induced motor activity in mice. Phar- macol Rep, 2006, 58, 101–106.

22. McIntire WE, MacCleery G, Garrison JC: The G protein beta subunit is a determinant in the coupling of Gs to the beta 1-adrenergic and A2A adenosine receptors.

J Biol Chem, 2001, 276, 15801–15809.

23. McVey M, Ramsay D, Kellett E, Rees S, Wilson S, Pope AJ, Milligan G: Monitoring receptor oligomeriza- tion using time-resolved fluorescence resonance energy transfer and bioluminescence resonance energy transfer.

(14)

The human delta-opioid receptor displays constitutive oligomerization at the cell surface, which is not regulated by receptor occupancy. J Biol Chem, 2001, 276, 14092–14099.

24. Mercier JF, Salahpour A, Angers S, Breit A, Bouvier M:

Quantitative assessment of beta 1- and beta 2-adrenegic receptor homo- and heterodimerization by biolumines- cence resonance energy transfer. J Biol Chem, 2002, 277, 44925–44931.

25. Milligan G: G protein-coupled receptor dimerization:

function and ligand pharmacology. Mol Pharmacol, 2004, 66, 1–7.

26. Milligan G, Bouvier M: Methods to monitor the quater- nary structure of G protein-coupled receptors. FEBS J, 2005, 272, 2914–2925.

27. Morelli M, Wardas J: Adenosine A2A receptor antago- nists: potential therapeutic and neuroprotective effects in Parkinson’s disease. Neurotox Res, 2001, 3, 545–556.

28. Patel RC, Kumar U, Lamb DC, Eid JS, Rocheville M, Grant M, Rani A et al.: Ligand binding to somatostatin receptors induces receptor-specific oligomer formation in live cells. Proc Natl Acad Sci USA, 2002, 99, 3294–3299.

29. Patel RC, Lange DC, Patel YC: Photobleaching fluores- cence resonance energy transfer reveals ligand-induced oligomer formation of human somatostatin receptor sub- types. Methods, 2002, 27, 340–348.

30. Patterson G, Day RN, Piston D: Fluorescent protein spectra. J Cell Sci, 2001, 114, 837–838.

31. Raymond JR, Mukhin YV, Gettys TW, Garnovskaya MN: The recombinant 5-HT1A receptor: G protein coupling and signaling pathways. Br J Pharmacol, 1997, 127, 1751–1764.

32. Rios CD, Jordan BA, Gomes I, Devi LA: G-protein- coupled receptor dimerization: modulation of receptor function. Pharmacol Ther, 2001, 92, 71–87.

33. Salim K, Fenton T, Bacha J, Urien-Rodriguez H, Bon- nert T, Skynner HA, Watts E et al.: Oligomerization of G-protein-coupled receptors shown by selective co-immunoprecipitation. J Biol Chem, 2002, 277, 15482–15485.

34. Sambrook J, Fritsch EF, Maniatis T: Molecular Cloning:

A Laboratory Manual. Cold Spring Harbor Laboratory Press, New York, 1996.

35. Scarselli M, Novi F, Schallmach E, Lin R, Baragli A, Colzi A, Griffon N et al.: D2/D3 dopamine receptor heterodimers exhibit unique functional properties.

J Biol Chem, 2001, 276, 30308–30314.

36. Seeman P, Guan HC, Civelli O, Van Tol HH, Sunahara RK, Niznik HB: The cloned dopamine D2 receptor re-

veals different densities for dopamine receptor antagonist ligands. Implications for human brain positron emission tomography. Eur J Pharmacol, 1992, 227, 139–146.

37. Seeman P, Van Tol HH: Dopamine receptor pharmacolo- gy. Trends Pharmacol Sci, 1994, 15, 264–270.

38. Stanasila L, Perez JB, Vogel H, Coteechia S: Oligomeri- zation of the alpha 1a- and alpha 1b-adrenergic receptor subtypes. Potential implications in receptor internaliza- tion. J Biol Chem, 2003, 278, 40239–40251.

39. Strange PG: Dopamine D4 receptors: curiouser and curi- ouser. Trends Pharmacol Sci, 1994, 15, 317–319.

40. Terrillon S, Durroux T, Mouilac B, Breit A, Ayoub MA, Taulan M, Jockers R et al.: Oxytocin and vasopressin V1a and V2 receptors form constitutive homo- and heterodimers during biosynthesis. Mol Endocrinol, 2003, 17, 677–691.

41. Thaler C, Vogel SS, Ikeda SR, Chen H: Photobleaching of YFP does not produce a CFP-like species that affects FRET measurements. Nat Methods, 2006, 3, 491.

42. Tucker AL, Jia LG, Holeton D, Taylor AJ: Dominance of G(s) in doubly G(s)/G(i)-coupled chimeric A(1)/A(2A) adenosine receptors in HEK-293 cells. Biochem J, 2000, 352, 203–210.

43. Valentin G, Verheggen C, Piolot T, Neel H, Coppey- Moisan M, Bertrand E: Photoconversion of YFP into a CFP-like species during acceptor photobleaching FRET experiments. Nat Methods, 2005, 2, 801.

44. Verrier SE, Söling HD: Photobleaching of YFP does not produce a CFP-like species that affects FRET measure- ments. Nat Methods, 2006, 3, 491–492.

45. Vila-Coro AJ, Mellado M, Martin de Ana A, Lucas P, del Real G, Martinez AC, Rodriguez-Frade JM: HIV-1 infection through the CCR5 receptor is blocked by re- ceptor dimerization. Proc Natl Acad Sci USA, 2000, 97, 3388–3393.

46. Vilardaga JP, Bunemann M, Krasel C, Castro M, Lohse ML: Measurement of the millisecond activation switch of G protein-coupled receptors in living cells. Nat Bio- technol, 2003, 21, 807–812.

47. Wreggett KA, Wells JW: Cooperativity manifest in the binding properties of purified cardiac muscarinic recep- tors. J Biol Chem, 1995, 270, 22488–22499.

48. Wurch T, Matsumoto A, Pauwels PJ: Agonist-indepen- dent and dependent oligomerization of dopamine D(2) receptors by fusion to fluorescent proteins. FEBS Lett, 2001, 507, 109–113.

Received:

May 28, 2007; in revised form: August 2, 2007.

Cytaty

Powiązane dokumenty

structural modifications of pyrimido- or diazepino- [2,1-f]-purines on the 5-HT 1A receptors affinity and 5-HT 1A /5-HT 2A selectivity; in consequence, pharma- cological assays

To substantiate the role of 5-HT 1B receptors in in- centive motivation for amphetamine, in the present paper, we used an extinction/reinstatement model to examine the effects of

synaptic 5-HT ) receptor activation. Gómez-Wong E, Martí MJ, Tolosa E, Valls-Solé J: Sen- sory modulation of the blink reflex in patients with ble- pharospasm. Grillon C, Ameli

In addition, intra-SNc injection of 8-hydroxy-2-[di-n-propylamino]tetralin (8-OH-DPAT; 10 µg/rat), a 5-HT 1A receptor agonist, decreased 6-OHDA-induced catalepsy. Our study

The aim of the present study was to investigate whether the selective pharmacological blockade or activation of 5-HT 6 receptors altered the maintenance of cocaine

In order to ascertain whether the integrity of 5-HT neurons was necessary to reveal the antidepres- sant-like activity of the selective 5-HT 6 receptor an- tagonist, the

The most striking result was that in the saturation radioligand binding experiments on HEK293 cells transiently transfected with the respective h5-HT 3A receptor subunit cDNA, B max

A new class of arylpiperazine derivatives: The library synthesis on SynPhase lanterns and biological evaluation on serotonin 5-HT 1A and 5-HT 2A receptors. Scott, W.L.;