ArticlePDF Available

Locking the Dimeric GABAB G-Protein-Coupled Receptor in Its Active State

Authors:

Abstract and Figures

G-protein-coupled receptors (GPCRs) play a major role in cell-cell communication in the CNS. These proteins oscillate between various inactive and active conformations, the latter being stabilized by agonists. Although mutations can lead to constitutive activity, most of these destabilize inactive conformations, and none lock the receptor in an active state. Moreover, GPCRs are known to form dimers, but the role of each protomer in the activation process remains unclear. Here, we show that the heterodimeric GPCR for the main inhibitory neurotransmitter, the GABA(B) receptor, can be locked in its active state by introducing two cysteines expected to form a disulphide bridge to maintain the binding domain of the GABA(B1) subunit in a closed form. This constitutively active receptor cannot be inhibited by antagonists, but its normal functioning, activation by agonists, and inhibition by antagonists can be restored after reduction with dithiothreitol. These data show that the closed state of the binding domain of GABA(B1) is sufficient to turn ON this heterodimeric receptor and illustrate for the first time that a GPCR can be locked in an active conformation.
Content may be subject to copyright.
Cellular/Molecular
Locking the Dimeric GABA
B
G-Protein-Coupled Receptor in
Its Active State
Julie Kniazeff,
1
Pierre-Philippe Saintot,
1
Cyril Goudet,
1
Jianfeng Liu,
1
Annie Charnet,
2
Gilles Guillon,
2
and
Jean-Philippe Pin
1
1
Laboratory for Functional Genomic, Department of Molecular Pharmacology, Centre National de la Recherche Scientifique Unite´ Propre de Recherche-
2580, and
2
Institut National de la Sante´ et de la Recherche Me´dicale U469, 34094 Montpellier Cedex 5, France
G-protein-coupled receptors (GPCRs) play a major role in cell– cell communication in the CNS. These proteins oscillate between various
inactive and active conformations, the latter being stabilized by agonists. Although mutations can lead to constitutive activity, most of
these destabilize inactive conformations, and none lock the receptor in an active state. Moreover, GPCRs are known to form dimers, but
the role of each protomer in the activation process remains unclear. Here, we show that the heterodimeric GPCR for the main inhibitory
neurotransmitter, the GABA
B
receptor, can be locked in its active state by introducing two cysteines expected to form a disulphide bridge
to maintain the binding domain of the GABA
B1
subunit in a closed form. This constitutively active receptor cannot be inhibited by
antagonists, but its normal functioning, activation by agonists, and inhibition by antagonists can be restored after reduction with
dithiothreitol. These data show that the closed state of the binding domain of GABA
B1
is sufficient to turn ON this heterodimeric receptor
and illustrate for the first time that a GPCR can be locked in an active conformation.
Key words: receptor; baclofen; GABA; metabotropic glutamate receptor; glutamate; activation mechanism
Introduction
G-protein-coupled receptors (GPCRs) play a major role in cell–
cell communication, especially in the brain, where they are in-
volved in the tuning of fast synaptic transmission. Several classes
of GPCRs can be defined based on sequence similarity (Kola-
kowski, 1994; Bockaert and Pin, 1999; Fredriksson et al., 2003).
GPCRs for the two main neurotransmitters, GABA and gluta-
mate [the GABA
B
and metabotropic glutamate receptors
(mGluRs)] are part of the class III GPCRs, together with those
activated by Ca
2
, sweet molecules, and some pheromones (Pin
et al., 2003). These receptors form dimers, but the GABA
B
recep-
tor is an obligatory heterodimer constituted of the homologous
GABA
B1
and GABA
B2
subunits (Jones et al., 1998; Kaupmann et
al., 1998; White et al., 1998). GABA
B1
is responsible for GABA
recognition (Galvez et al., 2000a; Kniazeff et al., 2002), whereas
GABA
B2
is necessary for the correct trafficking of GABA
B1
to the
cell surface (Margeta-Mitrovic et al., 2000; Pagano et al., 2001)
and is involved in G-protein activation (Galvez et al., 2001;
Margeta-Mitrovic et al., 2001b; Robbins et al., 2001; Duthey et al.,
2002).
The ligand-binding site of class III GPCRs is located in a large
extracellular domain (Takahashi et al., 1993) homologous to
some periplasmic-binding proteins (O’Hara et al., 1993) and to
the binding domain of other receptors such as ionotropic gluta-
mate, atrial natriuteric peptide (ANP), and some tyrosine kinase
receptors (Felder et al., 1999; Vicogne et al., 2003). X-ray struc-
tures have been solved for the extracellular domains of mGluR
type 1 (mGlu1) (Kunishima et al., 2000; Tsuchiya et al., 2002) and
ANP receptor (ANPR) types A and C (He et al., 2001; van den
Akker, 2001) produced as soluble proteins. These domains are
bilobate proteins often called Venus flytrap modules (VFTMs)
and share similar sequence homology with the extracellular do-
main of the GABA
B
subunits (Fig. 1a). The mGlu1 VFTM was
observed either in an open or closed conformation, whereas that
of the ANPR is only found in a closed conformation.
Both mGlu1 and ANP VFTMs homodimerize at the level of
their lobe I, leaving the two lobe II (connected to the membrane
domain) far apart in the absence of agonists (Fig. 1b,c). In the
presence of agonists, a change in conformation brings together
lobe II, possibly forcing the membrane domains to interact dif-
ferently (Fig. 1b,c). In the case of mGlu1, this results from: (1)
closure of at least one VFTM; and (2) a change in the relative
orientation of the VFTMs (Fig. 1b). In the case of the ANPR, this
results from the binding of ANP between the two VFTMs (Fig.
1c). Whether the dimer of GABA
B
VFTMs functions like that of
mGlu or ANPRs remains unknown.
Here, we show that the introduction of two cysteines expected
to lock the GABA
B1
VFTM in a closed state by a disulphide bridge
is sufficient to lock the entire receptor in an almost fully active
Received July 1, 2003; revised Nov. 10, 2003; accepted Nov. 10, 2003.
This work was supported by grants from the Centre National de la Recherche Scientifique (CNRS), Action Con-
certe´e Incitative “Mole´cules etCibles The´rapeutiques” from Institut Nationalde la Sante´ et de laRecherche Me´dicale,
and the French government (J.P.P.). J.L. and A.C. were supported by Groupement d’Intéreˆt Public Hoechst Marion
Roussel/Aventis and CisBio International, respectively. J.K. and C.G. were supported by CNRS and Fondation pour la
Recherche Me´dicale fellowships, respectively. We thank Drs. T. Galvez, G. Labesse, J. Neyton, M. L. Parmentier, L.
Pre´zeau, and P. Paoletti for constructive discussions and critical reading of this manuscript. We also thank Drs. W.
Froestl and K. Kaupmann (Novartis, Basel, Switzerland) for the supply of GABA
B
ligands and Dr. P. Rondard for the
construction of GB1 and GB2.
Correspondence should be addressed to Dr. Jean-Philippe Pin, Laboratory for Functional Genomic, Centre Na-
tional de la Recherche Scientifique Unite´ Propre de Recherche-2580, Department of Molecular Pharmacology, 141
rue de la Cardonille, F-34094 Montpellier Cedex 5, France. E-mail: jppin@ccipe.cnrs.fr.
DOI:10.1523/JNEUROSCI.3141-03.2004
Copyright © 2004 Society for Neuroscience 0270-6474/04/240370-08$15.00/0
370 The Journal of Neuroscience, January 14, 2004 24(2):370 –377
state. This reveals a similar activation mechanism for mGlu and
GABA
B
receptors and represents the first example of a GPCR that
has been locked in its active state. As such, this study brings much
information on the way the GPCRs for the main neurotransmit-
ters, glutamate and GABA, are activated.
Materials and Methods
Materials. GABA, baclofen, and CGP64213 were gifts from Drs.
W. Froestl and K. Kaupman (Novartis Pharma, Basel, Switzerland).
[
125
I]CGP64213 was purchase from Anawa (Zurich, Switzerland). FBS,
culture media, and other solutions used for cell culture were from
Invitrogen-Life Technologies, Inc. (Cergy Pontoise, France). [
3
H]Myo-
inositol (23.4 Ci/mol) was purchased from PerkinElmer Life Sciences
(Paris, France). All other reagents used were of molecular or analytical
grade, where appropriate.
Phylogenetic analysis. Sequence alignment of the VFTMs of crystallized
amide-binding protein AmiC (pdb:1pea), leucineisoleucinevaline-
binding protein (pdb: 2liv), natriuretic peptide receptors A and C (pdb:
1dp4 and 1jdn, respectively), and mGlu1 (pdb:1ewk) was deduced after
superimposition of their structures using SwissPdbViewer (version 3.7)
(Guex and Peitsch, 1997). Sequences of VFTMs of the rat NR2A subunit
of the NMDA receptor, GABA
B1
and GABA
B2
subunits (GB1 and GB2),
and tyrosine kinase receptor type 1 (RTK1) from Schistosoma mansoni
were aligned on the structural multiple alignment according to Paoletti et
al. (2000), Kniazeff et al. (2002), and Vicogne et al. (2003), respectively.
The phylogenetic tree was then constructed using the neighbor-joining
method (Saitou and Nei, 1987) with the command interface of the
Clustal W 1.60 program (Thompson et al., 1994) using the default pa-
rameters and not excluding gaps. Bootstrap values were calculated using
1000 trials with seeds of 111. The tree was drawn using TreeView (version
1.6.2) (Page, 1996).
Molecular modeling. Three-dimensional models of the open and closed
forms of GABA
B1
VFTM were built as described previously (Kniazeff et
al., 2002) using Modeler 6.0
(Sali and Blundell, 1993). In silico mu-
tagenesis and disulphide bridges modeling were performed using the
SwissPdbViewer program (version 3.7) (Guex and Peitsch, 1997) and
software default parameters. The figures were prepared using
SwissPdbViewer.
Plasmids and site-directed mutagenesis. The plasmids encoding the
wild-type GABA
B1a
and GABA
B2
subunits epitope tagged at their
N-terminal ends (pRK-GABA
B1a
-HA and pRK-GABA
B2
-cMyc), under
the control of a cytomegalovirus promoter, were described previously
(Galvez et al., 2001; Pagano et al., 2001).
Mutant subunits, carrying single or multiple mutations, were obtained
using the Quick-Change strategy (Stratagene, La Jolla, CA). Briefly, for
each mutagenesis, two complementary 27-mers primers (Eurogentec,
Bruxelles, Belgium) were designed to contain the desired mutation. To
allow a rapid screening of mutated clones, primers carried an additional
silent mutation introducing a new restriction site. The presence of the
desired mutations and the absence of additional ones were confirmed by
DNA sequencing (Genaxis, ˆmes, France). For double mutants, two
Quick-Change reactions were performed successively.
Cell culture and expression in HEK 293. Human embryonic kidney
(HEK) 293 cells were cultured in DMEM supplemented with 10% FCS
and transfected by electroporation as described previously (Brabet et al.,
1998; Franek et al., 1999). Unless stated otherwise, 10 10
6
cells were
transfected with plasmid DNA containing hemagglutinin (HA)-tagged
GABA
B1
(2
g) and cMyc-tagged GABA
B2
(2
g) and were completed to
a total amount of 10
g of plasmid DNA with pRK
6
. For determination of
inositol phosphate (IP) accumulation, the cells were also transfected with
the chimeric G
qi9 G-protein, which allows the coupling of the recom-
binant heteromeric GABA
B
receptor to phospholipase C (PLC) (Franek
et al., 1999).
Determination of IP accumulation. Determination of IP accumulation
in transfected cells was performed in 96-well plates (0.2.10
6
cells/well)
after overnight labeling with
3
H-myo-inositols (0.5
Ci/well), as de-
scribed previously (Chengalvala et al., 1999), with some modifications.
The stimulation was conducted for 30 min in a medium containing 10
mMLiCl and the indicated concentration of agonist or antagonist. The
reaction was stopped by replacing the medium with 0.1 Mformic acid.
Supernatants were recovered, and IP was purified by ion exchange chro-
matography using DOWEX AG1-X8 resin (Biorad, Marnes-la-Coquette,
France) in 96-well filter plates (ref:MAHVN4550; Millipore, Bedford,
MA). Total radioactivity remaining in the membrane fractions was
counted after treatment of cells with a solution containing 10% Triton
X-100 and 0.1N NaOH. Radioactivity was quantified using Wallac 1450
MicroBeta liquid scintillation counter. Data were expressed as the
amount of total IPs produced over the amount of radioactivity remaining
in the membranes plus the produced IP. Unless stated otherwise, all
data are means SEM of at least three independent experiments and
expressed in percentage of wild-type activity after a 30 min applica-
tion of 1 mMGABA. The dose–response curves were fitted using the
Figure1. Phylogenic analysis ofvarious VFTMs ( a) and comparison of the inactive and active
conformations of mGlu1 ( b) and NPRC ( c) VFTM dimers. a, The phylogenetic tree was con-
structed using the sequences of the VFTMs of the mGlu1 receptor, the amide-binding protein
(AmiC) from the amidase operon, the NR2A subunit of the rat NMDA receptor, leucine–
isoleucine–valine-bindingprotein(LIVBP),thenatriureticpeptidereceptortypesAandC(NPRA
and NPRC, respectively), RTK1 from Schistosoma mansoni, and the rat GABA
B1
and GABA
B2
subunits. Only branches with bootstrap values 600 are shown. Note that the RTK1 VFTM is
closer to the GABA
B
VFTMs than to any other VFTMs. b, Ribbon view of the inactive (left; pdb:
1ewt) and active (right; pdb:1ewk) forms of the dimer of mGlu1 VFTMs. Note the change in the
relative orientation between the two VFTMs and the closure of one VFTM (the blue– green one
in front) that bring together lobe II. c, Ribbon view of the inactive (left; pdb:1jdn) and active
(right; pdb:1jdp) forms of the dimer of NPRC VFTMs. Note that ANP binding at the interface
between the two VFTMs brings together lobe II.
Kniazeff et al. Locking the GABA
B
Receptor in Its Active State J. Neurosci., January 14, 2004 24(2):370 –377 371
Graph Pad (San Diego, CA) Prism program and the following equation:
y[(y
max
y
min
)/(1 (x/EC
50
)
nH
)] y
min
, where the EC
50
is the
concentration of the compound necessary to obtain 50% of the maximal
effect, and nH is the Hill coefficient.
Ligand-binding assay. Ligand-binding assay on intact HEK 293 cells
was performed as described previously using 0.1 nM[
125
I]CGP64213
(Galvez et al., 2001). Displacement curves were performed with at least
seven different concentrations of the displacer, and the curves were fitted
according to the equation: y [(y
max
y
min
)/(1 (x/IC
50
)
nH
)] y
min,
where the IC
50
is the concentration of the compound that inhibits 50% of
bound radioligand, and nH is the Hill coefficient. K
i
values were calcu-
lated according to the equation IC
50
K
i
(1 [RL]/ K
d
), where [RL] and
K
d
are the concentration and dissociation constant of the radioligand,
respectively. K
d
was determined assuming K
i
K
d
in the case of
CGP64213.
Western Blotting. Western blotting was performed as described previ-
ously (Kniazeff et al., 2002) using the rabbit polyclonal anti-HA antibody
(Zymed, San Francisco, CA) and the anti-rabbit HRP antibody (Amer-
sham, Saclay, France). The signal was revealed using an ECL assay.
Anti-HA ELISA assay for quantification of cell surface expression.
Twenty-four hours after transfection [10 10
6
cells, HA-tagged
GABA
B1
(2
g), and cMyc-tagged GABA
B2
(2
g) subunits], cells were
fixed with 4% paraformaldehyde and then blocked with PBS plus 5%
FBS. After a 30 min reaction with primary antibody (monoclonal
anti-HA clone 3F10; 0.5
g/ml; Roche, Basel, Switzerland) in the same
buffer, the goat anti-rat antibody coupled with HRP (Jackson Immu-
noresearch, West Grove, PA) was applied for 30 min at 1
g/ml. After
intense washes with PBS, secondary antibody was detected and quanti-
fied instantaneously by chemiluminescence using Supersignal ELISA
femto maximum sensitivity substrate (Pierce, Rockford, IL) and a Vic-
tor
2
luminescence counter (Wallac, Turku, Finland).
GTP-
-
35
S binding measurements. Cells were transfected using Poly-
Fect transfection reagent (Qiagen, Hilden, Germany) under optimized
conditions. Complexes were formed using a total amount of 8
gof
plasmid DNA with 60
l of PolyFect in 300
l of serum-free antibiotic-
free DMEM for 10 min and then added to cells at 4060% confluency.
According to expression results, the amount of DNA is 2
g of GABA
B1
,
1
g of GABA
B2
,2
gofG
o1c, and 3
gofpRK
6
for wild-type receptor
and 2
g of GABA
B1
,2
g of GABA
B2
,2
gofG
o1c, and 2
gofpRK
6
for CC1 and CC2 mutants.
Forty-eight hours after transfection, cells were scraped in lysis buffer
(15 mMTris, 2 mMMgCl2, and 0.3 mMEDTA, pH 7.4) and centrifuged
twice. The pellet was resuspended in Tris (50 mM), MgCl2 (3 mM), EGTA
(0.2 mM), and NaCl (60 mM), pH 7.4, using a potter. GTP-
-[
35
S] (1099
Ci/mmol; Amersham, Little Chalfont, UK) binding was performed in
96-well filtration plates (ref:MAFCN0B50; Millipore) equilibrated with
Tris (50 mM) and MgCl2 (5 mM), pH7.4. Membranes (5
g/well) were
preincubated or not with GABA (15 min; 1 mM; final volume, 20
l). The
plate was incubated for 1 hr at 30°C after the addition of 60
l of incu-
bation buffer (50 mMTris, 1 mMEDTA, 10
MGDP, 5 mMMgCl2, 0.01
mg/ml leupeptine, 100 mMNaCl, and 0.4 nMGTP-
-[
35
S]) and 20
lof
H
2
O. After vacuum filtration, plate-filter washing (three times with 250
lof50mMTris), and drying, the radioactivity was measured using a
1450 MicroBeta liquid scintillation counter (Wallac).
Results
Double cysteine mutations expected to lock the GABA
B1
VFTM in a closed state generate constitutively
active receptors
Based on three-dimensional models of the open and closed forms
of the GABA
B1
VFTM (Galvez et al., 1999, 2000a; Kniazeff et al.,
2002), some positions were selected to introduce cysteines that
could form a disulphide bridge in the closed state only. As de-
picted in Figure 2, the side chains of Ser247 or Ser246 from lobe I
and those of Thr315 or Glu316 from lobe II are far apart in a
model of the open form (Fig. 2a) but are in close proximity in the
closed form (Fig. 2b,c). Indeed, in the double mutants S247C-
T315C (CC1) (Fig. 2d) and S246C-E316C (CC2) (data not
shown), the distance between the sulfur atoms in the closed form
models is compatible with the formation of a disulphide bond
(1.9 and 2.1 Åfor CC1 and CC2, respectively). Double cysteine
mutations were introduced in GABA
B1
, and the CC1 and CC2
mutants were analyzed for function after coexpression with wild-
type GABA
B2
. Expression of the CC mutants in HEK 293 cells led
to a high constitutive activity in the absence of agonist reaching
69.8 4.8% (CC1) and 48.8 4.3% (CC2) (n3) of the max-
imal GABA-induced activity measured with the wild-type recep-
tor (Fig. 3a). In contrast, no significant increase in constitutive
activity can be detected with any of the single cysteine mutants
Figure 2. Three-dimensional models of the open ( a) and closed ( b) forms of the GABA
B1
VFTM and possible covalent linkage of both lobes in a closed form by a disulphide bridge. Lobe
I and lobe II are shown in red and blue ribbons, respectively. Residues subjected to mutagenesis
are represented. A closer view of these residues in the closed form model is shown in c.Ind, this
same region is shown for the CC1 mutant receptor, in which Ser247 and Thr315 were mutated
into cysteines.
Figure 3. The double cysteine mutants display a DTT-sensitive constitutive activity. Basal
andGABA (1 mM)-induced IP formation (black and white columns,respectively) weremeasured
in cells expressing wild-type (WT) or double cysteine GABA
B1
mutants, CC1 or CC2 ( a), or single
cysteine mutants S247C, T316C, and E316C ( b). c, Same as in a, but the GTP
S binding was
measured in membranes prepared from cells expressing the indicated GABA
B1
subunit, the
wild-type GABA
B2
subunit, and the Go1
subunit. d, Same as in a, but after a 30 min treatment
with 10 mMDTT, pH 8. Data are expressed as the percentage of the response obtained with the
wild-type receptor after 1 mMGABA stimulation and are means SEM of three independent
experiments (a,b,d) performed in triplicate. In c, data are means SEM of triplicate determi-
nations from one typical experiment.
372 J. Neurosci., January 14, 2004 24(2):370 –377 Kniazeff et al. Locking the GABA
B
Receptor in Its Active State
(Fig. 3b). Such a property of the CC mutants can be detected
using either the artificial coupling of the GABA
B
receptor to PLC
with the chimeric Gqi9 protein (Fig. 3a), but also through the
direct activation of Go protein as shown by GTP
[
35
S] binding
(Fig. 3c).
A disulphide bridge is involved in the constitutive activity of
CC mutants
In agreement with the involvement of a disulphide bond in the
high constitutive activity of CC mutants, this activity can no
longer be detected after reduction with DTT (Fig. 3d). This later
effect does not result from a destabilization of any of the subunits
attributable to reduction of native disulphide bonds for two rea-
sons. First, DTT treatment of cells expressing the wild-type re-
ceptor had no significant effect on the basal activity, and only a
small decrease in GABA-mediated response was observed (Fig.
3d). Second, after reduction, CC mutants can still be activated by
agonists (Fig. 3d) with an affinity similar to that measured on the
wild-type receptor (Table 1).
The constitutive activity of CC mutants is not inhibited by
competitive antagonists
Competitive antagonists of mGluRs have been shown to bind in
the open form of the VFTM, as illustrated with the mGlu1
(Tsuchiya et al., 2002) and mGlu8 (Bessis et al., 2002) receptors.
Accordingly, a receptor locked in a closed state would not be
expected to bind competitive antagonists. As shown in Figure 4a,
the antagonist CGP64213 did not inhibit the basal activity of the
CC mutants, whereas it was able to (1) inhibit the basal activity of
the wild-type receptor and (2) antagonize GABA-mediated re-
sponses of single cysteine mutants (Table 1). Moreover, after
DTT treatment, this antagonist become effective in fully inhibit-
ing GABA activation of the CC mutants (Fig. 4b) demonstrating
that a disulphide bridge, but not the presence of the two cysteines,
is responsible for the absence of antagonist action. These data
show that the constitutively active receptor is locked in its active
state and cannot return to the inactive antagonist-stabilized state.
Not all surface-expressed CC1 mutants are locked in an
active state
Although high constitutive activity can be observed in cells ex-
pressing the CC1 mutant, GABA [as well as its chlorophenyl de-
rivative baclofen (data not shown)] could still further increase the
response with a potency identical to that measured on the wild-
type receptor (Fig. 5a). This could result from the disulphide
bridge-stabilized receptors being in a partially active conforma-
tion. However, the agonist-induced response can be fully antag-
onized by CGP64213 with the same potency as that measured on
the wild-type receptor (Fig. 5b), indicating that the antagonist has
access to agonist-activated receptors. These data suggest that
there are two populations of receptors, some constitutively active
and insensitive to antagonists, and some that can be activated by
agonists and inhibited by CGP64213. In agreement with this pro-
posal, a small amount of specific [
125
I]CGP64213 binding can be
detected in cells expressing the CC1 mutant, and this binding was
largely increased after DTT treatment (approximately fivefold) in
contrast to the wild-type receptor (Fig. 6a). This effect was not
attributable to a change in the CGP64213 affinity because the
same K
i
values for this antagonist could be determined by dis-
placement studies on CC1-expressing cells before and after DTT
treatment (Table 1). This shows that reduction reveals new bind-
Table 1. Potencies of the agonist GABA and the antagonist CGP64213 on wild-type (WT), CC1, CC2, and the single cysteine mutants measured either with a functional assay
(IP production) or by displacement of [
125
I]CGP64213 binding under control condition or after DTT treatment
IP production [
125
I]CGP64213 binding
Control DTT Control DTT
GABA
EC
50
(
M)
CGP64213
IC
50
(nM)
GABA
EC
50
(
M)
CGP64213
IC
50
(nM)
GABA
K
i
(
M)
CGP64213
K
i
(nM)
GABA
K
i
(
M)
CGP64213
K
i
(nM)
WT 0.23 0.02 10.5 1.3 0.34 0.06 12.6 0.8 4.74 0.61 1.28 0.13 2.75 0.97 1.53 0.21
CC1 0.43 0.07 13.1 2.7 0.26 0.08 10.3 0.3 n.t. 3.61 0.81 1.59 0.34 3.43 1.00
CC2 0.33 0.07 12.8 0.7 0.29 0.05 12.0 0.7 n.t. 2.80 0.96 n.t. n.t.
S247C 0.36 0.06 n.t. n.t. n.t. 8.41 0.80 1.67 0.73 n.t. n.t.
T315C 0.28 0.06 n.t. n.t. n.t. 9.50 0.46 1.88 0.98 n.t. n.t.
E316C 0.85 0.017 n.t. n.t. n.t. 5.92 1.5 2.13 0.73 n.t. n.t.
Values are means SEM of at least three independent experiments. n.t., Not tested.
Figure 4. Constitutive activity of CC mutants is not inhibited by the GABA
B
competitive
antagonist CGP64213. a, The high basal IP production measured with the CC mutants (black
columns) is not inhibited by the GABA
B
antagonist CGP64213 (300 nM; gray columns), in con-
trast to the basal activity of the wild-type (WT) receptor. b, CGP64213 antagonizes with the
same potency GABA (10
M)-stimulated IP formation in cells expressing the wild-type GABA
B
receptor under control condition (black circles) or after DTT treatment (gray circles), or in cells
expressing the CC1 mutant after DTT treatment (gray squares). Data are means SEM of
triplicates from one representative of three independent experiments.
Figure 5. Cells expressing CC1 remain sensitive to both GABA and CGP64213 with the same
potency as cells expressing wild-type (WT) receptor. Concentration-dependent effect of GABA
(a) or CGP64213 (in the presence of 10
MGABA) ( b) on IP production in cells expressing
wild-type (circles, dashed line) or CC1 (squares, solid line) GABA
B
receptor. Data are means
SEM of triplicates from one representative of three independent experiments.
Kniazeff et al. Locking the GABA
B
Receptor in Its Active State J. Neurosci., January 14, 2004 24(2):370 –377 373
ing sites in cells expressing the CC1 mutant. Finally, the specific
binding of [
125
I]CGP64213 after DTT treatment represented half
of that determined on cells expressing the wild-type receptor, in
agreement with a twofold lower expression level of this receptor
(Fig. 6b,c). Therefore, most CGP64213-binding sites have been
unmasked after DTT reduction. Taken together, these data show
that 75.5 6.9% of the binding sites in cells expressing the CC1
mutant are inaccessible to CGP64213 because of a disulphide
bridge. Thus, the effect of GABA on these cells likely results from
the 25% remaining receptors because this effect is fully inhibited
by this antagonist. Another conclusion from these data are that
GABA is unlikely able to activate the constitutively active,
CGP64213-insensitive form of the receptor.
We then examined whether the proportion of disulphide-
locked receptors could be modulated by oxidizing agents 5,5-
dithio-bis(2-nitrobenzoic acid) or Cu-Phenantroline by agonists
that are expected to stabilize the closed state or by antagonists
expected to prevent the formation of the closed state. None of
these treatments alone or in combination lead to a modification
of constitutive activity (Fig. 7). Moreover, after DTT treatment,
constitutive activity of the CC1 mutant could not be restored
after 30 min of exposure to the oxidizing agents (data not shown).
This suggests that all disulphide-locked receptors are formed in-
side the cells rather than after plasma membrane insertion.
Constitutive activity of the disulphide-locked CC1 mutant is
close to the agonist-induced activity of the wild-type receptor
We then examined whether the constitutive activity of the CC1
mutant corresponds to a fully active form of the receptor. Surface
expression was determined with an ELISA assay, and second mes-
senger formation was measured in cells expressing various
amount of the GABA
B
heterodimer. A saturating curve was ob-
served, and the slope at the origin of the curves is indicative of the
specific activity of the receptor (amount of second messengers
produced per receptor) (Fig. 8). In the absence of agonist, the
specific activity of CC1 is about half the specific activity of the
wild-type receptor activated by GABA (0.7 10
3
and 1.4
10
3
arbitrary units, respectively), but, after DTT treatment, this
activity is decreased to the level of the basal activity of the wild-
type receptor. Because only 75% of the CC1 receptors are ex-
pected to be stabilized in an active form by a disulphide bridge
(see above), this indicates that the specific activity of the consti-
tutively active form of CC1 receptors is close to the specific activ-
ity of the agonist-activated wild-type receptors (1.0 10
3
and
1.4 10
3
arbitrary units, respectively).
The curves obtained clearly saturate, indicating a maximal IP
formation could be measured with a nonsaturating amount of
receptors expressed at the cell surface. This is in contrast to what
can be observed under similar conditions with the Gq-coupled
mGlu5 receptor (Goudet et al., 2004). This may, therefore, result
from the use of the chimeric G-protein
subunit Gqi9 cotrans-
fected with the receptor to make it able to activate PLC. It is
possible that either the amount of Gqi9 or endogenous
␤␥
sub-
units is limiting when the receptor is overexpressed.
Double cysteine mutations in the GABA
B2
VFTM does not
influence receptor function
Can the closure of the GABA
B2
VFTM also lead to receptor acti-
vation? To test this possibility, cysteines were introduced in the
GABA
B2
VFTM at the same positions as in GABA
B1
, but neither
constitutive activity nor changes in agonist efficacy and potency
were detected (data not shown). To examine whether such cys-
teines could form a disulphide bond in the GABA
B2
VFTM, and
as such to lock this domain in a closed state, Western blot exper-
iments were conducted under reducing and nonreducing (100
mMDTT, pH 8) conditions. Such an experiment performed with
the full-length receptor did not lead to a clear conclusion, likely
because of the large size of these receptor subunits. As such, con-
structs were created (GB1 and GB2), in which a stop codon
Figure 6. Cells expressing CC1 GABA
B
receptors possess CGP64213-binding sites at their
surface, and new sites are unmasked after DTT treatment. a,[
125
I]CGP64213 binding under
control condition (white column) or after DTT treatment (black column) on intact cells express-
ing GABA
B2
and wild-type (WT) or CC1 GABA
B1
subunits. Data are means of triplicates from one
representative of three independent experiments. b, Western blot on total membranes of cells
mock-transfected or transfected with plasmid expressing HA-tagged wild-type or CC1 mutant.
c, Quantification of cell surface expression of wild-type (WT) and CC1 mutant GABA
B1
subunit
using an ELISA assay in HEK 293-transfected cells.
Figure 7. Neither oxidizing treatment nor overnight incubation with agonist or antagonist
increasetheconstitutive activity. Basal and GABA (1 mM)-induced IP formation (black and white
columns, respectively) were measured under control (CTR) or after various treatment: 0.5 mM
5,5-dithio-bis(2-nitrobenzoic acid (DTNB) for 30 min, 1 mMCuSO
4
plus 4 mMphenanthroline
(CuP) for 30 min, CuP in the presence of 10
MGABA for 30 min, and overnight incubation with
CGP64213 (100 nM) or GABA (1 mM) applied immediately after transfection.
374 J. Neurosci., January 14, 2004 24(2):370 –377 Kniazeff et al. Locking the GABA
B
Receptor in Its Active State
was introduced after TM1, leading to the expression of the VFTM
attached to the membrane by the first transmembrane domain.
As shown in Figure 9, these constructs migrated faster under
nonreducing conditions, in agreement with the proposed exis-
tence of native disulphide bridges in the VFTM of both GABA
B1
and GABA
B2
(Galvez et al., 2000a; Kniazeff et al., 2002). Of inter-
est, both GB1-CC and GB2-CC migrated slightly faster than
the wild types under the nonreducing condition, consistent with
the existence of an additional disulphide bridge in these
constructs.
Discussion
The present study indicates that the introduction of two cysteines
expected to lock the GABA
B1
VFTM in a closed form generates a
constitutively active heterodimeric GABA
B
receptor.
A disulphide bond locks the GABA
B
receptor in an active state
GPCRs are assumed to oscillate between various inactive (R) and
active (R*) conformations (Lefkowitz, 1993; Samama et al., 1993;
Leff, 1995; Farrens et al., 1996; Christopoulos and Kenakin,
2002), the latter being stabilized by agonists, whereas the former
is stabilized by inverse agonists. Mutations have been identified
that lead to constitutive activity (Lefkowitz, 1993; Samama et al.,
1993; Leff, 1995; Christopoulos and Kenakin, 2002; Parnot et al.,
2002), but these have been shown to destabilize the inactive con-
formations rather than to stabilize an active one (Parnot et al.,
2002). As such, these mutations are expected to displace the
R7R* equilibrium toward R*, as demonstrated by the inhibition
of constitutive activity by inverse agonists, but did not bring
much information on the active conformation of the receptor.
The GABA
B1
CC mutants appear to be locked in an active state
through the formation of an additional disulphide bond. Indeed,
the R7R* equilibrium seems to no longer exist because the re-
ceptor cannot return to the inactive state even in the presence of
a high concentration of the inverse agonist CGP64213. Only DTT
treatment allowed the receptor to return to an inactive state. This
represents the first example of a constitutively active form of a
GPCR resulting from the locking of an active state.
A disulphide bond locks the GABA
B1
VFTM in a closed state
Several data are consistent with the two cysteines introduced in
the CC1 and CC2 mutants involved in a disulphide bridge that
locks the GABA
B1
VFTM in a closed state. First, the VFTM of the
CC1 mutant migrates faster than the wild type in acrylamide gels
under nonreducing conditions. Second, the distance between the
two sulfydryl groups introduced is consistent with the formation
of a disulphide bond in a closed form model of the GABA
B1
VFTM only. Third, the GABA
B
antagonist CGP64213 is unable to
(1) inhibit the constitutive activity of the CC mutants and (2)
bind on the disulphide-locked receptor, although this antagonist
can bind and exert its normal antagonist action after DTT treat-
ment. Indeed, as observed for the mGlu1 (Tsuchiya et al., 2002)
and mGlu8 (Bessis et al., 2002) receptors, many GABA
B
compet-
itive antagonists are larger than agonists and cannot fit into a
closed form model of the binding site. Finally, modeling and
mutagenesis studies suggested that the active form of the GABA
B1
VFTM corresponds to a closed form because residues from both
lobes are involved in agonist binding (Galvez et al., 1999, 2000a;
Bernard et al., 2001; Costantino et al., 2001; Kniazeff et al., 2002).
However, the absence of modulation by oxidants or ligands
expected to help or prevent the formation of the disulphide
bridge was surprising. Accordingly, the disulphide-locked recep-
tors are likely formed during the synthesis of the receptor in the
endoplasmic reticulum. When the receptor is correctly folded
and at the cell surface, in the absence of the preformed disulphide
bond or after its reduction with DTT, the sulfydryl groups may
not be in a correct orientation to spontaneously form a disul-
phide bond. Indeed, in our three-dimensional model, the side
chains of mutated residues had to be reoriented manually for the
disulphide bridge to form. Moreover, this area in the GABA
B1
VFTM has been proposed to constitute a Ca
2
-binding site
(Galvez et al., 2000b; Costantino et al., 2001), thus stabilizing a
precise position of the side chains of the surrounding residues
and possibly preventing the formation of a disulphide bond.
The disulphide-locked active state is close to the agonist-
stabilized active state
Although cells expressing the CC1 or CC2 mutants display a high
constitutive PLC activity, IP formation could still be increased by
Figure 8. Constitutive activity of CC1 mutant receptors is close to GABA-mediated activity of
the wild-type receptor. Basal (diamonds) and GABA-mediated (squares) IP formation were
measured in cells transfected with HA-tagged wild-type (open symbols, thin lines) or CC1 mu-
tant (filled symbols, bold lines) GABA
B1
subunit (2
g) and various amounts of GABA
B2
-
expressing plasmids (0.05–2
g). The amount of GABA
B
receptors at the cell surface was
measuredusingELISAandaHAantibody.BasalIPformationincellsexpressing various amounts
of CC1 mutant containing GABA
B
receptor was also measured after DTT treatment (closed tri-
angles). Data are means SEM of triplicate determinations from a single experiment. Similar
data were obtained in three independent experiments. Data points were fitted according to the
saturation equation: IP IPmax Exp/(Cte Exp), where IP is the amount of IPs produced
under the indicated condition, IPmax is the maximal IP response obtained with a high expres-
sion level of the receptor, Exp is the signal obtained with the ELISA, and Cte is a constant.
Figure 9. Migration of the VFTMs of GABA
B1
and GABA
B2
is affected by the presence of the
two additional cysteines. Total proteins from cells expressing the wild-type (WT) or the CC
version of GB1 or GB2 (GABA
B1
and GABA
B2
, in which a stop codon has been introduced
after TM1) were separated on an acrylamide gel under nonreducing conditions or after treat-
ment with 100 mMDTT, pH 8. After transfer, the proteins were detected using an anti-HA
antibody directed against the HA epitope inserted after the signal peptide. Data shown are
representative of three independent experiments.
Kniazeff et al. Locking the GABA
B
Receptor in Its Active State J. Neurosci., January 14, 2004 24(2):370 –377 375
GABA
B
agonists. This may seem surprising in the case of the CC2
mutant because Ser246 (mutated into Cys in this mutant) has
been proposed previously to form an H-bond with the carboxylic
function of GABA
B
ligands (Galvez et al., 2000a; Bernard et al.,
2001; Costantino et al., 2001; Kniazeff et al., 2002). However,
because an SH group can also form H-bonds, it is likely that the
replacement of the OH group of Ser246 by an SH group has
minor effect on ligand-binding affinities.
In the case of the ionotropic glutamate receptor subtypes,
partial agonists stabilized a partially closed form of the
glutamate-binding domain (Armstrong and Gouaux, 2000; Arm-
strong et al., 2003). Therefore, one possibility to explain the
agonist-induced response in CC mutant-expressing cells was that
the disulphide bond locked the receptor in a partially closed form
that could be closed further in the presence of agonist. However,
our data indicate that this is unlikely the case but rather that the
agonist-mediated response is attributable to a certain fraction of
the receptors at the cell surface that are not locked in their active
state by a disulphide bridge. Indeed, a fraction of the CC mutants
can bind with a normal affinity to the antagonist CGP64213, and
the agonist-induced activity can be inhibited by the antagonist
with a wild-type K
i
. Taking this into consideration, together with
the receptor density at the cell surface, we estimated that the
specific activity of the constitutively active disulphide-locked
CC1 mutant is close to that of the agonist-stabilized form of the
wild-type receptor. This suggests that the conformation of the
disulphide-locked CC1 VFTM is close to that of the agonist-
bound form of the wild-type receptor.
Comparison with mGlu1 and ANP receptors
As shown in Figure 1a, the GABA
B
VFTMs are as distant from the
mGlu1 VFTM than from any other VFTMs. Moreover, the large
insertions found in mGlu VFTMs and conserved in the related
Ca
2
-sensing, pheromone, and taste receptors are not found in
the GABA
B
VFTMs. Finally, whereas the mGlu-like receptors
possess a cysteine-rich domain that connects their VFTM to the
heptahelical domain, the GABA
B
subunits do not. In contrast,
the GABA
B
VFTMs share a significant higher similarity with the
VFTM of the monotopic receptor RTK1 (Vicogne et al., 2003),
leaving open the possibility that the dimer of VFTMs of the
dimeric GABA
B
receptor functions like those of dimeric mono-
topic receptors. However, in the case of the monotopic receptor
for the natriuretic peptide, both VFTMs are in a closed confor-
mation even in the inactive state (He et al., 2001; van den Akker,
2001) (Fig. 1c), whereas agonists seem to stabilize a closed form of
the VFTM in the case of the mGluRs (Kunishima et al., 2000;
Bessis et al., 2002; Tsuchiya et al., 2002) (Fig. 1b). Accordingly,
our data suggest the dimer of GABA
B
VFTMs functions like that
of mGluRs despite some structural dissimilarities between these
two types of receptors.
Role of GABA
B2
VFTM in GABA
B
receptor activation
GABA
B2
possesses a VFTM similar to that of GABA
B1
. Although
required for GABA
B
receptor activation (Galvez et al., 2001;
Margeta-Mitrovic et al., 2001a), the GABA
B2
VFTM does not
seem to bind any ligand (Kniazeff et al., 2002). Our data show
that introduction of two cysteines at the same position as in CC1
and CC2 mutants in the GABA
B2
VFTM does not change the
properties of the heteromeric GABA
B
receptor. However, as ob-
served with the GABA
B1
CC mutant, the GABA
B2
CC migrates
faster than the wild type under nonreducing conditions, consis-
tent with the existence of an additional disulphide bridge in this
mutant. This suggests that the closure of GABA
B2
is not sufficient
to activate the receptor. Indeed, either GABA
B2
VFTM is always
closed or the closure of GABA
B2
VFTM has no detectable effect
on the function of the receptor. This is consistent with the closure
of GABA
B1
VFTM being sufficient for GABA
B
receptor activa-
tion, in agreement with our proposal that GABA
B
agonists bind
in the GABA
B1
VFTM only (Kniazeff et al., 2002). Moreover, our
data also suggest that even an artificial ligand interacting in the
cleft of GABA
B2
is not expected to have an important effect on the
functioning of the heterodimer.
Allostery between the four main domains of the
heterodimeric GABA
B
receptor
Although GABA binds to the GABA
B1
VFTM only, the GABA
B2
7TM is critical for G-protein coupling (Galvez et al., 2001;
Margeta-Mitrovic et al., 2001b; Robbins et al., 2001; Duthey et al.,
2002). How can the closure of the GABA
B1
VFTM lead to the
change in conformation of the GABA
B2
7TM domain necessary
for G-protein activation? In the case of mGluRs, it has been pro-
posed that the change in the relative orientation of the two
VFTMs, observed on glutamate binding, may be required for
activation (Kunishima et al., 2000; Jensen et al., 2001, 2002).
Based on this hypothesis, the closure of the GABA
B1
VFTM pos-
sibly leads to a change in the relative orientation of the two
VFTMs in the heterodimer as proposed for the mGlu1 receptor,
no matter whether the GABA
B2
VFTM is in a closed or open
conformation.
Conclusion
Our present study brings much information on how GABA acti-
vates the GABA
B
receptor and on the specific roles played by each
subunit in this heteromeric receptor. Moreover, such informa-
tion may be of interest for the understanding of the activation
process of many other GPCRs, Indeed, the rhodopsin-like class I
GPCRs also form dimers (Bouvier, 2001; Fotiadis et al., 2003). It
is still unknown whether such dimers are required for activation
and whether, in such a case, agonist occupation of a single pro-
tomer is sufficient for full activation of the receptor. Taking into
account our data and the fact that retina can detect a single pho-
ton although rhodopsin forms oligomers of dimers (Fotiadis et
al., 2003; Liang et al., 2003), one may propose that an initial
change in conformation in a single subunit may be sufficient to
trigger the activation of some dimeric GPCRs.
References
Armstrong N, Gouaux E (2000) Mechanisms for activation and antagonism
of an AMPA-sensitive glutamate receptor: crystal structures of the GluR2
ligand binding core. Neuron 28:165181.
Armstrong N, Mayer M, Gouaux E (2003) Tuning activation of the AMPA-
sensitive GluR2 ion channel by genetic adjustment of agonist-induced
conformational changes. Proc Natl Acad Sci USA 100:57365741.
Bernard P, Guedin D, Hibert M (2001) Molecular modeling of the GABA/
GABA(B) receptor complex. J Med Chem 44:2735.
Bessis A-S, Rondard P, Gaven F, Brabet I, Triballeau N, Pre´zeau L, Acher F,
Pin J-P (2002) Closure of the Venus flytrap module of mGlu8 receptor
and the activation process: insights from mutations converting antago-
nists into agonists. Proc Natl Acad Sci USA 99:1109711102.
Bockaert J, Pin JP (1999) Molecular tinkering of G protein-coupled recep-
tors: an evolutionary success. EMBO J 18:17231729.
Bouvier M (2001) Oligomerization of G-protein-coupled transmitter re-
ceptors. Nat Rev Neurosci 2:274286.
Brabet I, Parmentier ML, De Colle C, Bockaert J, Acher F, Pin JP (1998)
Comparative effect of L-CCG-I, DCG-IV and gamma-carboxy-L-
glutamate on all cloned metabotropic glutamate receptor subtypes. Neu-
ropharmacology 37:10431051.
Chengalvala M, Kostek B, Frail DE (1999) A multi-well filtration assay for
376 J. Neurosci., January 14, 2004 24(2):370 –377 Kniazeff et al. Locking the GABA
B
Receptor in Its Active State
quantitation of inositol phosphates in biological samples. J Biochem Bio-
phys Methods 38:163170.
Christopoulos A, Kenakin T (2002) G protein-coupled receptor allosterism
and complexing. Pharmacol Rev 54:323374.
Costantino G, Macchiarulo A, Entrena Guadix A, Pellicciari R (2001) QSAR
and molecular modeling studies of baclofen analogues as GABA(B) ago-
nists. Insights into the role of the aromatic moiety in GABA(B) binding
and activation. J Med Chem 44:18271832.
Duthey B, Caudron S, Perroy J, Bettler B, Fagni L, Pin J-P, Pre´zeau L (2002)
A single subunit (GB2) is required for G-protein activation by the het-
erodimeric GABAB receptor. J Biol Chem 277:32363241.
Farrens DL, Altenbach C, Yang K, Hubbell WL, Khorana HG (1996) Re-
quirement of rigid-body motion of transmembrane helices for light acti-
vation of rhodopsin. Science 274:768770.
Felder CB, Graul RC, Lee AY, Merkle HP, Sadee W (1999) The Venus flytrap
of periplasmic binding proteins: an ancient protein module present in
multiple drug receptors. AAPS PharmSci 1:120.
Fotiadis D, Liang Y, Filipek S, Saperstein DA, Engel A, Palczewski K (2003)
Atomic-force microscopy: rhodopsin dimers in native disc membranes.
Nature 421:127128.
Franek M, Pagano A, Kaupmann K, Bettler B, Pin J-P, Blahos II J (1999) The
heteromeric GABA-B receptor recognizes G-protein
subunit
C-termini. Neuropharmacology 38:16571666.
Fredriksson R, Lagerstrom MC, Lundin LG, Schioth HB (2003) The
G-protein-coupled receptors in the human genome form five main fam-
ilies. Phylogenetic analysis, paralogon groups, and fingerprints. Mol
Pharmacol 63:12561272.
Galvez T, Parmentier ML, Joly C, Malitschek B, Kaupmann K, Kuhn R, Bit-
tiger H, Froestl W, Bettler B, Pin JP (1999) Mutagenesis and modeling of
the GABAB receptor extracellular domain support a Venus flytrap mech-
anism for ligand binding. J Biol Chem 274:1336213369.
Galvez T, Prezeau L, Milioti G, Franek M, Joly C, Froestl W, Bettler B, Ber-
trand HO, Blahos J, Pin JP (2000a) Mapping the agonist-binding site of
GABAB type 1 subunit sheds light on the activation process of GABAB
receptors. J Biol Chem 275:4116641174.
Galvez T, Urwyler S, Pre´zeau L, Mosbacher J, Joly C, Malitschek B, Heid J,
Brabet I, Froestl W, Bettler B, Kaupmann K, Pin J-P (2000b) Ca
2
-
requirement for high affinity
-aminobutyric acid (GABA) binding at
GABA
B
receptors: involvement of serine 269 of the GABA
B
R1 subunit.
Mol Pharmacol 57:419426.
Galvez T, Duthey B, Kniazeff J, Blahos J, Rovelli G, Bettler B, Prezeau L, Pin JP
(2001) Allosteric interactions between GB1 and GB2 subunits are re-
quired for optimal GABA(B) receptor function. EMBO J 20:21522159.
Goudet C, Gaven F, Kniazeff J, Vol C, Liu J, Cohen-Gonsaud M, Acher F,
Prézeau L, Pin JP (2004) Heptahelical domain of metabotropic gluta-
mate receptor 5 behaves like rhodopsin-like receptors. Proc Natl Acad Sci
USA, in press.
Guex N, Peitsch MC (1997) SWISS-MODEL and the Swiss-PdbViewer:
an environment for comparative protein modeling. Electrophoresis
18:27142723.
He X-L, Chow D-C, Martick MM, Garcia KC (2001) Allosteric activation of
a spring-loaded natriuretic peptide receptor dimer by hormone. Science
293:16571662.
Jensen AA, Sheppard PO, Jensen LB, OHara PJ, Bra¨uner-Osborne H (2001)
Construction of a high affinity zinc binding site in the metabotropic glu-
tamate receptor mGluR1: noncompetitive antagonism from the amino-
terminal domain of a family C G-protein-coupled receptor. J Biol Chem
276:1011010118.
Jensen AA, Greenwood JR, Bra¨uner-Osborne H (2002) The dance of the
clams: twists and turns in the family C GPCR homodimer. Trends Phar-
macol Sci 23:491493.
Jones KA, Borowsky B, Tamm JA, Craig DA, Durkin MM, Dai M, Yao WJ,
Johnson M, Gunwaldsen C, Huang LY, Tang C, Shen Q, Salon JA, Morse
K, Laz T, Smith KE, Nagarathnam D, Noble SA, Branchek TA, Gerald C
(1998) GABA(B) receptors function as a heteromeric assembly of the
subunits GABA(B)R1 and GABA(B)R2. Nature 396:674679.
Kaupmann K, Malitschek B, Schuler V, Heid J, Froestl W, Beck P, Mosbacher
J, Bischoff S, Kulik A, Shigemoto R, Karschin A, Bettler B (1998)
GABA(B)-receptor subtypes assemble into functional heteromeric com-
plexes. Nature 396:683687.
Kniazeff J, Galvez T, Labesse G, Pin JP (2002) No ligand binding in the GB2
subunit of the GABA(B) receptor is required for activation and allosteric
interaction between the subunits. J Neurosci 22:73527361.
Kolakowski LF (1994) GCRDb: a G-protein-coupled receptor database. Re-
ceptors Channels 2:17.
Kunishima N, Shimada Y, Tsuji Y, Sato T, Yamamoto M, Kumasaka T, Nakanishi
S, Jingami H, Morikawa K (2000) Structural basis of glutamate recognition
by a dimeric metabotropic glutamate receptor. Nature 407:971977.
Leff P (1995) The two-state model of receptor activation. Trends Pharmacol
Sci 16:8997.
Lefkowitz RJ (1993) Turned on to ill effect. Nature 365:603604.
Liang Y, Fotiadis D, Filipek S, Saperstein DA, Palczewski K, Engel A (2003)
Organization of the G protein-coupled receptors rhodopsin and opsin in
native membranes. J Biol Chem 278:2165521662.
Margeta-Mitrovic M, Jan YN, Jan LY (2000) A trafficking checkpoint con-
trols GABA(B) receptor heterodimerization. Neuron 27:97106.
Margeta-Mitrovic M, Jan YN, Jan LY (2001a) Ligand-induced signal trans-
duction within heterodimeric GABA(B) receptor. Proc Natl Acad Sci USA
98:1464314648.
Margeta-Mitrovic M, Jan YN, Jan LY (2001b) Function of GB1 and GB2
subunits in G protein coupling of GABA(B) receptors. Proc Natl Acad Sci
USA 98:1464914654.
OHara PJ, Sheppard PO, Thøgersen H, Venezia D, Haldeman BA, McGrane
V, Houamed KM, Thomsen C, Gilbert TL, Mulvihill ER (1993) The
ligand-binding domain in metabotropic glutamate receptors is related to
bacterial periplasmic binding proteins. Neuron 11:4152.
Pagano A, Rovelli G, Mosbacher J, Lohmann T, Duthey B, Stauffer D, Ristig
D, Schuler V, Meigel I, Lampert C, Stein T, Pre´zeau L, Blahos J, Pin J-P,
Froestl W, Kuhn R, Heid J, Kaupmann K, Bettler B (2001) C-terminal
interaction is essential for surface trafficking but not for heteromeric
assembly of GABA
B
receptors. J Neurosci 21:11891202.
Page RDM (1996) TreeView: an application to display phylogenetic trees on
personal computers. Comput Appl Biosci 12:357358.
Paoletti P, Perin-Dureau F, Fayyazuddin A, Le Goff A, Callebaut I, Neyton J
(2000) Molecular organization of a zinc binding N-terminal modulatory
domain in a NMDA receptor subunit. Neuron 28:911925.
Parnot C, Miserey-Lenkei S, Bardin S, Corvol P, Clauser E (2002) Lessons
from constitutively active mutants of G protein-coupled receptors.
Trends Endocrinol Metab 13:336343.
Pin JP, Galvez T, Prezeau L (2003) Evolution, structure, and activation
mechanism of family 3/C G-protein-coupled receptors. Pharmacol Ther
98:325354.
Robbins MJ, Calver AR, Filippov AK, Hirst WD, Russell RB, Wood MD, Nasir
S, Couve A, Brown DA, Moss SJ, Pangalos MN (2001) GABA
B2
is essen-
tial for G-protein coupling of the GABA
B
receptor heterodimer. J Neuro-
sci 21:80438052.
Saitou N, Nei M (1987) The neighbor-joining method: a new method for
reconstructing phylogenetic trees. Mol Biol Evol 4:406425.
Sali A, Blundell TL (1993) Comparative protein modelling by satisfaction of
spatial restraints. J Mol Biol 234:779815.
Samama P, Cotecchia S, Costa T, Lefkowitz RJ (1993) A mutation-induced
activated state of the
2-adrenergic receptor. J Biol Chem 268:46254636.
Takahashi K, Tsuchida K, Tanabe Y, Masu M, Nakanishi S (1993) Role of
the large extracellular domain of metabotropic glutamate receptors in
agonist selectivity determination. J Biol Chem 268:1934119345.
Thompson JD, Higgins DG, Gibson TJ (1994) Clustal W: improving the
sensitivity of progressive multiple sequence alignment through sequence
weighting, position-specific gap penalties and weight matrix choice. Nu-
cleic Acids Res 22:46734680.
Tsuchiya D, Kunishima N, Kamiya N, Jingami H, Morikawa K (2002)
Structural views of the ligand-binding cores of a metabotropic glutamate
receptor complexed with an antagonist and both glutamate and Gd
3
.
Proc Natl Acad Sci USA 99:26602665.
van den Akker F (2001) Structural insights into the ligand binding domains
of membrane bound guanylyl cyclases and natriuretic peptide receptors. J
Mol Biol 311:923937.
Vicogne J, Pin JP, Lardans V, Capron M, Noel C, Dissous C (2003) An
unusual receptor tyrosine kinase of Schistosoma mansoni contains a Ve-
nus flytrap module. Mol Biochem Parasitol 126:5162.
White JH, Wise A, Main MJ, Green A, Fraser NJ, Disney GH, Barnes AA,
Emson P, Foord SM, Marshall FH (1998) Heterodimerization is re-
quired for the formation of a functional GABA(B) receptor. Nature 396:
679682.
Kniazeff et al. Locking the GABA
B
Receptor in Its Active State J. Neurosci., January 14, 2004 24(2):370 –377 377
... To our knowledge, this was the first experimental suggestion that GPCRs form oligomers. GPCRs that have been reported to generate homomeric or heteromeric complexes include opioid receptors (ORs) [2,3], dopamine D 1,2,3 receptors [4,5], metabotropic glutamate receptor 5 (mGluR5) [6,7], g-aminobutyric acid receptors (GABA B1,B2 ) [8,9] and a 2 -adrenergic/M3 muscarinic receptors [10]. Most of the evidence supporting the existence of GPCR dimers also corroborates the existence of GPCR oligomers; such possibilities cannot be readily distinguished by currently available techniques. ...
... The authors envisaged that at least two polyethylene glycol (PEG) units, in addition to a biphenyltriazole-based moiety, would be necessary to achieve that. Inspection of the ligand-bound crystal structures of D 3 R and NTS1R clearly revealed that the 4 0 -position on the pyrrole ring of eticlopride (a D 3 R antagonist) and the N terminus amino of NT (8)(9)(10)(11)(12)(13) [neurotensin (8)(9)(10)(11)(12)(13) peptide, an NTS1R agonist] are accessible from the extracellular side, and were therefore chosen as the attachment points for bridging the two lead compounds; the designed bivalent compound connected the two pharmacophores at these points (Fig. 2d) [42]. These compounds showed high selectivity (up to three orders of magnitude) and binding affinities in the picomolar range for cells co-expressing D 2 R and NTS1R, compared with cells only expressing D 2 R. ...
... The authors envisaged that at least two polyethylene glycol (PEG) units, in addition to a biphenyltriazole-based moiety, would be necessary to achieve that. Inspection of the ligand-bound crystal structures of D 3 R and NTS1R clearly revealed that the 4 0 -position on the pyrrole ring of eticlopride (a D 3 R antagonist) and the N terminus amino of NT (8)(9)(10)(11)(12)(13) [neurotensin (8)(9)(10)(11)(12)(13) peptide, an NTS1R agonist] are accessible from the extracellular side, and were therefore chosen as the attachment points for bridging the two lead compounds; the designed bivalent compound connected the two pharmacophores at these points (Fig. 2d) [42]. These compounds showed high selectivity (up to three orders of magnitude) and binding affinities in the picomolar range for cells co-expressing D 2 R and NTS1R, compared with cells only expressing D 2 R. ...
Article
G protein-coupled receptors (GPCRs) have been exploited as primary targets for drug discovery, and GPCR dimerization offers opportunities for drug design and disease treatment. An important strategy for targeting putative GPCR dimers is the use of bivalent ligands, which are single molecules that contain two pharmacophores connected through a spacer. Here, we discuss the selection of pharmacophores, the optimal length and chemical composition of the spacer, and the choice of spacer attachment points to the pharmacophores. Furthermore, we review the most recent advances (from 2018 to the present) in the design, discovery and development of bivalent ligands. We aim to reveal the state-of-the-art design strategy for bivalent ligands and provide insights into future opportunities in this promising field of drug discovery.
... Fig. 3b shows that UV irradiation for LUVs in the 'OFF' state led to a decrease in 31 P NMR resonances at d HPNP = −6.0 ppm and the appearance of new 31 P NMR resonances at d cyP = 14.5 ppm, demonstrating the generation of cyP and the formation of the 'ON' state. Additionally, the generation of PNP and cyP was conrmed by electrospray ionization mass spectra (ESI-MS). ...
... G-protein coupled receptors) usually correspond to the active and inactive states of signal transduction. 31 Therefore, achieving reversible control of articial systems by conformational conversion is of importance for understanding the natural signal transduction process. Herein, the 'ON/OFF' switches of the tAzo-LA-TACN/a-CD signaling system were also achieved by UV and visible light-controlled conformational conversion. ...
Article
Full-text available
Inspired by nature, it is of significant importance to design and construct biomimetic signaling systems to mimic natural signal transduction. Herein, we report an azobenzene/α-cyclodextrin (α-CD)-based signal transduction system with three functional modules: a light-responsive headgroup, lipid-anchored group, pro-catalyst tailgroup. The transducer can be inserted into the vesicular membrane to trigger the transmembrane translocation of molecules under the activation of light, forming a ribonuclease-like effector site and leading to the transphosphorylation of the RNA model substrate inside the vesicles. Moreover, the transphosphorylation process can be reversibly turned 'ON/OFF' over multiple cycles by the activation and deactivation of the pro-catalyst. This artificial photo-controlled signal transduction successfully constructs a signal responsive catalysis system across the membrane to utilize light to reversibly control the internal transphosphorylation process of an RNA model substrate, which might provide a new strategy for future design to utilize exogenous signals for implementing endogenous enzyme manipulation and gene regulation.
... To investigate this, the subunits of all eight mGluRs were specifically labeled by incorporating either the cyan (CFP) or the yellow (YFP) fluorescent protein in the intracellular i2 loop 35,36 and the receptors were co-expressed in Xenopus oocytes to high expression levels (Methods and Supplementary Fig. 3, Supplementary Data 6). To optimize the signal-to-noise ratio we used the GABA B -based quality control-system consisting of an ER-retention signal (GABA B1 ) and a masking sequence (GABA B2 ) at the C-terminus, as described previously [46][47][48][49] . This ensures that donor-containing constructs in the membrane are the desired dimers with the acceptor constructs by retaining donor/donor dimers (GABA B1 /GABA B1 ) in the ER, thereby generating also an enhanced apparent FRET-efficiency (Supplementary Fig. 4). ...
... To exclude that our results on heterodimers depend on the particular combination of the fluorescence label (CFP or YFP) and the GABA B sequence of the quality control system (C1 and C2) (see Methods) 49 , we tested for the examples mGluR1/5 and Fig. 12, Supplementary Data 6). A similar test was done also for dequenching analysis. ...
Article
Full-text available
Dimeric metabotropic glutamate receptors (mGluRs) are abundantly expressed in neurons. In mammals, eight subunit isoforms, mGluR1-8, have been identified, forming the groups I, II, and III. We investigated receptor dimerization and kinetics of these mGluR isoforms in excised membrane patches by FRET and confocal patch-clamp fluorometry. We show that 5 out of 8 homodimeric receptors develop characteristic glutamate-induced on- and off-kinetics, as do 11 out of 28 heterodimers. Glutamate-responsive heterodimers were identified within each group, between groups I and II as well as between groups II and III, but not between groups I and III. The glutamate-responsive heterodimers showed heterogeneous activation and deactivation kinetics. Interestingly, mGluR7, not generating a kinetic response in homodimers, showed fast on-kinetics in mGluR2/7 and mGluR3/7 while off-kinetics retained the speed of mGluR2 or mGluR3 respectively. In conclusion, glutamate-induced conformational changes in heterodimers appear within each group and between groups if one group II subunit is present.
... Studies on α 2A -AR/-opiod heterodimers as well as mGLuR1 homodimers suggest that only one of the protomers is activated (Hlavackova et al., 2005;Vilardaga et al., 2008). However, it appears clear now that activation of both protomers is required for full activation (Kniazeff et al., 2004;Grushevskyi et al., 2019). ...
Thesis
Full-text available
The CXC chemokine receptor 4 (CXCR4) and the atypical chemokine receptor 3 (ACKR3) are seven transmembrane receptors that are involved in numerous pathologies, including several types of cancers. Both receptors bind the same chemokine, CXCL12, leading to significantly different outcomes. While CXCR4 activation generally leads to canonical GPCR signaling, involving Gi proteins and β‐arrestins, ACKR3, which is predominantly found in intracellular vesicles, has been shown to signal via β‐arrestin‐dependent signaling pathways. Understanding the dynamics and kinetics of their activation in response to their ligands is of importance to understand how signaling proceeds via these two receptors. In this thesis, different Förster resonance energy transfer (FRET)‐based approaches have been combined to individually investigate the early events of their signaling cascades. In order to investigate receptor activation, intramolecular FRET sensors for CXCR4 and ACKR3 were developed by using the pair of fluorophores cyan fluorescence protein and fluorescence arsenical hairpin binder. The sensors, which exhibited similar functional properties to their wild‐type counterparts, allowed to monitor their ligand-induced conformational changes and represent the first RET‐based receptor sensors in the field of chemokine receptors. Additional FRET‐based settings were also established to investigate the coupling of receptors with G proteins, rearrangements within dimers, as well as G protein activation. On one hand, CXCR4 showed a complex activation mechanism in response to CXCL12 that involved rearrangements in the transmembrane domain of the receptor followed by rearrangements between the receptor and the G protein as well as rearrangements between CXCR4 protomers, suggesting a role of homodimers in the activation course of this receptor. This was followed by a prolonged activation of Gi proteins, but not Gq activation, via the axis CXCL12/CXCR4. In contrast, the structural rearrangements at each step of the signaling cascade in response to macrophage migration inhibitory factor (MIF) were dynamically and kinetically different and no Gi protein activation via this axis was detected. These findings suggest distinct mechanisms of action of CXCL12 and MIF on CXCR4 and provide evidence for a new type of sequential signaling events of a GPCR. Importantly, evidence in this work revealed that CXCR4 exhibits some degree of constitutive activity, a potentially important feature for drug development. On the other hand, by cotransfecting the ACKR3 sensor with K44A dynamin, it was possible to increase its presence in the plasma membrane and measure the ligand‐induced activation of this receptor. Different kinetics of ACKR3 activation were observed in response to CXCL12 and three other agonists by means of using the receptor sensor developed in this thesis, showing that it is a valuable tool to study the activation of this atypical receptor and pharmacologically characterize ligands. No CXCL12‐induced G protein activation via ACKR3 was observed even when the receptor was re-localized to the plasma membrane by means of using the mutant dynamin. Altogether, this thesis work provides the temporal resolution of signaling patterns of two chemokine receptors for the first time as well as valuable tools that can be applied to characterize their activation in response to pharmacologically relevant ligands.
... The large substituents at either side of antagonists physically prevent VFTD GB1 closure, which stabilizes VFTD GB1 in an open conformation (Geng et al. 2013). Conversely, agonists induce VFTD GB1 closure (Geng et al. 2012;Kniazeff et al. 2004). Mutations that stabilize VFTD GB1 closure therefore lead to constitutive activity. ...
Chapter
GABAB receptors (GBRs), the G protein-coupled receptors for the inhibitory neurotransmitter γ-aminobutyric acid (GABA), activate Go/i-type G proteins that regulate adenylyl cyclase, Ca²⁺ channels, and K⁺ channels. GBR signaling to enzymes and ion channels influences neuronal activity, plasticity processes, and network activity throughout the brain. GBRs are obligatory heterodimers composed of GB1a or GB1b subunits with a GB2 subunit. Heterodimeric GB1a/2 and GB1b/2 receptors represent functional units that associate in a modular fashion with regulatory, trafficking, and effector proteins to generate receptors with distinct physiological functions. This review summarizes current knowledge on the structure, organization, and functions of multi-protein GBR complexes.
... Likewise, initial studies of mGluRs indicated that only one of the two protomers becomes activated (Hlavackova et al., 2005). However, it now appears clear that activation of both protomers is required for full activation (Kniazeff et al., 2004;Grushevskyi et al., 2019). The CXC chemokine receptor (CXCR) 4 appears to be a facultative homodimer, and dimer formation may contribute to its signaling function. ...
Article
Full-text available
G protein–coupled receptors (GPCRs) are biologic switches that transduce extracellular stimuli into intracellular responses in the cell. Temporally resolving GPCR transduction pathways is key to understanding how cell signaling occurs. Here, we investigate the kinetics and dynamics of the activation and early signaling steps of the CXC chemokine receptor (CXCR) 4 in response to its natural ligands CXC chemokine ligand (CXCL) 12 and macrophage migration inhibitory factor (MIF), using Förster resonance energy transfer–based approaches. We show that CXCR4 presents a multifaceted response to CXCL12, with receptor activation (≈0.6 seconds) followed by a rearrangement in the receptor/G protein complex (≈1 seconds), a slower dimer rearrangement (≈1.7 seconds), and prolonged G protein activation (≈4 seconds). In comparison, MIF distinctly modulates every step of the transduction pathway, indicating distinct activation mechanisms and reflecting the different pharmacological properties of these two ligands. Our study also indicates that CXCR4 exhibits some degree of ligand-independent activity, a relevant feature for drug development. SIGNIFICANCE STATEMENT The CXC chemokine ligand (CXCL) 12/CXC chemokine receptor (CXCR) 4 axis represents a well-established therapeutic target for cancer treatment. We demonstrate that CXCR4 exhibits a multifaceted response that involves dynamic receptor dimer rearrangements and that is kinetically embedded between receptor–G protein complex rearrangements and G protein activation. The alternative endogenous ligand macrophage migration inhibitory factor behaves opposite to CXCL12 in each assay studied and does not lead to G protein activation. This detailed understanding of the receptor activation may aid in the development of more specific drugs against this target.
... Plasmids. The pRK5 plasmids encoding the wild type rat mGlu 2 , GABA B1 and GABA B2 receptors with HA-tag, Flag-tag and/or SNAP-tag inserted just after the signal peptide (HA-ST-mGlu 2 , HA-GABA B1a/b , HA-ST-GABA B1a/b , Flag-GABA B2 , Flag-ST-GABA B2 ) under control of the CMV promoter were previously described 16,20,55 . Plasmids for controlling the number of SNAP-tags per mGlu 2 dimer with and without the V699C mutation (mGlu 2 -C1 KKXX , ST-mGlu 2 -C2 KKXX , mGlu 2 -V699C-C1 KKXX , ST-mGlu 2 -V699C-C2 KKXX ) were previously described 20,22 . ...
Article
Full-text available
G protein coupled receptors (GPCRs) play essential roles in intercellular communication. Although reported two decades ago, the assembly of GPCRs into dimer and larger oligomers in their native environment is still a matter of intense debate. Here, using number and brightness analysis of fluorescently labeled receptors in cultured hippocampal neurons, we confirm that the metabotropic glutamate receptor type 2 (mGlu2) is a homodimer at expression levels in the physiological range, while heterodimeric GABAB receptors form larger complexes. Surprisingly, we observed the formation of larger mGlu2 oligomers upon both activation and inhibition of the receptor. Stabilizing the receptor in its inactive conformation using biochemical constraints also led to the observation of oligomers. Following our recent observation that mGlu receptors are in constant and rapid equilibrium between several states under basal conditions, we propose that this structural heterogeneity limits receptor oligomerization. Such assemblies are expected to stabilize either the active or the inactive state of the receptor.
Chapter
Synopsis The relationship between chemical structure and gustatory response is discussed in substantial detail for each of the primary tastes: sweet, bitter, umami, salty and sour. Structural formulas, empirical formulas, molecular weights, and gustatory potencies are provided for most stimuli. Chemical structure/gustatory response reviews are preceded by reviews of current understandings of the biochemical pathways which mediate these gustatory experiences. The relationship between chemical structure and sweet taste is very broad ranging from the common carbohydrates to synthetic sweeteners to natural non-caloric sweeteners and even to some minerals which exhibit sweet taste. The relationship between chemical structure and bitter taste is even more diverse covering essentially all structural classes of organic and inorganic compounds. On the other hand, structures of chemical compounds which exhibit umami taste are narrowly tuned around the structure of monosodium glutamate, the prototypical umami taste compound. This is also true for salty and sour tastes where chemical structures of compounds exhibiting these tastes are all salts and acids, respectively.
Chapter
The GABAB receptor is activated by the main inhibitory neurotransmitter of the central nervous system, the γ-aminobutyric acid (GABA). The receptor is expressed in almost all neuronal and glial cells and plays a central role in the modulation of many physiological and pathological processes. The GABAB receptor has been considered for years as an interesting target for the treatment of spasticity, pain, addiction, anxiety or depression. This has prompted many studies aiming at understanding the activation of the receptor and its modulation. While it belongs to the super-family of G protein-coupled receptors (GPCRs), it was rapidly evident that the GABAB receptor is peculiar in the variety of allosteric modulations governing its activation. Here, I wish to gather the different aspects of the GABAB receptor allosteric modulation. After presenting the main small molecule allosteric modulators known to date, the intramolecular transitions controlling the receptor activation will be summarized. In addition, recent findings obtained in the last decade on the existence of GABAB receptor complexes and their influence on the receptor function will be introduced, including the GABAB receptor oligomers and the auxiliary proteins associated with the receptor. These new concepts will certainly be of major interest in the future analysis of GABAB receptor allosteric modulation.
Article
The orphan G-protein-coupled receptor (GPCR) GPR158 is expressed in the brain, where it is involved in the osteocalcin effect on cognitive processes, and at the periphery, where it may contribute to glaucoma and cancers. GPR158 forms a complex with RGS7-β5, leading to the regulation of neighboring GPCR-induced Go protein activity. GPR158 also interacts with αo, although no canonical Go coupling has been reported. GPR158 displays three VCPWE motifs in its C-terminal domain that are putatively involved in G-protein regulation. Here, we addressed the scaffolding function of GPR158 and its VCPWE motifs on Go. We observed that GPR158 interacted with and stabilized the amount of RGS7-β5 through a 50-residue region downstream of its transmembrane domain and upstream of the VCPWE motifs. We show that two VCPWE motifs are involved in αo binding. Using a Gαo-βγ bioluminescence resonance energy transfer (BRET) sensor, we found that GPR158 decreases the BRET signal as observed upon G-protein activation; however, no constitutive activity of GPR158 could be detected through the measurement of various G-protein-mediated downstream responses. We propose that the effect of GPR158 on Go is unlikely due to a canonical activation of Go, but rather to the trapping of Gαo by the VCPWE motifs, possibly leading to its dissociation from βγ Such action of GPR158 is expected to prolong the βγ activity, as also observed with some activators of G-protein signaling. Taken together, our data revealed a complex functional scaffolding or signaling role for GPR158 controlling Go through an original mechanism.
Article
Full-text available
Recent studies on G-protein-coupled receptors revealed that they can dimerize. However, the role of each subunit in the activation process remains unclear. The -amino-n-butyric acid type B (GABAB) receptor is comprised of two subunits: GB1 and GB2. Both consist of an extracellular domain (ECD) and a heptahelical domain composed of seven transmembrane -helices, loops and the C-terminus (HD). Whereas GB1 ECD plays a critical role in ligand binding, GB2 is required not only to target GB1 subunit to the cell surface but also for receptor activation. Here, by analysing chimeric GB subunits, we show that only GB2 HD contains the determinants required for G-protein signalling. However, the HD of GB1 improves coupling efficacy. Conversely, although GB1 ECD is sufficient to bind GABAB ligands, the ECD of GB2 increases the agonist affinity on GB1, and is necessary for agonist activation of the receptor. These data indicate that multiple allosteric interactions between the two subunits are required for wild-type functioning of the GABAB receptor and highlight further the importance of the dimerization process in GPCR activation.
Article
A new method called the neighbor-joining method is proposed for reconstructing phylogenetic trees from evolutionary distance data. The principle of this method is to find pairs of operational taxonomic units (OTUs [= neighbors]) that minimize the total branch length at each stage of clustering of OTUs starting with a starlike tree. The branch lengths as well as the topology of a parsimonious tree can quickly be obtained by using this method. Using computer simulation, we studied the efficiency of this method in obtaining the correct unrooted tree in comparison with that of five other tree-making methods: the unweighted pair group method of analysis, Farris's method, Sattath and Tversky's method, Li's method, and Tateno et al.'s modified Farris method. The new, neighbor-joining method and Sattath and Tversky's method are shown to be generally better than the other methods.
Article
In a previous study we reported that the addition of a carboxylic group to the mGlu receptor agonist aminocyclopentane-1,3-dicarboxylate (ACPD) changes its properties from agonist to antagonist at both mGlu1 and mGlu2 receptors, and resulted in an increase in affinity at mGlu4 receptors, with isomers being either agonists or antagonists. In the present study, the effect of gamma-carboxy-L-glutamic acid (Gla) and (2S,2'R,3'R)-2-(2,3-dicarboxycyclopropyl)glycine (DCG-IV), two carboxylic derivatives of non-selective agonists, were examined on all cloned mGlu receptors. We found that this additional carboxylic group on glutamate prevents its interaction with group-I mGlu receptors and generates a potent group-II antagonist (K(B) = 55 microM on mGlu2). At group-III mGlu receptors, Gla was found to be either an antagonist (mGlu7 and mGlu8 receptors) or a partial agonist (mGlu4 and mGlu6 receptors). We show here that L-CCG-I is a general mGlu receptor agonist activating all cloned receptors. We also confirm that DCG-IV, which corresponds to L-CCG-I with an additional carboxylic group, is a selective group-II agonist. However, this additional COOH group changes the properties of L-CCG-I from an agonist to an antagonist at all group-III receptors, making this compound one of the most potent group-III mGlu receptor antagonist known so far. These observations will be useful for the development of more potent and selective mGlu receptor agonists and antagonists.
Article
The recently published high-resolution crystal structures of the amino-terminal domains (ATDs) of the metabotropic glutamate 1 (mglu1) receptor homodimer present an exciting milestone in the study of the molecular pharmacology of family C G-protein-coupled receptors (GPCRs). In this article, we outline recent developments in the understanding of signal transduction by family C GPCR homodimers, with particular emphasis on the conformational movements of the two ATDs, in addition to allosteric modulation and competitive and noncompetitive antagonism of these processes.
Article
Comparative protein modeling is increasingly gaining interest since it is of great assistance during the rational design of mutagenesis experiments. The availability of this method, and the resulting models, has however been restricted by the availability of expensive computer hardware and software. To overcome these limitations, we have developed an environment for comparative protein modeling that consists of SWISS-MODEL, a server for automated comparative protein modeling and of the SWISS-PdbViewer, a sequence to structure workbench. The Swiss-PdbViewer not only acts as a client for SWISS-MODEL, but also provides a large selection of structure analysis and display tools. In addition, we provide the SWISS-MODEL Repository, a database containing more than 3500 automatically generated protein models. By making such tools freely available to the scientific community, we hope to increase the use of protein structures and models in the process of experiment design.