Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Neurochemistry International 55 (2009) 467–475

Contents lists available at ScienceDirect

Neurochemistry International
journal homepage: www.elsevier.com/locate/neuint

Comparative pharmacology of adrenergic a2C receptors coupled to


Ca2+ signaling through different Ga proteins
Dalma Kurko *, Zsófia Bekes, Anikó Gere, Andrea Baki, András Boros, Sándor Kolok,
Gyula Bugovics, József Nagy, Zsolt Szombathelyi, Györgyi Ignácz-Szendrei
Pharmacological and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary

A R T I C L E I N F O A B S T R A C T

Article history: Adrenergic a1, a2 and b receptors are members of the G-protein-coupled receptor families (GPCRs)
Received 9 March 2009 mediating physiological responses to adrenaline (epinephrine) and noradrenaline (norepinephrine).
Received in revised form 24 April 2009 Since GPCRs are major targets for potential therapeutic agents, development of robust, reliable and cost
Accepted 28 April 2009
effective functional screening methods for these receptors is in the focus of pharmacological research.
Available online 6 May 2009
For this reason, the aim of the present study was to develop an intracellular calcium assay for
investigating the pharmacology of the a2C type of adrenergic receptors (a2C-AR). Although activation of
Keywords:
a2C-AR is not linked to calcium mobilization, co-expression of these receptors with the chimeric Gaqi5
a2C-AR
Chimeric Gaqi5 protein
protein, containing the five carboxyl-terminal amino acids from Gi, or promiscuosus Ga16 protein can
Promiscuosus Ga16 protein divert receptor signaling to the Gq pathway generating Ca2+ release from intracellular stores.
Fluorometric Ca2+ assay In order to assess the functional potency of a2-AR agonists and antagonists, we established a
High-throughput screening fluorometric Ca2+ assay using cell lines stably and constitutively co-expressing a2C-AR and Gaqi5 or Ga16
proteins (Gaqi5/a2C and Ga16/a2C). As part of the pharmacological characterization, we measured the
changes in cytoplasmic Ca2+ levels due to activation of the chimeric Gaqi5 or Ga16 coupled recombinant
a2C receptors as a function of increasing concentration of several agonists (noradrenaline, brimonidine,
oxymetazoline, clonidine, moxonidine) and antagonists (MK912, yohimbine). The binding affinities of
a2-AR agonist and antagonists and the inhibition of the forskolin-stimulated cAMP accumulation in a2C-
AR expressing cells were also measured. These results confirmed that the Gaqi5/a2C and Ga16/a2C
recombinant systems can be useful for modelling the native Gi-coupled system. Our results indicate that
a plate-reader based fluorometric Ca2+ assay may be suitable in high-throughput screening for a2C-AR
ligands as well.
ß 2009 Elsevier Ltd. All rights reserved.

1. Introduction depression, obesity, diabetes (Serle et al., 1991; Ruffolo et al.,


1993,1995; Ruffolo and Hieble, 1994; Lakhlani et al., 1997;
The a2-adrenergic receptors (a2-ARs) constitute a distinct Arnsten, 1998; Kable et al., 2000; Crassous et al., 2007; Sanders
family of catecholamine receptors which mediate physiological and Maze, 2007). Several line of evidence suggests that a2-ARs may
responses to noradrenaline and adrenaline. Pharmacological and also play a role in gastric mucosal defense pointing to the
molecular biological studies indicate that a2-ARs consist of three gastroprotective effect of the a2-AR agonists (Gyires et al., 2007).
homologous subtypes: namely a2A, a2B, and a2C (Bylund, 1992, Moreover, as presynaptic release-modulating receptors represent
2005; Hieble et al., 1995; Docherty, 1998; Calzada and Artinano, promising targets for pharmacological intervention it is possible
2001). There are some reports of existence of a fourth subtype, a2D, that a new generation of drugs with actions on transmitter release
but it is generally acknowledged to be the rodent homologue of the through autoreceptors and heteroreceptors may become a
human a2A-AR subtype (Link et al., 1992; MacDonald et al., 1997). generation of novel drugs with useful therapeutic properties,
The a2-adrenergic receptor subtypes play a pivotal role in many including some unmet medical needs of today (Langer, 2008).
diverse cell functions and physiological processes, therefore these However, the therapeutic use of the currently available a2-
receptors have been considered as important drug targets for drugs is limited because of severe side effects, including sedation
treatment of pain, hypertension, ischemia, glaucoma, spasticity, and cardiovascular effects (Kamibayashi and Maze, 2000; Maze
and Fujinaga, 2000). Recent studies on transgenic animals lacking
or overexpressing different a2-AR subtypes have greatly enhanced
* Corresponding author. Fax: +36 1 8898400. our understanding of subtype specific physiological functions and
E-mail address: d.kurko@richter.hu (D. Kurko). have highlighted the potential for the development of subtype

0197-0186/$ – see front matter ß 2009 Elsevier Ltd. All rights reserved.
doi:10.1016/j.neuint.2009.04.015
468 D. Kurko et al. / Neurochemistry International 55 (2009) 467–475

selective therapeutic agents with improved efficacy and/or More recently, functional assays that measure various signaling
reduced side effects when compared to existing drugs (Lakhlani events downstream to receptor activation have become widely
et al., 1997; Kable et al., 2000; Scholz and Tonner, 2000; Philipp utilized (Chambers et al., 2003; Monteith and Bird, 2005). Ca2+
et al., 2002; Knaus et al., 2007). Therefore, screening of compound assays are now recognized as efficient tools for the characteriza-
libraries, molecular modeling and synthesis of novel adrenergic tion of GPCRs signaling via Gq proteins. Ca2+ measurement can be
agonists and antagonists without therapy-limiting side effects, an option for evaluation of non-Gq-coupled receptors as well using
with better subtype selectivity and specificity are still under chimeric and/or promiscuous G-proteins (Kassack et al., 2002;
thorough investigation (Wong et al., 1994, 2000; Balogh et al., Eglen, 2005; New and Wong, 2005). It has been reported that the
2007). As such, cell lines that stably express a2C-ARs are extremely carboxyl-terminal end of the a subunit of G-proteins plays an
useful in identifying selective ligands of this receptor class. important role in the specific interaction between the G-protein
a2-ARs belong to the GPCR superfamily, which comprises one and the receptor. Accordingly, the exchange of three to nine amino
of the largest gene families in the human genome (Ji et al., 1998; acids of the C-terminal residues of the PLC-b-activating Ga
Perez, 2005). Physiological signaling pathways linked to the a2- subunit, i.e. the Gaq, by those of either Gai or Gao is sufficient to
AR subtypes are mostly mediated by pertussis toxin-sensitive G- allow these chimeric subunits to alter the signaling pathways of
proteins (Gi1, Gi2, Gi3 and Go) resulting in inhibition of adenylyl- many GPCRs (Conklin et al., 1993; Conklin et al., 1996; Liu et al.,
cyclase (AC) activity and consequent decreases in cellular cAMP, 1995; Kostenis et al., 1997). It has also been reported that the
activation of receptor operated K+ channels and inhibition of promiscuous Ga15 and Ga16 proteins, that activate PLC-b, can
voltage gated Ca2+ channels (Limbird, 1988; Abdulla and Smith, activate different GPCRs, including those recognized not to
1997; Saunders and Limbird, 1999). Stimulatory effects of a2-AR stimulate PLC-b per se (Offermanns and Simon, 1995; Milligan
agonists have also been described, including coupling to et al., 1996; Kostenis, 2001). To date, however, it is impossible to
stimulatory cholera toxin-sensitive Gs proteins with increased predict a priori whether a particular GPCR can interact effectively
cAMP generation (Eason et al., 1992, 1994; Pohjanoksa et al., with a specific chimeric or promiscuous G-protein.
1997; Jasper et al., 1998) and Ca2+ mobilization (Kukkonen et al., In order to identify an efficient receptor–G-protein combination
1998; Reynen et al., 2000). Although the preferential transduc- and to provide increased sensitivity as well as a high signal-to-
tion pathway of the a2C-subtype is through the inhibitory Gi/o noise ratio for ligand screens at a2C-ARs, we developed stable
proteins (Pohjanoksa et al., 1997), in recombinant systems they transfected cell lines co-expressing a2C-ARs with either chimeric
can also activate Gq when aq subunit is overexpressed in HEK- Gaqi5 or promiscuous Ga16 proteins. We report the successful
293 cells (Conklin et al., 1992). Ca2+ mobilization by a2C-ARs has application of these G-proteins for functional characterization of
also been demonstrated in CHO-K1 cells stably expressing the ligands acting at a2C-AR by establishing a fluorometric Ca2+ assay
receptor; however, the pertussis toxin sensitivity of this compatible even with high-throughput evaluation.
response points that it is likely to be mediated by the bg
subunits of Gi/o proteins (Kukkonen et al., 1998). Furthermore, 2. Experimental procedures
GPCR signaling exhibits even greater diversity and complexity if 2.1. Generation of stably transfected cell lines expressing recombinant human a2C-ARs
several receptors are simultaneously activated since the signal and chimeric Gaqi5 or promiscuous Ga16 proteins
transduction pathways can cross-talk or interact (Fresco et al., Chinese hamster ovary (CHO-K1) cells expressing a2C-ARs (a2C-C1 cell line) were
2007). purchased from Euroscreen (cat. nr. ES-032-C, Bruessels, Belgium). Sequencing
Since GPCRs represent one of the most important drug revealed that the DNA region encoding human a2C-AR was identical to the gene
discovery targets, a variety of functional assays are available for published by GenBank with accession number of AY605898.1, corresponding to the
haplotype 1 of the human a2C-AR (i.e. an isoform where the sequence 322–325 is
screening GPCR ligands (Thomsen et al., 2004, 2005). Historically,
not deleted); it codes for a protein identical to that available in GenBank under the
screening for GPCR agonists or antagonists has been performed by accession number of AAT02221.1. Expression vectors pCEP-Gqi5-HA and pCEP-
filter binding assays utilizing high-affinity radioligands. However, Ga16 encoding the chimeric Gaqi5 and promiscuous Ga16 proteins, respectively,
an ideal screening system should be simple, non-radioactive, were obtained from Molecular Devices (Sunnyvale, CA, USA). For transfection a2C-
robust (i.e. characterized by high signal-to-noise ratio), homo- C1 cells were grown on 96-well plates to 50% confluency in Ham’s F12 medium
(nutrient mixture Ham’s F12 + L-Glutamine (GIBCO BRL, Gaithersburg, MD, USA),
genous, suitable in a microtiter plate format (96-, 384- or 1536-
containing 10% fetal bovine serum (GIBCO BRL), 2.5 mg/ml amphotericin B, 100 U/
well) and also utilizing minimal reagent addition steps to facilitate ml penicillin G, 100 mg/ml streptomycin and 400 mg/ml G418 (Invitrogen, Carlsbad,
robotic automation (Milligan and Rees, 1999). Further, these CA, USA). Cells were incubated with Gaqi5 or Ga16 cDNAs containing vectors (5 mg/
screening methods should allow a simple functional discrimina- ml) for 4 h in serum-free F12 in the presence of lipofectamine (Pfx-7, Invitrogen) in
a humidified atmosphere with 5% CO2 at 37 8C. Following the 4 h transfection
tion between agonists and antagonists and be applicable for a great
period cells were placed in serum containing F12 medium and next day they were
variety of receptors. One of the widely used methods for studying exposed to selection medium, containing hygromycin (200 mg/ml, Invitrogen) and
signaling through Gi proteins is the measurement of incorporation G418 (400 mg/ml, Invitrogen). The medium was replaced every 2 or 3 days for 2
of radiolabelled GTPgS into activated G-proteins. Since this weeks. Stable cell colonies were isolated and assayed for calcium responses using
method uses radioisotopes and requires purification of membrane the FlexStation II (described below).

components, furthermore, it lacks the advantages of intrinsic


signal amplification that occurs in a whole cell (Cerione, 1991), it is 2.2. Determination of cytosolic free concentration of calcium ions
not the most appropriate method as a drug screening assay. Changes in the cytoplasmic calcium ([Ca2+]i) were measured using the Ca2+-
Moreover, this assay is generally limited to Gi/o-coupled receptors, sensitive dye, fluo-4AM (Molecular Probes, Eugene, OR). Cells were seeded into 96-
because Gi/o proteins have a faster GDP–GTP exchange rate than well plates at a density of 40,000 cells/well and maintained overnight in a tissue
other G-proteins (Milligan, 2003). As activation of the Gi proteins culture incubator at 37 8C under an atmosphere of 5% CO2. The growth medium was
aspirated from the cells and replaced with 2 mM fluo-4AM in extracellular solution
results in a decreased AC activity, measuring the inhibition of
(140 mM NaCl, 5 mM KCl, 5 mM HEPES-Na, 5 mM HEPES, 2 mM CaCl2, 2 mM MgCl2,
forskolin-stimulated cAMP accumulation is another popular 10 mM glucose, pH 7.4). Cell loading was enhanced by premixing the hydrophobic
method for studying Gi signaling (Wong, 1994; Gabriel et al., dye AM-ester with low levels (final concentration 0.02%) of Pluronic-127 (Sigma–
2003). Since this assay generally has a low signal-to-noise ratio Aldrich, St. Louis, MO, USA). To inhibit the efflux of the dye from the cells during and
following loading 2 mM probenecid (anion transporter inhibitor) was added to the
(Williams, 2004) and a limited dynamic range, furthermore it is
cells. After the incubation period (60 min, 37 8C, 5% CO2/95% air), unloaded dye was
expensive, time-consuming, and because the inhibition of the removed from the cells by washing and 100 ml fresh extracellular medium with or
stimulation is rarely exceeds 60%, it is not ideal for higher without an a2-AR antagonist was added to the wells and the plate was incubated at
throughput screening. 37 8C for an additional 10 min. The plate was transferred to FlexStation II (Molecular
D. Kurko et al. / Neurochemistry International 55 (2009) 467–475 469

Devices, Sunnyvale, CA), a plate-reader fluorometer with integrated 8-channel fluid cDNA was performed using the Ready-To-Go You-Prime First-Strand Beads Kit
addition capability. Fluorescence measurements were carried out at 37 8C. The dye (Amersham Biosciences, New Jersey, USA) following the manufacturer’s instruc-
was excited at 485 nm, emission was sampled at 538 nm at 1.4-s intervals. The basal tions. The 20 ml total volume of the PCR reaction solution contained 1 ml cDNA
(unstimulated) level of cytosolic Ca2+ ion concentration was recorded for 20 s template from each sample, 1–1 ml forward and reverse primer, 10 ml 2 Master
followed by agonist stimulation. The 25 ml 5 concentrated agonist solution was Mix (Promega Corporation, Madison, WI, USA) and 7 ml dH2O.
added to the cells using the pipettor of FlexStation and fluorescence was monitored for Primers were designed using the Primer 2.0 software and were synthesized by
an additional 40 s. Results were expressed as DF/F values using SOFTMax Pro software IDT (Leuven, Belgium). Sequences of Gaqi5 and Ga16 specific primers generating an
(Molecular Devices), where F was the resting fluorescence preceding agonist 293 and 186 base-pair (bp) amplicons, respectively, were as follows: forward
application and DF was the increase in fluorescence at a given time (DF = maximum primer: 50 -CTCTGGAGTCCATCATGGCA-30 , reverse primer: 50 -GATCTTGAGCGTGTC-
fluorescence intensity values after stimulation minus average fluorescence intensity CATCG-30 ; and forward primer: 50 -GCTACCTGGACGAGATCAAC-30 , reverse primer:
values for unstimulated level of cytosolic Ca2+ ion concentration). In all experiments, 50 -TGAGAGGCAGAGAAGAGAGC-30 . PCR analysis was performed with a Biometra T
all treatments on the plates were measured in multiple wells (4–12) in parallel, and Gradient instrument. The PCR program consisted of an initial denaturation phase of
the mean DF/F values or inhibition percentages of those were used for analysis. 5 min at 95 8C, 20 cycles of denaturation at 94 8C (60 s), annealing at 55 8C (30 s) and
Sigmoid curves were fitted to the concentration–response data to obtain EC50 or IC50 extension at 72 8C (60 s) and a Wnal synthesis step of 10 min at 72 8C. The PCR assay
values. Curve fitting was done by Microcal Origin 6.0 software. For agonists, relative was performed simultaneously with negative control samples (cDNA obtained from
efficacy (Emax) values were calculated as fitted maximum of each concentration–effect non-transfected a2C-C1 cells and sterile water). The resulting PCR products were
curve expressed as a percentage of the response observed with a maximally effective separated using a Bioanalyzer 2100 Instrument (Agilent Technologies, Santa Clara,
concentration of brimonidine (UK14,304; 1 mM) from the same plate. All experiments CA, USA) and a DNA 7500 LabChip Kit. The specific PCR products were analyzed with
were performed at least three times, on different days. Data were expressed as Agilent Technologies 2100 Expert Software (Version Nr: B.02.02.SI238)
mean  S.E.M., unless otherwise stated.
2.6. Immunocytochemical analysis of Gaqi5
2.3. Measurement of intracellular cAMP levels
Internal hemagglutinin (HA) epitope in Gaqi5 cDNA allows analysis of protein
Chinese hamster ovary CHO-K1 cells expressing recombinant a2C-ARs were expression using flow cytometry based immunocytochemistry. CHO-K1 cells stably
grown as described above. For the measurements cells were seeded in 384-well plates expressing a2C-ARs and Gaqi5 proteins were cultured on 24-well plates. Cells were
and cultured overnight at 37 8C and 5% CO2. Following medium replacement with suspended in Dulbecco’s phosphate buffered saline (D-PBS), and then fixed in 4%
HBSS (Hank’s buffered saline solution) supplemented with 50 mM Ro 20-1724 buffered paraformaldehyde for 30 min at room temperature. After three washes,
(phosphodiesterase inhibitor, Sigma) cells were incubated for 20 min at 37 8C/5% CO2. samples were incubated overnight at 4 8C with mouse monoclonal anti-
Cells were then treated with an a2-AR agonist, UK14,304, in a concentration range of hemagglutinin (HA)–FITC antibody (1:20, Roche, Basel, Switzerland) in D-PBS
3 nM to 3 mM for 15 min and were stimulated with forskolin (1 mM) for an additional containing 0.2% Triton X-100 and 2% BSA. The same staining procedure was applied
15 min at 37 8C. In antagonists experiments, cells were treated first with MK912 to Gaqi5 non-transfected a2C-C1 cells as well to determine the non-specific staining
(25 mM), and after 15 min incubation, with UK14,304 (25 mM). After agonist properties of the antibody. After washing, the FITC fluorescence was analyzed using
treatment cells were also incubated for 15 min, followed by forskolin (0.03–100 mM) a FACScan flow cytometer (Becton Dickinson, San Jose, CA, USA), equipped with
treatment and an additional 15 min incubation (37 8C; 5% CO2). Stimulated cells were 15 mW air-cooled 488 nm argon ion laser. Fluorescence signals were measured in
lysed and cAMP levels were measured immediately via a competitive, fluorescence the Fl-1 window (530  30 nm band pass filter). Data of 3000 events per sample were
intensity based immunoassay. Concentration of cAMP was determined with the recorded in the list mode at a flow rate of approximately 300 cell/s following
CatchPointTM kit (Molecular Devices) according to the manufacturer’s instructions. logarithmic amplification. Data were analyzed using CellQuest software (version: 3.1.f;
cAMP, derived from treated and lysed cells competes with horseradish peroxidase- Becton Dickinson, San Jose, CA, USA). Cells were gated in the forward scatter (FSC) vs.
labelled cAMP conjugate for binding sites on the anti-cAMP antibodies. After a single side scatter (SSC) dot plot. The arithmetic means of the FITC-fluorescence intensities
wash step, any unbound HRP–cAMP is removed. In the absence of cAMP, most of the were calculated from fluorescence histograms for the gated population.
HRP–cAMP is bound to the antibody. Increasing concentrations of cAMP competi-
tively decrease the amount of bound conjugate, thus decreasing the measured HRP 2.7. Drugs
activity. In the presence of HRP and hydrogen peroxide, a fluorescent substrate is
oxidized. The recorded fluorescent intensity (excitation: 530 nm; emission: 590 nm) UK14,304 (5-bromo-N-[4,5-dihydro-1H-imidazol-2-yl]-6-quinoxalinamine),
is proportional to the amount of HRP and inversely proportional to the cAMP oxymetazoline (2-(3-hydroxy-2,6-dimethyl-4-t-butylbenzyl)-2-imidazoline), clo-
concentration present in the reaction. Fluorescence was measured with the Polarstar nidine (2-[2,6-dichloroaniline]-2-imidazoline HCl), moxonidine (4-chloro-N-(4,5-
Optima (BMG Labtech) multimode microplate reader. The fluorescence intensity dihydro-1H-imidazol-2-yl)-6-methoxy-2-methylpyrimidin-5-amine), yohimbine,
values of the forskolin-stimulated samples were extracted from those of the MK912 ((2S,12bS)10 ,30 -dimethylspiro(1,3,4,50 ,6,60 ,7,12b-octahydro-2H-benzo[b]-
unstimulated control samples (the highest value). The DRFU calculated is furo[2,3-a]quinolizine)-2,40 -pyrimidin-20 -one), U-73122 (1-(6-((17b-3-methox-
proportional to the cAMP concentration present in the sample. yestra-1,3,5(10)-trien-17-yl)amino)hexyl)-1H-pyrrole-2,5-dione) were obtained
from Sigma–Aldrich.
2.4. Radioligand binding assay

Membranes were prepared from CHO-K1 cells stably expressing human a2C-ARs.
3. Results
Cells were harvested and centrifuged at 500  g. The cell pellet was broken with a
Polytron PTA 10 S homogenizer (setting 5, 30 s burst) in ice-cold homogenization buffer 3.1. Cell lines co-expressing recombinant human a2C-ARs with
containing 5 mM Tris–HCl, 5 mM EDTA, 5 mM EGTA, 0.1 mM phenylmethylsulfonyl chimeric Gaqi5 or promiscuous Ga16 proteins
fluoride (PMSF); pH 7.4, followed by two consecutive centrifugation steps at 30,000  g
for 15 min separated by a wash step in the same buffer. Membranes were resuspended
in TRIS buffer containing 50 mM Tris–HCl, 1 mM EDTA, 5 mM MgCl2, 0.1 mM PMSF; pH After transfection of a2C-C1 cells with pCEP-Gqi5-HA or pCEP-
7.4. Protein content was determined and was adjusted to 1 mg protein/ml. The final Ga16 expression vectors, we obtained cell lines stably co-
suspension was aliquoted and was frozen at 70 8C. For saturation analysis the expressing human a2C-ARs with chimeric Gaqi5 or promiscuous
radiolabelled agonist ligand [3H]UK14,304 was used in a concentration range of 0.1– Ga16 proteins, respectively. More than two dozens of cell colonies
15 nM with an incubation time of 30 min at room temperature.
To determine the binding affinities of some reference drugs the radioligand
resistant for the selecting agents (i.e. hygromycin and neomycin)
[3H]UK14,304 was used in a concentration of 2 nM. Non-specific binding was were picked and pre-screened for functional activity by assessing
determined in the presence of phentolamine (10 mM). The binding procedure was UK14,304-evoked cytoplasmic Ca2+ release. Stable expression of
stopped by filtration on Unifilter GF/B glass-fiber filter presoaked in 0.5% the a2C-AR in CHO-K1 cells alone did not result in Ca2+ elevation in
polyethyleneimine. The filters were washed five times with 1 ml TRIS buffer and
response to 1 mM UK14,304 stimulation, but co-expression of the
the radioactivity of the filters was counted by liquid scintillation spectrometry in a
Pachard TopCount NXT microplate scintillation 8 luminescence counter. All receptor with chimeric Gaqi5 or promiscuous Ga16 proteins
experiments were performed at room temperature. IC50 values were calculated resulted in robust Ca2+ responses with a high signal-to-background
from concentration displacement curves by sigmoidal fitting using Microcal Origin ratio. The maximal changes in relative fluorescence units were up
6.0 software, Ki values were calculated using the Cheng–Prusoff equation (Cheng to 45,000 and 20,000, respectively (Fig. 1). UK14,304 generated
and Prusoff, 1973).
typical responses consisting of a rapid initial peak in fluorescence
(maximal 8–12 s after agonist addition) followed by a rapid decline
2.5. Determination of mRNA expression by RT-PCR
towards the basline (over a period of 40 s).
Total RNA from transfected and non-transfected a2C-C1 cells was extracted using Two clones, designated as Gaqi5/H7 and Ga16/B6, which
the RNeasy Mini Kit (Qiagen, Hilden, Germany). Reverse transcription (RT) into exhibited the highest agonist-induced [Ca2+]i responses, were
470 D. Kurko et al. / Neurochemistry International 55 (2009) 467–475

Fig. 1. Ca2+ response in CHO-K1 cells expressing the a2C-AR alone (circle) or co-
transfected with chimeric Gaqi5 (square) or promiscuous Ga16 proteins (triangle)
following stimulation with 1 mM UK14,304. Cells were loaded with the Ca2+
indicator dye Fluo-4, transferred to FlexStationII and after recording baseline (0–
20 s) stimulated with UK14,304. Raw fluorescence data from a representative
experiment is shown with each data point representing the mean value  S.E.M.
from 6 wells.

chosen and characterized further in subsequent experiments. RT-


PCR analysis of the expression of Gaqi5 or Ga16 mRNA clearly
verified the expression of these mRNAs in both cells lines (data not
shown). The expression of the Gaqi5 protein tagged with
hemagglutinin (HA) could be detected by a flow cytometry based
immunocytochemical method as well. The level of expression was
assessed by comparing the fluorescence histograms obtained from
measurements on the non-expressing (a2C-C1) and the Gaqi5
expressing (Gaqi5/H7) cells stained with anti-HA–FITC antibody.
The intensity of specific immunolabelling was about six times
higher in Gaqi5/H7 compared to a2C-C1 cells (data not shown).
The mechanism of agonist-evoked increase of cytoplasmic Fig. 2. (a) Concentration-dependence of agonist-induced Ca2+ responses in Gaqi5/
calcium was also examined in the two different G-protein H7 cells co-expressing the a2C-AR and chimeric Gaqi5 proteins. Agonist responses
were expressed as a percentage of the response observed with a maximally effective
expressing cell lines and was found to be consistent with Gq concentration of UK14,304 (1 mM). Data points represent mean  S.E.M. from three
signal transduction. 5 min preincubation with the PLC inhibitor U- to seven independent experiments. (b) Concentration-dependence of agonist-induced
73122 reduced Ca2+ elevation in response to UK14,304 (IC50 values Ca2+ responses in Ga16/B6 cells co-expressing the a2C-AR and promiscuous Ga16
were 1.51  0.18 and 1.36  0.12 mM in Gaqi5/H7 and Ga16/B6 cell proteins. Agonist responses were expressed as a percentage of the response observed
with a maximally effective concentration of UK14,304 (1 mM). Data points represent
lines, respectively).
mean  S.E.M. from three to seven independent experiments.
Functional stability of the generated cell lines was also
monitored by comparing the maximum responses to UK14,304
observed in cells following several passages. We could detect adrenaline > moxonidine > oxymetazoline > clonidine. In compari-
proper calcium responses up to 30 passages and the DF/F values son to UK14,304, whose efficacy was considered as 100%,
(see Section 2) were stable during this period. noradrenaline displayed similar maximal efficacies in Gaqi5/H7 and
Ga16/B6 cells (97.4  0.4% and 95.8  2.5%) while moxonidine (90.3 
3.2. Pharmacological properties of recombinant human a2C-ARs 3.9% and 80.1  2.2%, respectively), oxymetazoline (82.3  2.7% and
61.5  2.7%, respectively) and clonidine (77.3  0.6% and 46.3  7.1%,
Further detailed characterization of the a2C-AR-coupled Ca2+ respectively), resulted in lower maximal responses suggesting partial
signal facilitated by chimeric Gaqi5 or promiscuous Ga16 proteins agonist responses for a2C-ARs.
was undertaken using established receptor agonists and antago- To examine the effect of antagonists, cells were pre-treated
nists. All of the agonists tested caused a concentration-dependent with yohimbine and MK912. The compounds tested in presence of
increase in [Ca2+]i; the generated concentration–response curves 40 nM UK14,304 (corresponding approximately to the concentra-
are illustrated in Fig. 2. The rank order of potency for examined tion of the agonist generating 90% activity [EC90]) resulted in a
agonists was UK14,304 (EC50, 1.7  0.1 nM) = noradrenaline (2.1  concentration-dependent inhibition of agonist-induced Ca2+
0.3 nM) > oxymetazoline (2.9  0.5 nM) > clonidine (7.0  0.6 nM) mobilization, as shown in Fig. 3. Calculated IC50 values for
> moxonidine (41.0  4.7 nM) for Gaqi5/H7 cell line and UK14,304 MK912 were 0.73  0.05 nM and 0.20  0.04 nM in Gaqi5/H7 and
(EC50, 5.1  0.3 nM) > oxymetazoline (7.4  1.2 nM) > noradrenaline Ga16/B6 cell lines, respectively. Estimated IC50 values for the
(15.3  2.7 nM) > clonidine (29  3.8 nM) > moxonidine (321  inhibitory effect of yohimbine were 17.8  2.9 nM and
34 nM) for Ga16/B6 cell line, respectively. Although UK14,304 and 3.1  0.8 nM, respectively. None of the antagonists tested increased
noradrenaline were equally efficacious in all the clones, marked [Ca2+]i in any of the cell lines (data not shown). In the case of Ga16/B6
differences in response to the other agonists were seen resulting in a cell line, as it can be seen in Fig. 3b, the maximum inhibition % of
rank order of efficacy as follows in both cell lines: UK14,304 = nor- antagonists varied between 94% and 106%. However, the differences
D. Kurko et al. / Neurochemistry International 55 (2009) 467–475 471

Fig. 4. Determination of forskolin-stimulated cAMP levels by Catch PointTM kit.


Stimulation of membrane bound AC with forskolin resulted in cAMP production and
accumulation in a concentration-dependent manner. The inhibitory effect of the
agonist UK14,304 depends on forskolin concentration. 1 mM forskolin seemed to be
optimal, at higher concentrations the inhibitory efficacy of UK14,304 gradually
decreased. Average raw fluorescence intensity values of unstimulated control
samples were 4013  53 (buffer) and 4182  81 (25 mM UK14,304), respectively.
Each column reperesents the mean value  S.E.M from a representative experiment
performed in triplicate.

between the maximal inhibitions of the tested antagonists were


not significant. The rank order of potencies and relative efficacies
of various a2-AR ligands for both cell lines are summarized in
Table 1.
UK14,304 was also evaluated for its ability to inhibit forskolin-
induced cAMP elevation in CHO-K1 cells expressing recombinant
a2C-ARs alone. Stimulation of membrane bound adenylyl-cyclase
with forskolin resulted in cAMP production and accumulation in a
dose-dependent manner with an EC50 value of 343 nM. The
inhibitory effect of UK14,304 was dependent on forskolin
Fig. 3. (a) Concentration-dependent inhibition of yohimbine and MK912 on the concentration. 1 mM forskolin seemed to be optimal for a near
cytoplasmic Ca2+ concentration rises evoked by 40 nM UK14,304 in Gaqi5/H7 complete inhibition of forskolin-induced cAMP elevation by full
cells. Antagonists were equilibrated with cells for 10 min before agonist agonists. At higher concentrations (>3 mM) the percentage of
stimulation. Preliminary experiments confirmed that both yohimbine and
inhibition gradually decreased (Fig. 4). UK14,304 concentration-
MK912 were devoid of agonist activity. Data points represent mean  S.E.M of 3
and 4 independent experiments, respectively. (b) Concentration-dependent dependently decreased the cAMP accumulation induced by
inhibition of yohimbine and MK912 on the cytoplasmic Ca2+ concentration rises forskolin (1 mM) with an IC50 value of 11.2  3.3 nM (Fig. 5). Pre-
evoked by 40 nM UK14,304 in Ga16/B6 cells. Antagonists were equilibrated treatment of agonist-stimulated a2C-C1 cells with 25 mM MK912
with cells for 10 min before agonist stimulation and preliminary experiments reversed the agonist effect, as shown in Fig. 6.
confirmed that both yohimbine and MK912 were devoid of agonist activity.
Data points represent mean  S.E.M of 3 and 4 independent experiments,
For comparative purposes we have developed a competitive
respectively. binding assay in membranes derived from a2C-AR expressing
CHO-K1 cells using [3H]UK14,304 as a radioligand. All compounds

Table 1
Pharmacological profile of a2C-ARs as determined in fluorometric [Ca2+]i and radioligand binding experiments.

Compounds [Ca2+]i [3H]UK14,304 binding

EC50/IC50 (nM) Emax (%) Ki (nM)

Gaqi5/H7 Ga16/B6 Gaqi5/H7 Ga16/B6 a2C-C1


UK14,304 1.7  0.1 5.1  0.3 100 100 1.7  0.4
Noradrenaline 2.1  0.3 15.3  2.7 97.4  0.4 95.8  2.5 2.0  0.2
Oxymetazoline 2.9  0.5 7.4  1.2 82.3  2.7 61.5  2.7 6.9  2.5
Clonidine 7.0  0.6 29  3.8 77.3  0.6 46.3  7.1 7.3  2.2
Moxonidine 41  4.7 321  34 90.3  3.9 80.1  2.2 63  5.4
MK912 0.73  0.05 0.20  0.04 0.21  0.04
Yohimbine 17.8  2.9 3.1  0.8 4.33  0.9

Agonist activity (EC50, nM) was deduced from concentration response curves of various compounds and percent efficacy (representing the relative maximum response, Emax
%) was normalized to 1 mM UK14,304. IC50s (nM) were determined against 40 nM UK14,304 (corresponding to an EC90 concentration). a2C-AR ligand affinities (Ki, nM) were
determined by inhibition studies using the radioligand [3H]-UK14,304. FlexStationII data were derived from cells co-expressing the a2C-AR and the chimeric Gaqi5 or
promiscuous Ga16 proteins, while binding data were derived from cells expressing only the a2C-ARs. Data in the table represent the mean  S.E.M. from three to seven
independent experiments.
472 D. Kurko et al. / Neurochemistry International 55 (2009) 467–475

Thus, our primary objective was the generation of an in vitro


model system in which pharmacology of a2C-ARs could be
investigated. Therefore, in addition to constructing appropriate
cell lines, we also developed an assay system by which functional
interaction of compounds with this receptor subtype could be
conveniently evaluated. Since a2C-ARs naturally use a Gai
signaling pathway, in this study we examined the feasibility of
using the chimeric as well as the promiscuous G-protein approach
to couple this adrenergic receptor subtype to a Ca2+signaling
readout facilitating ligand screening and applying fluorometry
beneficial for high-throughput evaluations.
Fluorometric Ca2+ assays have been used efficiently for
characterization of GPCRs signaling via Gaq proteins mobilizing
cytoplasmic [Ca2+]i. There are growing amount of evidence for their
potential use for investigating non-Gaq-coupled receptors as well
since it has been reported that replacement of five amino acids of
the C-terminal of the Gaq sequence with the corresponding Gai
Fig. 5. Inhibition of the forskolin-stimulated cAMP production by UK14,304. Agonist sequence can convert the signaling pathway of Gi/o-coupled
binding to a2C-ARs prior to forskolin-induced AC activation inhibits cAMP receptors towards the PLC-b pathway. A significant number of
production in a dose-dependent manner with an IC50 value of 11.2 nM. Each Gi-linked GPCRs have been successfully coupled to such chimeric
point represents the mean  S.E.M. of 3 separate experiments.
G-proteins, e.g. A1 adenosine, D2 dopamine, a2-adrenergic
receptors (Conklin et al., 1993), neuropeptide Y receptors (hY1,
hY2, hY4; Dauzenberg et al., 2005), mGluR2, mGluR4 (Gomeza
et al., 1996; Kowal et al., 2003; Downey et al., 2005), H3 histamine
(Uvegas et al., 2002), mel-1a and mel-1b melatonin (Yokoyama
et al., 2003), D3, D4 dopamine receptors (Moreland et al., 2004).
However, there are some receptors (like somatostatin sst1 and
cannabinoid CB1 receptors) where such an exchange failed to
produce functional systems (Conklin et al., 1996). Alternatively,
receptors can be also co-expressed with certain promiscuous G-
proteins (i.e. Ga15, Ga16) that are capable of interacting with a
variety of Gs-, Gi,o-, and Gq-coupled receptors. Calcium release has
been detected upon activation of b2-adrenergic, muscarinic M2,
dopamine D1, vasopressin V2, adenosine A2A, 5-HT1A, fMLP, m-
opioid, thromboxane A2 receptors when co-expressed with
promiscuous G-proteins (Offermanns and Simon, 1995). However,
the promiscuous G-proteins can not be considered as truly
universal adaptors, as they either poorly couple to or fail to
activate the Ca2+ pathway in the case of some Gi-selective GPCRs
(e.g. dopamine D3, somatostatin sst1, melatonin MT1c, chemokines
CCR1 and CCR2) (Milligan et al., 1996; Marchese et al., 1999; Mody
et al., 2000; Kostenis, 2001; Liu et al., 2003). Several Gi/o-coupled
Fig. 6. Forskolin-induced cAMP elevations (square) and reversal of UK14,304 receptors, such as the mGluR2 and mGluR4 (Gomeza, 1996), 5-
(25 mM, circle) reduced cAMP levels by the a2-AR antagonist MK912 (25 mM, HT1A receptors (Offermanns and Simon, 1995; Kowal et al., 2002)
triangle) in the original a2C-AR expressing CHO cells. Each point represents the
d-, k- and m-opioid (Coward et al., 1999) receptors, efficiently
mean  S.E.M. of 3 separate experiments.
activate chimeric and promiscuous G-proteins as well, but
depending on the receptors these recombinant systems do not
tested in Ca2+ assay bound to the receptor and concentration- always activate the different G-proteins equally.
dependently inhibited [3H]UK14,304 binding with the following In order to identify an effective receptor–G-protein combina-
rank order of affinity (Ki) MK912 (0.21  0.04 nM) > UK14,304 tion useful for screening potential a2C-AR ligands, we compared
(1.7  0.4 nM) > noradrenaline (2  0.2 nM) > yohimbine (4.3  the pharmacological profile of two cell lines stably expressing the
0.9 nM) > oxymetazoline (6.9  2.5 nM)  clonidine (7.3  2.2 receptor together with either a chimeric (Gaqi5) or a promiscuous
nM) > moxonidine (63  5.4 nM). The Hill slopes for adrenergic (Ga16) G-protein in the same cellular background. Although the
receptors ligands did not differ significantly from unity, indicating their transient co-transfection of a2A and a2B subtypes of adrenergic
competition with [3H]UK14,304 radioligand for the a2C-ARs (data not receptors with Ga15 promiscuous proteins (Pauwels and Colpaert,
shown). Data deduced from concentration–response curves for 2000a; Pauwels et al., 2000) and the transient co-transfection of
inhibition of [3H]UK14,304 binding are summarized in Table 1. a2C-ARs with chimeric Gaq/i1 proteins (Pauwels and Colpaert,
2000b) have been reported previously, our study is the first to
4. Discussion evaluate the use of the promiscuous Ga16 protein co-expression
strategy in the generation of stable cell lines co-expressing the a2C-
Recombinant receptors expressed in heterologous cellular ARs.
systems are extensively used as experimental models for studying For characterization of the expression of chimeric Gaqi5 or
receptor function and mechanism of action. Functional cell-based promiscuous Ga16 proteins in the cell lines we have obtained, RT-
assays are also widely employed to assess the pharmacological PCR and immunocytochemical analyses were applied. To test the
properties of the expressed receptor and their ligands (Sautel and functionality and to examine the pharmacological profile of
Milligan, 2000). recombinant a2C-ARs, radioligand binding and fluorometric
D. Kurko et al. / Neurochemistry International 55 (2009) 467–475 473

measuring techniques to determine cAMP-inhibition and calcium naline (100, EC50 = 3.98 nM) > oxymetazoline (81, EC50 = 4.68
levels were used. Although a2C-ARs were able to couple to both nM) > clonidine (75, EC50 = 13.18 nM). Other earlier investiga-
recombinant G-proteins, it seems that receptors most efficiently tions showed a similar rank order of agonists obtained by
couple to the chimeric Gaqi5 proteins. Transfection of Gaqi5 or measuring extracellular acidification, [35S]-GTPgS binding,
Ga16 proteins into CHO-K1 cells expressing a2C-ARs resulted in a Ca2+ and GTPase responses in CHO and HEK-293 cells stably
robust agonist-induced transient Ca2+ response displaying similar transfected with a wt a2C-AR (Pihlavisto and Scheinin, 1999;
kinetics in both cell lines, although the signal-to-background ratio Jasper et al., 1998; Umland et al., 2001; Kukkonen et al., 1998;
was approximately 2.5-fold higher in Gaqi5/H7 cells. Ca2+ Jansson et al., 1999). Some modest quantitative differences in
elevations were sensitive to the phospholipase C inhibitor U- agonist activities were, however, noted presumably due to
73122 suggesting that the Ca2+ response is due to PLC-b activation. differences in host cell lines used, coupling efficiency of native
Similar differences in preferential coupling have been seen in HEK- vs. chimeric G-proteins, and the measured endpoints.
293 cells co-expressing mGluR2 or mGluR4 and either chimeric MK912 and yohimbine, known to be potent and selective a2-AR
(Gaqi5, Gaqi9) or promiscuous G-proteins (Ga15, Ga16) (Gomeza antagonists with high affinity for the a2C-AR subtype (Schaak et al.,
et al., 1996). In the case of human d-, k- and m-opioid receptors 1997; Uhlén et al., 1994, 1997; Lalchandani et al., 2002; Mustafa
expressed in CHO-K1 cells, k- and m-opioid receptors signaled et al., 2005), inhibited UK14,304-mediated Ca2+ responses in a
most efficiently when they were co-transfected with Gaqi5. On the concentration-dependent manner. However, the antagonists were
contrary, in the case of the d-opioid receptor, its signaling was 4- to 6-fold less potent in the Gaqi5/H7 than the Ga16/B6 cell line,
more efficient with Ga16 co-transfection, although it worked well with higher IC50s in the concentration–inhibition curves. It is
with Gaqi5 too (Coward et al., 1999). possible that such discrepancy in antagonist potencies reflect the
Stable expression of the a2C-AR in CHO-K1 cells alone did not different coupling efficiencies of the a2C-ARs with the chimeric
result in cytoplasmic Ca2+ elevation in response up to 1 mM Gaqi5 or the promiscuous Ga16 proteins. The IC50 value of
UK14,304 stimulation. On the contrary, Kukkonen et al. (1998) yohimbine obtained from our Ca2+ assay was consistent with that
found that human a2-AR subtypes were coupled to elevation of obtained in a cAMP measurement assay using a cell-based
[Ca2+]i when heterologously expressed in CHO cells. However, their luciferase reporter gene assay on human a2C-ARs expressing
results suggest that this Ca2+ response is due to PLC-b activation by CHO cells (IC50 = 3.2 nM; Lalchandani et al., 2002). In contrast to
bg subunits from Gi proteins. According to literature data agonists yohimbine, in our Ca2+ assay the logistic slope of the concentra-
can differ in their potency to induce activation of the different G- tion–inhibition curve of MK912 was significantly greater than 1.
protein subunits. Results of Kukkonen et al. suggest that there is However, in contrast to the Hill slope, the logistic slope of
also a roughly150-fold decrease in the potency of UK14,304 when functional inhibition curves cannot be interpreted directly
Ca2+ elevation occurs through stimulation of PLC-b by bg subunits (Lazareno and Birdsall, 1993). Also, despite their prevalence, the
from Gi. As our aim was to examine whether the use of the chimeric physical events underlying steep concentration–effect curves in
or that of the promiscuous G-protein approach is more suitable to functional assays are poorly understood (Shoichet, 2006). It is not
model the primary signal transduction pathway of native Gi- clear what is behind the apparently different inhibitory patterns
coupled system, experiments with UK14,304 at concentrations produced by yohimbine and MK912 in our Ca2+ assay. Although a
higher than 1 mM have not been carried out. In addition, the recent study (Görnemann et al., 2007) found yohimbine and
marked differences between our and Kukkonen’s data could be due MK912 to be a competitive and noncompetitive antagonist,
to the different expression level of the a2C-ARs in the generated cell respectively, it has not been shown to our knowledge that the
lines, as Kukkonen et al. reported a 2040 fmol/mg protein inhibition curve of a noncompetitive antagonist should have
expression, while our saturation analysis revealed a 589.2 fmol/ higher logistic slope than a competitive one.
mg protein expression. Although agonist pharmacology did not seem altered in the
To examine whether the pharmacology of the receptor is fluorometric Ca2+ assay, it is important to verify subsequent
unaltered in the generated recombinant systems, a2C-coupled evaluation of compounds in assays reflecting endogenous G-
calcium signals were measured in response to a number of protein coupling of the recombinantly expressed receptors
reference agonists and antagonists. UK14,304 and noradrenaline (Kostenis et al., 2005). Our data indicate that the potency values
were full agonists, while oxymetazoline, clonidine and moxonidine of UK14,304 determined in the Ca2+ assay (1.9 nM and 5.1 nM in
behaved as partial agonist, in agreement with published data Gaqi5/H7 and Ga16/B6 cells, respectively) were relatively close to
(Munk et al., 1994; MacDonald et al., 1997; Parsley et al., 1999; that obtained when forskolin-stimulated cAMP accumulation was
Umland et al., 2001; Millan, 2002; Szabo, 2002; Stone et al., 2003). measured (11.2 nM). MK912 pre-treatment reversed the agonist
The efficacies of all partial agonists tended to be higher in the case effect. These data are in agreement with those acquired previously
of Gaqi5/H7 cells compared with Ga16/B6 cells. The potencies of in cAMP assays on native receptors or recombinant a2C-ARs
the agonists investigated were approximately in the same range in coupled to endogenous G-proteins. UK14,304 concentration-
both cell lines, with all compounds displaying a moderate decrease dependently inhibited forskolin-stimulated cAMP accumulation
in potency when tested in Ga16/B6 cells. The concentration– in human neuroblastoma SH-SY5Y cells (EC50 = 7.83 nM, Parsley
response curves were rightward shifted with 3- to 8-fold et al., 1999) and also in LM(tk-) and CHO-K1 cells stably transfected
differences in EC50 values. Except that oxymetazoline was some- with the cloned human a2C-AR (EC50 = 8.02 nM, Jeon et al., 1995;
what more potent than noradrenaline in this model, there were no EC50 = 2.1 nM, Umland et al., 2001).
differences between the rank orders of potencies. This may be a In addition to observing good agreement between the func-
result of either a better coupling of these receptors to this tional activities, our Ca2+ measurement data also correspond well
particular chimeric G-protein or of a higher level of expression of to those generated in radioligand binding studies using mem-
this a subunit, and subsequent a more efficient coupling to PLC-b. branes prepared from the a2C-C1 cell line. Compounds used in the
Similar a2C-AR subtype-dependent pharmacological profile Ca2+ assay were profiled using [3H]UK14,304 radioligand binding.
was reported for these agonists in a study using similar With the exception of oxymetazoline in Ga16/B6 cells, concentra-
fluorometric techniques (Pauwels and Colpaert, 2000b). Transient tion–response curves for inhibition of [3H]UK14,304 binding
co-transfection of the wild type (wt) a2C-AR with chimeric Gaq/i1 yielded essentially identical rank order of compound affinities
protein in CHO-K1 cells revealed the following rank order of when compared with the Ca2+ assay. In the case of the Gaqi5/H7
efficacies and potencies: UK14,304 (102, EC50 = 1.48 nM) = adre- cell line, EC50/IC50 values from the fluorometric Ca2+ measure-
474 D. Kurko et al. / Neurochemistry International 55 (2009) 467–475

ments and Ki values from binding tests were almost the same, only Crassous, P.A., Denis, C., Paris, H., Sénard, J.M., 2007. Interest of alpha2-adrenergic
agonists and antagonists in clinical practice: background, facts and perspec-
yohimbine had a 4-fold higher value in the functional assay. In the tives. Curr. Top. Med. Chem. 7 (2), 187–194.
case of Ga16/B6 cells up to 8-fold differences were observed Dauzenberg, F.M., Higelin, J., Pflieger, P., Neidhart, W., Guba, W., 2005. Estabilish-
between radioligand binding and fluorometric Ca2+ assays. More- ment of robust functional assays for the characterization of neuropeptide Y
(NPY) receptors: identification of 3-(5-benzoyl-thiazol-2-yamino)-benzonitrile
over, the radioligand binding data we obtained were very similar to as selective NPY type 5 receptor antagonist. Neuropharmacology 48, 1043–
those reported by other investigators (Piletz et al., 1996; Umland 1055.
et al., 2001; Uhlén et al., 1997; Lalchandani et al., 2002; Mustafa Docherty, J.R., 1998. Subtypes of functional a1- and a2-adrenoceptors. Eur. J.
Pharmacol. 361, 1–15.
et al., 2005). Binding affinities of noradrenaline (Ki = 1.7 nM), Downey, P.M., Lozza, G., Petrò, R., Diodato, E., Foglia, C., Bottazzoli, F., Brusa, R.,
UK14,304 (Ki = 2.2 nM), oxymetazoline (Ki = 7.4 nM) for displacing Asquini, T., Reggiani, A., Grilli, M., 2005. Ecdysone-based system for controlled
[3H]UK14,304 on human a2C-ARs expressed in CHO cells have been inducible expression of metabotropic glutamate receptor subtypes 2, 5, and 8. J.
Biomol. Screen. 10 (8), 841–848.
reported previously (Umland et al., 2001). Radioligand binding
Eason, M.G., Kurose, H., Holt, B.D., Raymond, J.R., Liggett, S.B., 1992. Simultaneous
analyses of antagonists revealed a Ki value of 0.063 nM for MK912 coupling of a2-adrenergic receptors to two G-proteins with opposing effects.
(Lalchandani et al., 2002) and Ki values of 0.68 nM (Lalchandani Subtype-selective coupling of a2C10, a2C4, and a2C2 adrenergic receptors to Gi
et al., 2002) and 0.88 nM (Mustafa et al., 2005) for yohimbine by and Gs. J. Biol. Chem. 267, 15795–15801.
Eason, M.G., Jacinto, M.T., Liggett, S.B., 1994. Contribution of ligand structure to
replacing [3H]rauwolscine. activation of a2-adrenergic receptor subtype coupling to Gs. Mol. Pharmacol.
In summary, in this study we demonstrated that the human 45, 696–702.
a2C-AR can be coupled to calcium signaling using chimeric Gaqi5 as Eglen, R.M., 2005. Functional G protein-coupled receptor assays for primary and
secondary screening. Comb. Chem. High Throughput Screen. 8, 311–318.
well as promiscuous Ga16 proteins. According to the radioligand Fresco, P., Oliveira, J.M., Kunc, F., Soares, A.S., Rocha-Pereira, C., Gonçalves, J., Diniz,
binding affinities of a2-AR agonist and antagonists and their C., 2007. A2A adenosine-receptor-mediated facilitation of noradrenaline release
modulatory potential on forskolin-evoked cAMP accumulation, in rat tail artery involves protein kinase C activation and bg subunits formed
after a2-adrenoceptor activation. Neurochem. Int. 51 (1), 47–56.
pharmacological properties of a2-AR ligands observed in our co- Gabriel, D., Vernier, M., Pfeifer, M.J., Dasen, B., Tenaillon, L., Bouhelal, R., 2003. High
expression systems proved to be similar to those reported in native throughput screening technologies for direct cyclic AMP measurement. Assay
Gai-coupled systems. However, our findings indicate that these Drug Dev. Technol. 1 (2), 291–303.
Gomeza, J., Mary, S., Brabet, I., Parmentier, M., Restituito, S., Bockaert, J., Pin, J., 1996.
recombinant systems activate the chimeric or promiscuous G- Coupling of metabotropic glutamate receptors 2 and 4 to Ga15, Ga16, and
proteins differently, as co-expression of Gaqi5 protein with a2C-AR chimeric Gaq/i proteins: characterization of new antagonists. Mol. Pharmacol.
provides a more sensitive cellular system for functional assays. The 50, 923–930.
Görnemann, T., von Wenckstern, H., Kleuser, B., Villalón, C.M., Centurión, D.,
established Ca2+ assay on dual recombinant protein expressing cell
Jähnichen, S., Pertz, H.H., 2007. Characterization of the postjunctional alpha
lines may substitute or complement the existing cAMP assay 2C-adrenoceptor mediating vasoconstriction to UK14304 in porcine pulmonary
formats and can enhance the capacity for characterization of a2C- veins. Br. J. Pharmacol. 151 (2), 186–194.
AR ligands providing a robust and reliable functional assay more Gyires, K., Zádori, Z.S., Shujaa, N., Minorics, R., Falkay, G., Mátyus, P., 2007. Analysis
of the role of central and peripheral a2-adrenoceptor subtypes in gastric
appropriate for high-throughput screening examinations. mucosal defense in the rat. Neurochem. Int. 51 (5), 289–296.
Hieble, J.P., Bondinell, W.E., Ruffolo Jr., R.R., 1995. Alpha- and beta-adrenoceptors:
from the gene to the clinic. 1. Molecular biology and adrenoceptor subclassi-
Acknowledgments fication. J. Med. Chem. 38 (18), 3415–3444.
Jansson, C.C., Pohjanoksa, K., Lang, J., Wurster, S., Savola, J.M., Scheinin, M., 1999. a2-
We thank for the excellent technical assistance of Mrs. E. Széll, Adrenoceptor agonists stimulate high-affinity GTPase activity in a receptor
subtype-selective manner. Eur. J. Pharmacol. 374, 137–146.
Mrs. E. Borók, Mrs. P. Unghy and Mrs. A. Garai. We are grateful to
Jasper, J.R., Lesnick, J.D., Chang, L.K., Yamanishi, S.S., Chang, T.K., Hsu, S.A., Daunt,
Dr. I. Tarnawa for invaluable help in discussions and for critically D.A., Bonhaus, D.W., Eglen, R.M., 1998. Ligand efficacy and potency at recom-
reading the manuscript. binant a2-adrenergic receptors: agonist-mediated [35S]GTPgS binding. Bio-
chem. Pharmacol. 55 (7), 1035–1043.
Jeon, Y.T., Luoa, C., Forrayb, C., Vaysseb, P.J.J., Branchekb, T.A., Gluchowskia, C., 1995.
References Pharmacological evaluation of UK-14,304 analogs at cloned human a adrener-
gic receptors. Bioorg. Med. Chem. Lett. 5 (19), 2255–2258.
Abdulla, F.A., Smith, P.A., 1997. Ectopic alpha2-adrenoceptors couple to N-type Ca2+ Ji, T.H., Grossmann, M., Ji, I., 1998. G protein-coupled receptors. I. Diversity of
channels in axotomized rat sensory neurons. J. Neurosci. 17, 1633–1641. receptor–ligand interactions. J. Biol. Chem. 273 (28), 17299–17302.
Arnsten, A.F., 1998. The biology of being frazzled. Science 280, 1711–1712. Kable, J.W., Murrin, L.C., Bylund, D.B., 2000. In vivo gene modification elucidates
Balogh, B., Jójárt, B., Wágner, Z., Kovác, s, P., Máté, G., Gyires, K., Zádori, Z., Falkay, G., subtype-specific functions of alpha(2)-adrenergic receptors. J. Pharmacol. Exp.
Márki, A., Viskolcz, B., Mátyus, P., 2007. 3D QSAR models for a2a-adrenoceptor Ther. 293 (1), 1–7.
agonists. Neurochem. Int. 51 (5), 268–276. Kamibayashi, T., Maze, M., 2000. Clinical uses of alpha2-adrenergic agonists.
Bylund, D.B., 1992. Subtypes of a1- and a2-adrenergic receptors. Fed. Am. Soc. Exp. Anesthesiology 93 (5), 1345–1349.
Biol. J. 6 (3), 832–839. Kassack, M.U., Hofgen, B., Lehmann, J., Eckstein, N., Quillan, J.M., Sadee, W., 2002.
Bylund, D.B., 2005. Alpha-2 adrenoceptor subtypes: are more better? Br. J. Phar- Functional screening of G protein-coupled receptors by measuring intracellular
macol. 144, 159–160. calcium with a fluorescence microplate reader. J. Biomol. Screen. 7, 233–246.
Calzada, B.C., Artinano, A.A., 2001. Alpha-adrenoceptor subtypes. Pharmacol. Res. Knaus, A.E., Muthig, V., Schickinger, S., Moura, E., Beetz, N., Gilsbach, R., Hein, L.,
44 (3), 195–208. 2007. a2-Adrenoceptor subtypes—unexpected functions for receptors and
Cerione, R.A., 1991. Reconstitution of receptor/GTP-binding protein interactions. ligands derived from gene-targeted mouse models. Neurochem. Int. 51 (5),
Biochim. Biophys. Acta 1071 (4), 473–501. 277–281.
Chambers, C., Smith, F., Williams, C., Marcos, S., Liu, Z.H., Hayter, P., Ciaramella, G., Kostenis, E., 2001. Is Ga16 the optimal tool for fishing ligands of orphan G protein-
Keighley, W., Gribbon, P., Sewing, A., 2003. Measuring intracellular calcium fluxes coupled receptors? Trends Pharmacol. Sci. 22 (11), 560–564.
in high throughput mode. Comb. Chem. High Throughput Screen. 6, 355–362. Kostenis, E., Degtyarev, M.Y., Conklin, B.R., Wess, J., 1997. The N-terminal extension
Cheng, Y., Prusoff, W.H., 1973. Relationship between the inhibition constant (Ki) and of Gaq is critical for constraining the selectivity of receptor coupling. J. Biol.
the concentration of inhibitor which causes 50 percent inhibition (IC50) of an Chem. 272, 19107–19110.
enzymatic reaction. Biochem. Pharmacol. 22, 3099–3108. Kostenis, E., Waelbroeck, M., Milligan, G., 2005. Techniques: promiscuous Ga
Conklin, B.R., Chabre, O., Wong, Y.H., Federman, A.D., Bourne, H.R., 1992. Recombi- proteins in basic research and drug discovery. Trends Pharmacol. Sci. 26
nant Gqa mutational activation and coupling to receptors and phospholipase. J. (11), 595–602.
Biol. Chem. 267, 31–34. Kowal, D., Nawoschik, S., Ochalski, R., Dunlop, J., 2003. Functional calcium coupling
Conklin, B.R., Farfel, Z., Lustig, K.D., Julius, D., Bourne, H.R., 1993. Substitution of with the human metabotropic glutamate receptor subtypes 2 and 4 by stable
three amino acids switches receptor specificity of Gqa to that of Gia. Nature co-expression with a calcium pathway facilitating G-protein chimera in Chinese
363, 274–276. hamster ovary cells. Biochem. Pharmacol. 66 (5), 785–790.
Conklin, B.R., Herzmark, P., Ishida, S., Voyno-Yasenetskaya, T.A., Sun, Y., Farfel, Z., Kowal, D., Zhang, J., Nawoschik, S., Ochalski, R., Vlattas, A., Shan, Q., Schechter, L.,
Bourne, H.R., 1996. Carboxyl-terminal mutations of Gqa and Gsa that alter the Dunlop, J., 2002. The C-terminus of Gi family G-proteins as a determinant of 5-
fidelity of receptor activation. Mol. Pharmacol. 50, 885–890. HT(1A) receptor coupling. Biochem. Biophys. Res. Commun. 294 (3), 655–659.
Coward, P., Chan, S.D.C., Wada, H.G., Humphries, G.M., Conklin, B.R., 1999. Chimeric Kukkonen, J.P., Renvaktar, A., Shariatmadari, R., Akerman, K.E., 1998. Ligand- and
G proteins allow a high-throughput signaling assay of Gi-coupled receptors. subtype-selective coupling of human alpha-2 adrenoceptors to Ca++ elevation in
Anal. Biochem. 270, 242–248. Chinese hamster ovary cells. J. Pharmacol. Exp. Ther. 287 (2), 667–671.
D. Kurko et al. / Neurochemistry International 55 (2009) 467–475 475

Lakhlani, P.P., MacMillan, L.B., Guo, T.Z., McCool, B.A., Lovinger, D.M., Maze, M., Perez, D., 2005. From plants to man: the GPCR ‘‘Tree of life’’. Mol. Pharmacol. 67,
Limbird, L.E., 1997. Substitution of a mutant alpha (2a)-adrenergic receptor via 1383–1384.
hit and run gene targeting reveals the role of this subtype in sedative, analgesic, Philipp, M., Brede, M., Hein, L., 2002. Physiological significance of a2-adrenergic
and anesthetic sparing responses in vivo. Proc. Natl. Acad. Sci. U.S.A. 94, 9950– receptor subtype diversity: one receptor is not enough. Am. J. Physiol.: Regul.
9955. Integr. Comp. Physiol. 283, 287–295.
Lalchandani, S.G., Lei, L., Zheng, W., Suni, M.M., Moore, B.M., Liggett, S.B., Miller, D.D., Pihlavisto, M., Scheinin, M., 1999. Functional assessment of recombinant human a2-
Feller, D.R., 2002. Yohimbine dimers exhibiting selectivity for the human alpha adrenoceptor subtypes with cytosensor microphysiometry. Eur. J. Pharmacol.
2C-adrenoceptor subtype. J. Pharmacol. Exp. Ther. 303 (3), 979–984. 385 (2–3), 247–253.
Langer, S.Z., 2008. Presynaptic autoreceptors regulating transmitter release. Neu- Piletz, J.E., Zhu, H., Cikkala, D.N., 1996. Comparison of ligand binding affinities at
rochem. Int. 52 (1–2), 26–30. human I1-imidazoline binding sites and the high affinity state of a2-adreno-
Lazareno, S., Birdsall, N.J.M., 1993. Estimation of competitive antagonist affinity ceptor subtypes. J. Pharmacol. Exp. Ther. 279, 694–702.
from functional inhibition curves using the Gaddum, Schild and Cheng–Prusoff Pohjanoksa, K., Jansson, C.C., Luomala, K., Marjamaki, A., Savola, J.M., Scheinin, M.,
equations. Br. J. Pharmacol. 109, 1110–1119. 1997. a2-Adrenoceptor regulation of adenylyl cyclase in CHO cells: dependence
Limbird, L.E., 1988. Receptors linked to inhibition of adenylate cyclase: additional on receptor density, receptor subtype and current activity of adenylyl cyclase.
signaling mechanisms. Fed. Am. Soc. Exp. Biol. J. 2, 2686–2695. Eur. J. Pharmacol. 335, 53–63.
Link, R., Daunt, D., Barsh, G., Chruscinski, A., Kobilka, B., 1992. Cloning of two mouse Reynen, P.H., Martin, G.R., Eglen, R.M., MacLennan, S.J., 2000. Characterization of
genes encoding alpha 2-adrenergic receptor subtypes and identification of a human recombinant a2A-adrenoceptors expressed in Chinese hamster lung
single amino acid in the mouse alpha2-C10 homolog responsible for an inter- cells using intracellular Ca2+ changes: evidence for cross-talk between recom-
species variation in antagonist binding. Mol. Pharmacol. 42 (1), 16–27. binant a2A- and native a1-adrenoceptors. Br. J. Pharmacol. 129 (7), 1339–1346.
Liu, A.M., Ho, M.K., Wong, C.S., Chan, J.H., Pau, A.H., Wong, Y.H., 2003. Galpha(16/z) Ruffolo Jr., R.R., Bondinell, W., Hieble, J.P., 1995. Alpha- and beta-adrenoceptors:
chimeras efficiently link a wide range of G protein-coupled receptors to calcium from the gene to the clinic. 2. Structure–activity relationships and therapeutic
mobilization. J. Biomol. Screen. 8, 39–49. applications. J. Med. Chem. 38 (19), 3681–3716.
Liu, J., Conklin, B.R., Blin, N., Yun, J., Wesss, J., 1995. Identification of a receptor/G- Ruffolo Jr., R.R., Hieble, J.P., 1994. a-Adrenoceptors. Pharmacol. Ther. 61, 1–64.
protein contact site critical for signaling specificity and G-protein activation. Ruffolo Jr., R.R., Nichols, A.J., Stadel, J.M., Hieble, J.P., 1993. Pharmacologic and
Proc. Natl. Acad. Sci. U.S.A. 92, 11642–11646. therapeutic applications of alpha 2-adrenoceptor subtypes. Annu. Rev. Phar-
MacDonald, E., Kobilka, B.K., Scheinin, M., 1997. Gene targeting-homing in on the macol. Toxicol. 33, 243–279.
a2-adrenoceptor-subtype function. Trends Pharmacol. Sci. 18, 211–219. Sanders, R.D., Maze, M., 2007. a2-Adrenoceptor agonists. Curr. Opin. Investig. Drugs
Marchese, A., George, S.R., Kolakowski Jr., L.F., Lynch, K.R., O’Dowd, B.F., 1999. Novel 8 (1), 25–33.
GPCRs and their endogenous ligands: expanding the boundaries of physiology Saunders, C., Limbird, L.E., 1999. Localization and trafficking of a2-adrenergic
and pharmacology. Trends Pharmacol. Sci. 20, 370–375. receptor subtypes in cells and tissues. Pharmacol. Ther. 84, 193–205.
Maze, M., Fujinaga, M., 2000. Alpha(2) adrenoceptors in pain modulation—which Sautel, M., Milligan, G., 2000. Molecular manipulation of G-protein-coupled recep-
subtype should be targeted to produce analgesia? Anesthesiology 92, tors: a new avenue into drug discovery. Curr. Med. Chem. 7 (9), 889–896.
934–936. Schaak, S., Cayla, C., Blaise, R., Quinchon, F., Paris, H., 1997. HepG2 and SK-N-MC:
Millan, M.J., 2002. Descending control of pain. Prog. Neurobiol. 66 (6), 355–474. two human models to study alpha-2 adrenergic receptors of the alpha-2C
Milligan, G., 2003. Principles: extending the utility of [35S]GTPgS binding assays. subtype. J. Pharmacol. Exp. Ther. 281 (2), 983–991.
Trends Pharmacol. Sci. 24, 87–90. Scholz, J., Tonner, P.H., 2000. a2-Adrenoceptor agonists in anaesthesia: a new
Milligan, G., Marshall, F., Rees, S., 1996. G16 as a universal G protein adapter: paradigm. Curr. Opin. Anaesthesiol. 13, 437–442.
implications for agonist screening strategies. Trends Pharmacol. Sci. 17 (7), Serle, J.B., Lustgarten, J.S., Podos, S.M., 1991. A clinical trial of metipranodol, a
235–237. noncardioselective beta-adrenergic antagonist, in ocular hypertension. Am. J.
Milligan, G., Rees, S., 1999. Chimaeric Ga proteins: their potential use in drug Ophthalmol. 112, 302–307.
discovery. Trends Pharmacol. Sci. 20, 118–124. Shoichet, B.K., 2006. Interpreting steep dose–response curves in early inhibitor
Mody, S.M., Ho, M.K., Joshi, S.A., Wong, Y.H., 2000. Incorporation of Gaz specific discovery. J. Med. Chem. 49, 7274–7277.
sequence at the carboxyl terminus increases the promiscuity of Ga16 toward Gi- Stone, L.S., Fairbanks, C.A., Wilcox, G.L., 2003. Moxonidine, a mixed a2-adrenergic
coupled receptors. Mol. Pharmacol. 57, 13–23. and imidazoline receptor agonist, identifies a novel adrenergic target for spinal
Monteith, G.R., Bird, G.St.J., 2005. Techniques: high-throughput measurement of analgesia. Ann. N.Y. Acad. Sci. 1009, 378–385.
intracellular Ca2+—back to basics. Trends Pharmacol. Sci. 26 (4), 218–223. Szabo, B., 2002. Imidazoline antihypertensive drugs: a critical review on their
Moreland, R.B., Nakane, M., Donnelly-Roberts, D.L., Miller, L.N., Chang, R., Uchic, mechanism of action. Pharmacol. Ther. 93, 1–35.
M.E., Terranova, M.A., Gubbins, E.J., Helfrich, R.J., Namovic, M.T., El-Kouhen, O.F., Thomsen, W., Frazer, J., Unett, D., 2005. Functional assays for screening GPCR
Masters, J.N., Brioni, J.D., 2004. Comparative pharmacology of human dopamine targets. Curr. Opin. Biotechnol. 16, 655–665.
D2-like receptor stable cell lines coupled to calcium flux through Gaqo5. Thomsen, W.J., Gatlin, J., Unett, D.J., Behan, D.P., 2004. Developing functional GPCR
Biochem. Pharmacol. 68, 761–772. screens. http://www.currentdrugdiscovery.com/pdf/2004/518083.pdf.
Munk, S.A., Gluchowski, C., Dolby, L., Wong, H., Burke, J., Kharlamb, A., Manlapaz, C., Uhlén, S., Porter, A.C., Neubig, R.R., 1994. The novel alpha-2 adrenergic radioligand
Padillo, E., Rodgers, D., Ohta, B., Wheeler, L., Garst, M., 1994. Analogues of UK [3H]-MK912 is alpha-2C selective among human alpha-2A, alpha-2B and alpha-
14,304 as R2-adrenoceptor agonists. Twist and agent polarity as design ele- 2C adrenoceptors. J. Pharmacol. Exp. Ther. 271 (3), 1558–1565.
ments. Bioorg. Med. Chem. Lett. 4, 459–462. Uhlén, S., Lindblom, J., Johnson, A., Wikberg, J.E., 1997. Autoradiographic studies of
Mustafa, S.M., Bavadekar, S.A., Ma, G., Moore, B.M., Fellerb, D.R., Millera, D.D., 2005. central alpha 2A- and alpha 2C-adrenoceptors in the rat using [3H]MK912 and
Synthesis and biological studies of yohimbine derivatives on human a2C- subtype-selective drugs. Brain Res. 770 (1–2), 261–266.
adrenergic receptors. Bioorg. Med. Chem. Lett. 15, 2758–2760. Umland, S.P., Wan, Y., Shah, H., Billah, M., Egan, R.W., Hey, J.A., 2001. Receptor
New, D.C., Wong, Y.H., 2005. Chimeric and promiscuous G proteins in drug dis- reserve analysis of the human a2C-adrenoceptors using [35S]GTPgS and cAMP
covery and the deorphanization of GPCRs. Drug Des. Rev. Online 2, 66–79. functional assays. Eur. J. Pharmacol. 411, 211–221.
Offermanns, S., Simon, I.M., 1995. Ga15 and Ga16 couple a wide variety of receptors Uvegas, A.J., Kowal, D., Zhang, Y., Spangler, T.B., Dunlop, J., Semus, S., Jones, P.G.,
to phospholipase C. J. Biol. Chem. 270 (25), 15175–15180. 2002. The role of transmembrane helix 5 in agonist binding to the human H3
Parsley, S., Gazi, L., Bobirnac, I., Loetscher, E., Schoeffter, P., 1999. Functional a2C- receptor. J. Pharmacol. Exp. Ther. 301, 451–458.
adrenoceptors in human neuroblastoma SH-SY5Y cells. Eur. J. Pharmacol. 372, Williams, C., 2004. cAMP detection methods in HTS: selecting the best from the rest.
109–115. Nat. Rev. Drug Discov. 3, 125–135.
Pauwels, P.J., Colpaert, F.C., 2000a. Disparate ligand-mediated Ca2+ responses by Wong, W.C., Wang, D., Forray, C., Vaysse, P.J.-J., Branchek, T.A., Gluchowski, C.A.,
wild-type, mutant Ser200Ala and Ser204Ala a2A-adrenoceptor: Ga15 fusion 1994. Convenient synthesis of 2-amino-2-oxazolines and their pharmacological
proteins: evidence for multiple ligand-activation binding sites. Br. J. Pharmacol. evaluation at cloned human a-adrenergic receptors. Bioorg. Med. Chem. Lett. 4,
130, 1505–1512. 2317–2322.
Pauwels, P.J., Colpaert, F.C., 2000b. Partial to complete antagonism by puative Wong, W.C., Sun, W., Cui, W., Chen, Y., Forray, C., Vaysse, P.J., Branchek, T.A.,
antagonists at wild-type a2C-adrenoceptors based on kinetics analyses of Gluchowski, C., 2000. 2-Amino-2-oxazolines as subtype selective a2-adreno-
agonist: antagonist interactions. Br. J. Pharmacol. 131, 1385–1390. ceptor agonists. J. Med. Chem. 43, 1699–1704.
Pauwels, P.J., Tardif, S., Wurch, T., Colpaert, F.C., 2000. Facilitation of constitutive Wong, Y.H., 1994. Gi assays in transfected cells. Methods Enzymol. 238, 81–94.
a2A-adrenoceptor activity by both single amino acid mutation (Thr373Lys) and Yokoyama, T., Katob, N., Yamada, N., 2003. Development of a high-throughput
Gao protein co-expression: evidence for inverse agonism. J. Pharmacol. Exp. bioassay to screen melatonin receptor agonists using human melatonin recep-
Ther. 292, 654–663. tor expressing CHO cells. Neurosci. Lett. 344, 45–48.

You might also like