Download A Molecular Model of Agonist and Nonpeptide Antagonist Binding to

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

5-Hydroxyeicosatetraenoic acid wikipedia , lookup

Hepoxilin wikipedia , lookup

Purinergic signalling wikipedia , lookup

NMDA receptor wikipedia , lookup

Cooperative binding wikipedia , lookup

G protein–coupled receptor wikipedia , lookup

Signal transduction wikipedia , lookup

Cannabinoid receptor type 1 wikipedia , lookup

Transcript
0022-3565/00/2941-0195$03.00/0
THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS
Copyright © 2000 by The American Society for Pharmacology and Experimental Therapeutics
JPET 294:195–203, 2000 /2366/831458
Vol. 294, No. 1
Printed in U.S.A.
A Molecular Model of Agonist and Nonpeptide Antagonist
Binding to the Human V1 Vascular Vasopressin Receptor1
MARC THIBONNIER, PATRICK COLES, DOREEN M. CONARTY, CHRISTINE L. PLESNICHER, and
MENACHEM SHOHAM
Departments of Medicine (M.T., D.M.C., C.L.M.) and Biochemistry (P.C., M.S.), Case Western Reserve University School of Medicine,
Cleveland, Ohio
Accepted for publication March 14, 2000
This paper is available online at http://www.jpet.org
The neurohypophysial hormone arginine vasopressin
(AVP) is a cyclic nonapeptide (Fig. 1) whose actions are
mediated by stimulation of specific G protein-coupled receptors (GPCRs) currently classified into V1 vascular (V1R), V2
renal (V2R), and V3 pituitary (V3R) AVP receptors (Thibonnier et al., 1998b). AVP is involved in numerous physiological
functions, including the regulation of body fluid osmolality,
blood volume, vascular tone, and blood pressure (Thibonnier,
1993). In addition, AVP belongs to the family of vasoactive
and mitogenic peptides involved in physiological and pathological cell growth and differentiation (Van Biesen et al.,
1996).
The various members of the family of human and animal
AVP-oxytocin (OT) receptors have been cloned (Birnbaumer
et al., 1992; Kimura et al., 1992; Lolait et al., 1992; Morel et
al., 1992; Gorbulev et al., 1993; De Keyser et al., 1994; Mahlmann et al., 1994; Sugimoto et al., 1994; Thibonnier et al.,
Received for publication December 8, 1999.
1
This work was supported in part by National Institutes of Health Grants
RO1-HL39757 and PO1-HL41618. P.C. was supported by an Undergraduate
Research Experience supplement to Grant MCB97-28420 from the National
Science Foundation awarded to M.S.
whereas the polar part is located on the surface of the extracellular side. The increased affinity of the G337A mutant is due
to two additional van der Waals contacts of the alanine methyl
group with carbon atoms on the antagonist. The I310V mutant
reduces the hydrophobicity in the vicinity of the polar oxygen
atom of the antagonist. The I224V mutant relieves overcrowding in a hydrophobic binding pocket involving the aromatic
residues Trp175, Phe179, Phe307, and Trp304. Finally, the E324D
mutant enables the formation of a hydrogen bond of the carboxylate side chain with the amide side chain of Gln311, which
in turn forms a hydrogen bond with the N57 nitrogen atom of
OPC-21268. Thus, a few residues, distinct from those involved
in agonist binding, control interspecies selectivity toward OPC21268 nonpeptide antagonist binding.
1994; Hutchins et al., 1995). Stable expression of these cloned
receptors in immortalized cell lines now allows the detailed
characterization of the ligand-binding pocket and the signal
transduction pathways coupled to a given AVP-OT receptor
subtype, without the possible interference from other receptor subtypes and endogenously bound hormone. Such characterization will facilitate the rational design of potent and
selective therapeutic agents.
The combination of receptor three-dimensional modeling
and site-directed mutagenesis experiments has suggested
that the AVP agonist binding domain is made of a 15- to
20-Å-deep central cavity defined by the transmembrane helices and surrounded by the extracellular loops of the receptor (Mouillac et al., 1995; Hibert et al., 1999). As shown for
other families of GPCRs, residues that are critical for peptide
agonist binding are not involved in antagonist binding to the
AVP-OT receptors. Furthermore, the determinants of nonpeptide AVP receptor antagonist binding were unknown before this work.
The first nonpeptide AVP V1R antagonist found by random
screening and optimization of chemical entities (Yamamura
et al., 1991), OPC-21268 (Fig. 1), has an excellent affinity for
ABBREVIATIONS: AVP, arginine vasopressin; OT, oxytocin; V1R, V1 vascular vasopressin receptor; V2R, V2 renal vasopressin receptor; V3R, V3
pituitary vasopressin receptor; GPCR, G protein-coupled receptor; CHO, Chinese hamster ovary; TMS, transmembrane segment.
195
Downloaded from jpet.aspetjournals.org at ASPET Journals on August 11, 2017
ABSTRACT
The affinity of the nonpeptide antagonist OPC-21268 is greater
for the rat V1 arginine vasopressin (AVP) receptor (V1R) than for
the human V1R. Site-specific mutagenesis was carried out to
identify the residues that determine interspecies selectivity for
nonpeptide antagonist binding. The introduction of rat amino
acids in position 224, 310, 324, or 337 of the human V1R
sequence dramatically altered OPC-21268 affinity for the receptor, whereas binding of AVP, the peptide V1R antagonist
d(CH2)5Tyr(Me)AVP, and the nonpeptide V1R antagonist
SR49059 was not altered by these mutations. Computer modeling explained the mutagenesis results. Docking of
OPC-21268 onto a homology-built model of the V1R receptor
yielded a model for the bound ligand in which the hydrophobic
part is deeply embedded in the transmembrane region,
196
Thibonnier et al.
Vol. 294
Fig. 1. Chemical structure of AVP,
the peptide V1R antagonist d(CH2)5
[Tyr(Me)2]AVP, and the nonpeptide
V1R antagonists OPC-21268 and SR49059.
Experimental Procedures
Materials. Standard reagents, unless stated otherwise, were purchased from Sigma Chemical Co. (St. Louis, MO). CHO-K1 cells were
obtained from American Type Culture Collection (Rockville, MD).
Cell culture media and geneticin were purchased from Life Technologies (Grand Island, NY). Fetal bovine serum was obtained from
Hyclone (Logan, UT). Restriction and modification enzymes were
obtained from Promega (Madison, WI). [3H]AVP (specific activity,
68.5 Ci/mmol) was obtained from DuPont-New England Nuclear
(Wilmington, DE). The XL2-Blue Escherichia coli strain was purchased from Stratagene (La Jolla, CA). The expression vector
pcDNA3.1 was purchased from Invitrogen (San Diego, CA). AVP and
the peptide V1R antagonist d(CH2)5Tyr(Me)AVP were purchased
from Bachem California (Torrance, CA). The nonpeptide V1R antagonist SR-49059 (batch no. MY10-075) was provided by Dr. C. Serradeil-Le Gal (Sanofi Recherché, Toulouse, France). The nonpeptide
V1R antagonist OPC-21268 (batch no. 93F92 M) was provided by Dr.
J. F. Liard (Otsuka America Pharmaceutical, Inc., Rockville, MD).
Site-Directed Mutagenesis of Human V1R. The human V1R
cDNA clone was isolated by screening a human liver cDNA library as
described previously (Thibonnier et al., 1994) and inserted into the
pcDNA3.1 expression vector. The mutations were introduced in the
human V1R wild-type sequence by using the Quickchange mutagenesis kit from Stratagene according to the manufacturer’s recommendations. The presence of the mutations was verified by doublestranded DNA sequencing with the Taq Dye Deoxy Terminator cycle
sequencing kit and a model 373A sequencer from Applied Biosystems
(Foster City, CA).
Stable transfection of CHO cells with the pcDNA3.1 expression
vector containing the sequence of the wild-type or mutated human
V1R cDNA clones was performed using the calcium phosphate precipitation method. Cells were grown in medium F12 supplemented
with 10% fetal calf serum, selected with the neomycin analog geneticin, and purified by the limiting dilution technique. Clones expressing high and comparable levels of AVP receptors were studied by
radioligand saturation and competition binding experiments as described later.
Radioligand Binding Assays in Intact Cells. Transfected
CHO cells were grown to confluence in 24-well dishes and washed
twice with PBS plus 10 mM MgCl2 and 0.2% BSA, pH 7.4. Saturation
binding experiments of AVP receptors of transfected CHO cells were
performed in 24-well dishes in duplicate with increasing concentrations of [3H]AVP with or without 1 ␮M unlabeled AVP (Thibonnier et
al., 1994). Affinity (Kd) and capacity (Bmax) values of the AVP receptors were calculated by a nonlinear least-squares analysis program.
Protein concentration was measured with BCA reagent (Pierce
Chemical Co., Rockford, IL) using albumin as an internal standard.
Competition binding experiments were performed as described previously (Thibonnier et al., 1994, 1998b) using one fixed concentration
of [3H]AVP and increasing concentrations of unlabeled peptide and
nonpeptide AVP analogs (n ⫽ 3– 8 for each analog) for 30 min at
30°C. IC50 values were derived from nonlinear least-squares analysis, and Ki values were calculated with the equation of Cheng and
Prusoff: Ki ⫽ IC50/(1 ⫹ Lf/Kd).
Three-Dimensional Molecular Modeling of V1R. Very little
direct structural information is available for GPCRs, and for many
years, molecular models of these receptors have been built based on
the crystal structure of bacteriorhodopsin. Although bacteriorhodopsin consists of the seven transmembrane helical domains by which
GPCRs are characterized, it shares very little sequence homology
with any of the GPCRs. Still, the use of bacteriorhodopsin to establish the orientation of the transmembrane domains of the V1R is the
only way to build a model based on an experimentally determined
high-resolution structure (Henderson et al., 1990). Coordinates of
bovine rhodopsin are also available but only for the seventh TMS
without any loops. As a basis for our model building of the V1R
receptor, we used a model of the seventh TMS of V1R generated by G.
Vriend with the program WHATIF (Vriend, 1990) based on the
crystal structure of bacteriorhodopsin (G Protein-Coupled Receptor
Data Base at http://swift.embl-heidelberg.de/7tm/htmls/consortium.
html; Rodriguez et al., 1998).
Downloaded from jpet.aspetjournals.org at ASPET Journals on August 11, 2017
the rat V1R (25 nM) but a poor affinity for the human V1R
(8800 nM) (Thibonnier et al., 1998a). The human and rat
V1Rs share a high degree of structural homology with 96%
sequence identity. The differing residues are presumably
involved in species-related variations in antagonist binding.
Comparison of the human and rat V1R sequences revealed
that only 20 amino acid differences are present in the extracellular loops and the upper portions of the transmembrane
segments (TMSs; see Fig. 3). We reasoned that these interspecies differences in amino acid sequence modulate the receptor affinity for nonpeptide compounds. In this work, we
produced a series of reverse mutations in which corresponding rat amino acids were introduced by site-directed mutagenesis into the human V1R sequence. The influence of
these interspecies amino acid differences on nonpeptide antagonist binding was subsequently tested. A single amino
acid substitution in the seventh TMS produced a 27-fold
increase in the affinity toward OPC-21268. To gain information about the location of the OPC-21268 binding site, a
model of this compound was docked onto a homology-built
three-dimensional model of the human V1R. The hydrophobic
moieties of this nonpeptide antagonist were found to be located deep within the transmembrane region, whereas the
polar part is on the extracellular surface. This model of the
ligand-receptor complex is consistent with the mutagenesis
results and provides an explanation for the increased affinity
of the mutants tested in this study.
2000
197
model of AVP assumed a type I ␤-turn structure, containing a hydrogen bond between the carbonyl oxygen of Tyr2 and the amide
proton of Asn5 (Fig. 2A).
A model of the nonpeptide V1R antagonist OPC-21268 was constructed with the program Alchemy 2000 (Fig. 2B; Tripos Inc., St.
Louis, MO). This compound was first drawn in two dimensions and
then extended into a three-dimensional model by a two-dimensionalto-three-dimensional builder incorporated in Alchemy 2000. Three
possible rotations, each differing by 120°, of the bond between the
lactam nitrogen and the piperidyl ring were considered. Each rotamer was subjected to an energy minimization, and the most stable of
the three rotamers was chosen. A conformational search was performed on the resulting structure, systematically stepping through
all possible rotations of the bonds of the piperidyl ring and of the
bond connecting this ring to the lactam nitrogen. A rotational increment of 3° was set for the bonds in the ring, whereas an increment of
30° was used for the bond connecting the ring to the lactam nitrogen.
Conformations with the lowest energy and devoid of any short contacts were saved. Three of the most stable conformations were subjected to yet another energy minimization, as were the corresponding
conformations with a 180° rotation about the amide bond of the
piperidyl nitrogen. Finally, the most stable conformation of these six
was subjected to an optimization using the program Gaussian 98
(Gaussian, Inc., Pittsburgh, PA). A similar strategy was used to build
a model of the nonpeptide V1R antagonist SR-49059 (Fig. 2C).
Docking of AVP and Antagonist Ligands onto V1R. Docking
of the ligands was done with the program LIGIN (Sobolev et al.,
1996), based on a built-in complementarity function. This function is
Fig. 2. Model of 8-AVP (A) and the nonpeptide V1R antagonists OPC-21268 (B) and SR49059 (C) in ball-and-stick representation. Note the disulfide
bridge between cysteines 1 and 6 of AVP.
Downloaded from jpet.aspetjournals.org at ASPET Journals on August 11, 2017
The three extracellular and three intracellular loops of the V1R
were subsequently constructed with program Look v3.5 (Molecular
Applications Group, Palo Alto, CA), using the spatial constraints for
the ends of each loop provided by the coordinates of the helical
bundle. Look v3.5 is a protein-modeling program that segmentally
builds a protein by aligning short stretches of its sequence with
homologous peptides of known structure and performs a full energy
refinement of the model (Levitt, 1992). Because the N- and C-terminal domains of the V1R are not involved in the binding of agonists or
antagonists, they were not included in this model (Mouillac et al.,
1995; Howl and Wheatley, 1996; Mendre et al., 1997; Thibonnier et
al., 2000).
A disulfide bridge exists between cysteines 124 and 203 located on
the second and third extracellular loops, respectively. Disruption of
this disulfide bridge is known to cause a significant drop in binding
affinity of ligands (Mouillac et al., 1995; Postina et al., 1996). Thus,
it was necessary to ascertain that these cysteine residues were close
enough in the model and that the sulfhydryl groups had the proper
orientation to be able to form the disulfide bridge. This was achieved
by performing an energy refinement in program X-PLOR (Brünger,
1993) with the constraint of forming this particular disulfide bridge.
The sulfur-sulfur distance refined to a value of 2.03 Å, consistent
with the formation of a disulfide bridge. The remainder of the structure was not significantly altered by this refinement procedure.
Molecular Modeling of AVP and Antagonist Ligands. Using
program Look v3.5, a model of 8-AVP was built based on the structure of OT obtained from the crystallographically determined structure of the neurophysin-OT complex (Fig. 1; Rose et al., 1996). This
Molecular Model of Vasopressin Receptor
198
Thibonnier et al.
Results
Radioligand Binding Characteristics of Wild-Type
and Mutated Human V1Rs. The amino acid differences
between human and rat V1Rs are presented in Fig. 3. Mutations that altered the charge or shape were produced by
introducing corresponding rat amino acid residues into the
wild-type human V1R sequence. Because mutations located
into the first two extracellular loops do not affect the affinity
of antagonists (Mouillac et al., 1995), we centered our attention on interspecies amino acid differences present in the
other components of the ligand binding pocket. An extensive
ligand binding characterization of these mutated human
AVP receptors was carried out in stably transfected Chinese
hamster ovary (CHO) cells. As shown previously, CHO cells
do not express endogenous AVP-OT receptors (Thibonnier et
al., 1994), and each clone tested in our experiments expressed a single AVP receptor clone.
The wild-type human V1R expressed in CHO cells displayed a high affinity not only for the endogenous hormone
AVP, but also for the reference V1R peptide antagonist
d(CH2)5Tyr(Me)AVP (“Maurice Manning’s V1R antagonist”)
and the V1R nonpeptide antagonist SR-49059 (Table 1). As
we observed previously, the affinity of the wild-type human
V1R for the OPC-21268 compound was quite weak.
None of the nine single mutations, two double mutations,
and one triple mutation engineered in this study interfered
with the normal folding of the V1R within the plasma membrane, and the levels of expression of all of the mutated
clones were similar to that of the wild-type human V1R (Bmax
⫽ 11,000 –25,000 fmol/mg of protein).
For all of these mutated human V1Rs, affinity for the
natural hormone AVP, for the V1R peptide antagonist
d(CH2)5Tyr(Me)AVP, and for the V1R nonpeptide antagonist
SR-49059 remained in a close range (Ki ⫽ 0.26 –1.75 nM;
Fig. 3. Amino acid differences between the human and the rat V1Rs.
The primary sequence and the putative transmembrane segments of the
human V1R are shown. The putative
ligand binding pocket is boxed by the
rectangle. Arrows pointing toward the
corresponding residues of the rat sequence mark nonconserved residues
between the human and rat V1R sequences.
Downloaded from jpet.aspetjournals.org at ASPET Journals on August 11, 2017
a sum of the surface areas of atomic contacts. These contacts are
weighted according to the types of atoms in contact, and another
term is included to prevent short contacts. After maximizing the
complementarity function, LIGIN optimizes the lengths of possible
hydrogen bonds. To take into account possible movements of the
receptor on ligand binding, steric overlap between the ligand and a
specified number of residues in the receptor can be allowed without
energy penalty.
The model of AVP was docked onto V1R by initially placing it in
the upper portion of the transmembrane region (the expected binding pocket) and allowing LIGIN to search for the binding site within
a 20- ⫻ 20- ⫻ 20-Å box around the original ligand position. A similar
procedure was used for the docking of the antagonist OPC-21268. In
the docking of both AVP and the antagonist, some steric overlap (one
to three residues) was allowed between the ligand and receptor.
Energy minimization with program X-PLOR relieved these short
contacts.
Molecular Modeling of Site-Directed Mutagenesis. After
docking the model of OPC-21268 onto wild-type V1R, the receptorligand complex was subjected to an energy refinement using program X-PLOR. Interactions between this antagonist and four residues on the receptor (Ile224, Ile310, Glu324, and Gly337) were analyzed.
Based on the mutagenesis results, mutations of these four residues
to Val224, Val310, Asp324, and Ala337 were modeled in the program O
(Jones et al., 1991). Interactions between the antagonist and the
mutated residues, as well as any other residues in close contact to
the ligand, were analyzed.
Data Analysis. Nucleotide and amino acid sequences were analyzed with the computer package MacVector (Oxford Molecular, Oxford, UK) on a Macintosh G3 computer. Binding parameters (Kd and
Bmax) of AVP receptors were calculated by a nonlinear least-squares
analysis program using the software package Kaleidagraph (Synergy
Software, Reading PA; Thibonnier and Roberts, 1985). Data were
expressed as mean ⫾ S.E. Statistical analysis was performed with
Kruskal-Wallis and Mann-Whitney nonparametric tests (StatView
statistical package; Abacus Concepts, Berkeley, CA). P ⬍ .05 was
considered statistically significant.
Vol. 294
2000
Molecular Model of Vasopressin Receptor
199
TABLE 1
Affinity of AVP and receptor antagonists for the wild-type and mutated forms of the human V1Rs stably expressed in CHO cells
The affinity constants (Ki) of the different compounds were determined in competition binding assays with [3H]AVP and increasing concentrations of unlabeled compounds.
Each value is the mean of four to eight independent experiments.
Compound
Mutation
AVP
d(CH2)5Tyr(Me)AVP
1.73 ⫾ 0.12
0.29 ⫾ 0.04*
0.48 ⫾ 0.02
0.63 ⫾ 0.03
0.52 ⫾ 0.03
0.56 ⫾ 0.01
0.87 ⫾ 0.04
0.73 ⫾ 0.05
0.49 ⫾ 0.05
0.95 ⫾ 0.05
0.95 ⫾ 0.05
1.45 ⫾ 0.03
0.62 ⫾ 0.04
0.98 ⫾ 0.12
1.59 ⫾ 0.05
0.66 ⫾ 0.02
0.59 ⫾ 0.10
1.21 ⫾ 0.07
0.52 ⫾ 0.03
0.54 ⫾ 0.01
0.86 ⫾ 0.04
0.36 ⫾ 0.05
0.48 ⫾ 0.02
1.36 ⫾ 0.13
1.75 ⫾ 0.11
1.27 ⫾ 0.02
1.25 ⫾ 0.03
0.26 ⫾ 0.01*
SR49059
OPC21268
1.06 ⫾ 0.08
0.46 ⫾ 0.01
0.79 ⫾ 0.04
0.79 ⫾ 0.02
0.34 ⫾ 0.01*
0.46 ⫾ 0.02
0.68 ⫾ 0.02
0.59 ⫾ 0.13
0.63 ⫾ 0.06
0.57 ⫾ 0.07
1.01 ⫾ 0.11
0.52 ⫾ 0.05
0.49 ⫾ 0.02
0.27 ⫾ 0.03*
8800 ⫾ 1200
5046 ⫾ 470
4764 ⫾ 398
1242 ⫾ 51*
3865 ⫾ 304*
5239 ⫾ 305
4780 ⫾ 451
4755 ⫾ 366
2655 ⫾ 204*
328 ⫾ 19*
185 ⫾ 25*
20 ⫾ 1*
29 ⫾ 2*
28 ⫾ 3*
nM
* P ⬍ .05 compared with the affinity of the wild-type receptor for the same ligand.
Table 1). These fluctuations are presumably not relevant
from a pharmacological viewpoint. At variance, significant
improvement in the human V1R affinity for OPC-21268 was
obtained by introducing single mutations in positions 224 in
the fifth TMS (7-fold improvement), 324 in the third extracellular loop (3-fold improvement), and 337 in the seventh
TMS (27-fold improvement). Simultaneous mutations of
E324D and G337A or I224V and G337A produced dramatic
improvement of the human V1R affinity for the OPC-21268
compound (303-/440-fold improvement), similar to the values
encountered for the rat V1R: 20 and 29 nM, respectively. The
combination of the three mutations G337A ⫹ E324D ⫹ I224V
did not further improve the affinity for OPC-21268.
Docking of Hormone AVP onto Human V1R. AVP has
a polar as well as a nonpolar surface. The exocyclic tripeptide
Pro7-Arg8-Gly9 and one side of the hormone ring (Gln4, Asn5)
are mainly hydrophilic, whereas the other part of the ring
(Cys1, Cys6, Tyr2, and Phe3) is essentially hydrophobic. This
dual surface property is reflected in the nature of the binding
pocket that is formed by residues from TMSs 1, 3, 4, 5, 6, and
7, as well as the first extracellular loop (Fig. 4). The bottom of
the cleft is mainly hydrophobic, closed by the aromatic and
hydrophobic residues Met135, Phe136, Phe179, Phe307, and
Ile330. The entrance to the binding pocket and one side of it
contain predominantly hydrophilic residues. The Arg8 guanido group at the entrance to the cleft forms a salt bridge
with Asp112 located on the first extracellular loop. Trp111
forms van der Waals contacts with the hydrophobic part of
Arg8. The ⑀-amino group of Lys128 forms a hydrogen bond to
the amide side chain nitrogen of Asn5. Other hydrogen bonds
are formed between the side chain moieties of Gln185 and
Ser182 with Gln4 and of Ser213 O␥ with Tyr2 OH. Another
wall of the pocket is lined with the hydrophobic residues Ile55
and Ile330.
Docking of Nonpeptide Antagonist OPC-21268 onto
Human V1R. The location of the bound antagonist OPC21268 is distinct from the AVP-binding pocket with only
partial overlap near the extracellular surface (Fig. 5). The
hydrophobic part is embedded in the transmembrane region
far deeper than AVP, whereas the polar part is located on the
surface of the extracellular side. The binding pocket is
Fig. 4. Docking of the hormone 8-AVP onto the model of the human V1R.
AVP and interacting receptor residues are shown in ball-and-stick representation. Residue numbers for AVP are in superscript. A salt bridge
between Arg8 of AVP and Asp112 of the receptor is shown by a broken line.
Two hydrogen bonds, one between Asn5 of AVP and Lys128 and the other
between Gln4 of AVP and Gln185 of the receptor, are shown by broken
lines. Distances (in angstroms) are indicated near the broken lines. The
secondary structure assignment of the interacting receptor residues is
shown by small tube representations of helical segments H1, H4, H6, and
H7, as well as extracellular loop 1 (el1). Met135, Phe136, Ser182, and Ser213
also interact with AVP. They are not shown here for reasons of clarity.
The former two residues are within van der Waals contact of Phe3,
whereas the latter two form hydrogen bonds with the amide side chain
nitrogen atom of Gln4 and the hydroxyl group of Tyr2, respectively.
formed by residues from TMSs 4, 5, 6, and 7, as well as the
third extracellular loop (Fig. 6). The 27-fold increase in the
affinity of the G337A mutant is explained by the formation of
two van der Waals contacts of the methyl carbon with carbon
atoms C22 and C28 of the bicyclic ring structure of OPC21268 at the bottom of the cleft (Fig. 7). The E324D mutant
Downloaded from jpet.aspetjournals.org at ASPET Journals on August 11, 2017
Wild type
G134A
G222S
I224V
I310V
P318G
M319N
S320F
E324D
G337A
G337A ⫹ I310V
G337A ⫹ E324D
G337A ⫹ I224V
G337A ⫹ E324D ⫹
I224V
200
Thibonnier et al.
Vol. 294
has an indirect effect. It enables the formation of a hydrogen
bond of the carboxylate side chain with the amide side chain
atom of Gln311. This causes a polarization of this amide
nitrogen atom and enables it in turn to form another hydrogen bond to the N57 nitrogen atom of OPC-21268 (Fig. 7).
The I310V mutant reduces the hydrophobicity in the vicinity
of the polar oxygen atom of the antagonist. The I224V mutant relieves overcrowding in a hydrophobic binding site involving the aromatic residues Trp175, Phe179, Phe307, and
Trp304. The smaller valine side chain allows for better positioning of the aromatic residues to interact with the bicyclic
ring structure of OPC-21268 (Fig. 8). Finally, the I310V
mutant reduces the hydrophobicity in the vicinity of the polar
oxygen atom of the antagonist. Thus, the model explains all
of the mutations that significantly increase the affinity toward OPC-21268.
Discussion
AVP receptors represent a logical target for drug development in several therapeutic fields. As a new class of therapeutic agents, orally active AVP analogs could be used in
several pathophysiological conditions. V2R agonists increase
the reabsorption of free water in central diabetes insipidus.
V1R antagonists could reduce the systemic vascular resistances noted in arterial hypertension, congestive heart failure, and peripheral arteriopathy. V2R antagonists could re-
verse the hyponatremia of Schwartz-Bartter syndromes,
congestive heart failure, and liver cirrhosis. Mixed V1/V2R
antagonists may prevent thromboembolic events in surgical
patients. V3R agonists and antagonists could be valuable
additions to the diagnosis, imaging, localization, and medical
treatment of adrenocorticotropic hormone-secreting tumors.
Finally, OT receptor antagonists could be used in the treatment of primary dysmenorrhea and premature labor (Thibonnier, 1998).
Three different strategies can be contemplated to develop
ligands with high affinity and selectivity for a given AVP
receptor subtype: 1) the systematic or rationale alterations of
the ligand structure, implemented by Maurice Manning and
collaborators who designed numerous peptide AVP and OT
analogs (Manning et al., 1995); 2) the random screening for
new chemical compounds, developed by pharmaceutical companies who isolated the first nonpeptide V1R and V2R antagonists (Yamamura et al., 1991, 1992; Serradeil-Le Gal et al.,
1993, 1996); and 3) structure-based drug design, requiring
the knowledge of the three-dimensional structure of both the
ligand and receptor. The AVP-OT receptors crystallographic
structure has yet to be established. However, modeling by
analogy based on the structure of bacteriorhodopsin has been
done for the seven TMSs of many GPCRs and has yielded
useful information (Ji et al., 1998).
These three strategies are complementary. For instance,
Downloaded from jpet.aspetjournals.org at ASPET Journals on August 11, 2017
Fig. 5. Superposition of the models of AVP and the nonpeptide antagonist OPC-21268 as bound to the human V1R. AVP and OPC-21268 in
ball-and-stick representation (A) and with the receptor shown in ribbons (B). The loops are labeled il1, il2, and il3 for the intracellular loops and el1,
el2, and el3 for the extracellular loops. The different binding modes of agonist and antagonist are clearly shown.
2000
Molecular Model of Vasopressin Receptor
201
conformational energy calculations carried out on three nonpeptide AVP-OT antagonists (OPC-21268, OPC-31260, and
penicilide) found that the affinity of these compounds and
their selectivity for AVP and OT receptors are probably connected with mimicking the aromatic rings of the Tyr2 and the
Ile3 OT residues or with mimicking the aromatic rings of the
Tyr2 and Phe3 AVP residues (Oldziej et al., 1995). Similarly,
this study illustrates that strategies 2 and 3 are indeed
complementary.
By random screening and subsequent optimization of
chemical entities, nonpeptide compounds were recently
shown to antagonize AVP receptors (Yamamura et al., 1991,
1992; Serradeil-Le Gal et al., 1993, 1996). They specifically
antagonize the V1R or the V2R and have different chemical
structures. The first AVP receptor antagonist OPC-21268
was found to be a potent entity in rat models but was subsequently found to display a poor affinity for human AVP
receptors (Thibonnier et al., 1998b). To expand our understanding of the molecular characteristics of the ligand-binding pocket of AVP receptors, we used the amino acid differences among mammalian species to search for the rat versus
human molecular determinants of nonpeptide V1R binding.
Our data confirm that the molecular determinants of agonist and antagonist binding as well as peptide versus nonpeptide compounds are distinct (Mouillac et al., 1995). Amino
acid residues that are important for peptide agonist binding
are not critical determinants in binding of the cyclic peptide
d(CH2)5Tyr(Me)AVP, of the linear peptide antagonist pheny-
lacetyl1-D-Tyr(Me)2-Phe3-Gln4-Asn5-Arg6-Pro7-Arg8-NH2,
and of the nonpeptide V1R antagonist SR49059 (Mouillac et
al., 1995). Similarly, the molecular determinants of peptide
antagonist binding to the OT receptor are different from
those involved in peptide agonist binding; they are TMSs 1, 2,
and especially 7. The introduction of just seven amino acids
of the upper part of the seventh TMS of the OT receptor into
the V2R sequence is sufficient to introduce high-affinity binding for an OT peptide antagonist into the V2R.
All point mutations affecting peptide agonist binding to
AVP receptors were found to have no or little effect on peptide antagonist binding, thus suggesting that peptide agonist
and antagonist binding requirements are physically distinct
(Phalipou et al., 1997). So far, there is little information
about the molecular determinants of nonpeptide antagonists
binding to AVP-OT receptors besides the fact that they are
different from those involved in peptide agonist and antagonist binding. Cotte et al. (1998) found that residues 202 in the
second extracellular loop and 304 in the seventh TMS of the
V2R which modulated species selectivity of cyclic peptide
antagonists containing a D-isoleucyl at position 2, did not
contribute to binding of nonpeptide antagonists OPC-31260
and SR-121463A.
The combination of site-directed mutagenesis and threedimensional modeling in our study identified key residues
involved in binding of the nonpeptide antagonist OPC-21268
to the V1R. Our data clearly identified a single residue in the
seventh TMS, explaining the different affinities of the human
Downloaded from jpet.aspetjournals.org at ASPET Journals on August 11, 2017
Fig. 6. Docking of the nonpeptide antagonist OPC-21268 onto the model of the human V1-vascular AVP receptor. A, side view. B, top view. The receptor
and the antagonist are shown in ribbon and ball-and-stick representation, respectively.
202
Thibonnier et al.
Vol. 294
Fig. 7. The stabilizing effect of the G337A and E324D mutations on
antagonist binding. Portions of helices 6 and 7 as well as of the extracellular loop 3 (el3) are shown in tube representation; the antagonist as well
as residues 311, 324, and 337 of the receptor are shown in ball-and-stick
representation. Distances are indicated in angstroms. The 27-fold increase in affinity of this glycine-to-alanine mutant can be explained by
the formation of two new van der Waals contacts of the methyl group with
the antagonist. The glutamic acid-to-aspratic acid muation at position
324 has an indirect effect. It enables the formation of a hydrogen-bond
between an oxygen atom of the carboxylate and the amide side chain
nitrogen atom of Gln311. This has a polarizing effect on this nitrogen atom
that in turn stabilizes another hydrogen bond of this atom with atom N57
of OPC-21268.
and rat V1Rs for OPC-21268. The docking model developed
for this study confirmed the importance of this single residue:
Ala337. Furthermore, the model predicts that a serine residue
at this position should cause an even tighter binding due to
the formation of a hydrogen bong between the serine O␥ atom
with the quinoline oxygen atom of OPC-21268, in addition to
the van der Waals interaction of the serine ␤-carbon with
carbon atoms 22 and 28 of this antagonist. This study also
suggests modifications to the antagonist to increase the affinity for the receptor. For example, elimination of the quinoline oxygen atom should stabilize the interactions with the
hydrophobic pocket deep inside the transmembrane region.
However, this may cause adverse solubility problems. A similar situation exists for residue 310 of the receptor and oxy-
gen 47 of the antagonist. A hydrophobic residue in the vicinity of this polar atom is clearly unfavorable. A valine at this
position, as found in the human sequence, is better than an
isoleucine, the corresponding rat residue, but a threonine
would be even better. Alternatively, replacement of oxygen
47 of the antagonist with a carbon atom should also increase
the affinity. With respect to residue 224, a valine at this
position seems to be optimal. This residue is located in a
rather crowded hydrophobic environment into which a valine
seems to fit better than the bulkier isoleucine.
Combination of the three mutations in positions 224, 324,
and 337 did not further improve the affinity of the V1R for
OPC-21268 compared with the two double mutations, thus
suggesting that alterations of the structure of the nonpeptide
antagonist will be required to further increase the affinity of
this compound.
The field of GPCRs has a lack of experimentally determined structures. Therefore, molecular modeling is a very
useful tool to derive structural information for the V1R. It
provides a framework to design and test new drugs, as well
as site-specific mutations, in a rational way. However, one
must keep in mind the limitations of molecular modeling.
The approach is based on the assumption that the seven
TMSs are similar in structure to bacteriorhodopsin. The
Achilles’ heels of this approach are the loops connecting the
helical regions as well as the N- and C-terminal nonhelical
segments. The former were built by sequence similarity to
known protein segments from a database within the program
LOOK, whereas the N- and C-terminal stretches were left
Downloaded from jpet.aspetjournals.org at ASPET Journals on August 11, 2017
Fig. 8. The stabilizing effect of the G337A, I224V, and I310V mutations
on antagonist binding. Portions of helices 4, 5, 6, and 7 are shown in tube
representation; the antagonist as well as selected residues of the receptor
are shown in ball-and-stick representation. Distances are indicated in
angstroms. The isoleucine-to-valine substitution at position 310 puts a
less hydrophobic residue in the vicinity of the polar O47 oxygen atom of
the OPC-21268 antagonist compound. This should have a slightly stabilizing effect, and it could explain the 2.3-fold increase in affinity. Residue
224 is located deep in the transmembrane region in a very crowded
environment, which constitutes a hydrophobic binding pocket for the
antagonist. The pocket consists of residues Trp175, Phe179, Phe307, and
Trp304 in addition to residue 224. Substitution of the smaller valine for
the larger isoleucine relieves overcrowding and allows for better positioning of the aromatic residues to interact with the bicyclic ring structure of
OPC-21268. This could explain the 7.1-fold increase in affinity for this
mutant.
2000
out altogether from the model because they are not involved
in ligand binding. The validity of the model is supported by
the experimentally determined affinities for the drugs. The
model explains very well all of our findings. It does not prove
that the model is correct, but the model is certainly consistent with the data, and it provides a tool for designing new
drugs and mutants.
In conclusion, this study provides for the first time the
structural basis of species-selective binding of a nonpeptide
antagonist to the V1R. These findings should generate new
ideas for drug development of nonpeptide AVP receptor antagonists and for optimizing drug-receptor interactions.
Acknowledgment
The program LIGIN was kindly provided by Vladmir Sobolev from
the Weizmann Institute of Science in Rehovot, Israel.
References
203
Mendre C, Dufour MN, Le Roux S, Seyer R, Guillou L, Calas B and Guillon G (1997)
Synthetic rat V1a vasopressin receptor fragments interfere with vasopressin binding via specific interaction with the receptor. J Biol Chem 272:21027–21036.
Morel A, O’Carroll AM, Brownstein MJ and Lolait SJ (1992) Molecular cloning and
expression of a rat V1a arginine vasopressin receptor. Nature (Lond) 356:523–526.
Mouillac B, Chini B, Balestre MN, Elands J, Trumpp-Kallmeyer S, Hoflack J, Hibert
M, Jard S and Barberis C (1995) The binding site of neuropeptide vasopressin V1a
receptor. J Biol Chem 270:25771–25777.
Oldziej S, Ciarkowski J, Liwo A, Shenderovich MD and Grzonka Z (1995) Conformational aspects of differences in requirements for oxytocin and vasopressin
receptors. J Recept Signal Transduct Res 15:703–713.
Phalipou S, Cotte N, Carnazzi E, Seyer R, Mahe E, Jard D, Barberis C and Mouillac
B (1997) Mapping peptide-binding domains of the human V1a vasopressin receptor
with a photoactivable linear peptide antagonist. J Biol Chem 272:26536 –26544.
Postina R, Kojro E and Fahrenholz F (1996) Separate agonist and peptide antagonist
binding sites of the oxytocin receptor defined by their transfer into the V2 vasopressin receptor. J Biol Chem 271:31593–31601.
Rodriguez R, Chinea G, Lopez N, Pons T and Vriend G (1998) Homology modelling,
model and software evaluation: Three related sources. CABIOS 14:523–528.
Rose JP, Wu CK, Hsiao CD, Breslow E and Wand BC (1996) Crystal structure of the
neurophysin-oxytocin complex. Nat Struct Biol 3:163–169.
Serradeil-Le Gal C, Lacour C, Valette G, Garcia G, Foulon L, Galindo G, Bankir L,
Pouzet B, Guillon G and Barberis C (1996) Characterization of SR-121463A, a
highly potent and selective, orally active vasopressin V2 receptor antagonist.
J Clin Invest 98:2729 –2738.
Serradeil-Le Gal C, Wagnon J, Guillon G, Garcia C, Lacour C, Cantau B, Guiraudou
P, Christophe B, Barberis C, Villanova G, Nisato D, Maffrand JP, Jard S and Le
Fur G (1993) Biochemical and pharmacological properties of SR49059, a new
potent, non-peptide antagonist of rat and human vasopressin V1a receptors. J Clin
Invest 92:224 –231.
Sobolev V, Wade RC, Vriend G and Edelman M (1996) Molecular docking using
surface complementarity. Prot Struct Funct Genet 25:120 –129.
Sugimoto T, Saito M, Mochizuki S, Watanabe Y, Hashimoto S and Kawashima H
(1994) Molecular cloning and functional expression of a cDNA encoding the human
V1b vasopressin receptor. J Biol Chem 269:27088 –27092.
Thibonnier M (1993) Antidiuretic hormone: Regulation, disorders, and clinical evaluation in neuroendocrinology, in Concepts in Neurosurgery (Selman W ed) pp
19 –30 Williams & Wilkins, Baltimore.
Thibonnier M (1998) Development and therapeutic indications of orally-active nonpeptide vasopressin receptor antagonists. Exp Opin Invest Drugs 7:729 –740.
Thibonnier M, Auzan C, Wilkins P, Berti-Mattera L, Madhun Z and Clauser E (1994)
Cloning, sequencing, and functional expression of the cDNA coding for the human
V1a vasopressin receptor. J Biol Chem 269:3304 –3310.
Thibonnier M, Berti-Mattera LN, Dulin N, Conarty DM and Mattera R (1998a)
Signal transduction pathways of the human V1-vascular, V2-renal, V3-pituitary
AVP, and oxytocin receptors, in Progress in Brain Research (Urban IJA, Burbach
JPH and De Wied D eds) pp 143–158, Elsevier Science BV, Amsterdam.
Thibonnier M, Conarty DM, Preston JA, Wilkins PL, Berti-Mattera LN and Mattera
R (1998b) Molecular pharmacology of human vasopressin receptors, in Vasopressin
and Oxytocin: Molecular, Cellular, and Clinical Advances (Zingg H, Bourque CW
and Bichet D eds) pp 251–276, Plenum Press, New York.
Thibonnier M, Preston J, Conarty D, Plesnicher C and Berti-Mattera L (2000) Role
of the C-terminus region of the human V1-vascular vasopressin receptor in signal
transduction (Abstract). Experimental Biology 99 meeting; 1999 April 17–21;
Washington, DC, in press.
Thibonnier M and Roberts JM (1985) Characterization of human platelet vasopressin receptors. J Clin Invest 76:1857–1864.
Van Biesen T, Luttrell LM, Hawes BE and Lefkowitz RJ (1996) Mitogenic signaling
via G protein-coupled receptors. Endocr Rev 17:698 –714.
Vriend G (1990) A molecular modeling and drug design program. J Mol Graph
8:52–56.
Yamamura Y, Ogawa H, Chihara T, Kondo K, Ongawa T, Nakamura S, Mori T,
Tominaga M and Yabuuchi Y (1991) OPC-21268, an orally effective nonpeptide
vasopressin V1 receptor antagonist. Science (Wash DC) 252:572–574.
Yamamura Y, Ogawa H and Yamashita H (1992) Characterization of a novel
aquaretic agent, OPC-31260, as an orally effectuve, nonpeptide vasopressin V2receptor antagonist. Br J Pharmacol 105:787–792.
Send reprint requests to: Dr. Marc Thibonnier, Room BRB431, Division of
Clinical and Molecular Endocrinology, Department of Medicine, Case Western
Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH
44106-4951. E-mail: [email protected]
Downloaded from jpet.aspetjournals.org at ASPET Journals on August 11, 2017
Birnbaumer M, Seibold A, Gilbert S, Ishido M, Barberis C, Antaramian A, Brabet P
and Rosenthal W (1992) Molecular cloning of the receptor for human antidiuretic
hormone. Nature (Lond) 357:333–335.
Brünger AT (1993) X-PLOR, Version 3.1 Manual: A System for X-Ray Crystallography and NMR. Yale University Press, New Haven, CT.
Cotte N, Balestre MN, Phalipou S, Hibert M, Manning M, Barberis C and Mouillac
B (1998) Identification of residues responsible for the selective binding of peptide
antagonists and agonists in the V2 vasopressin receptor. J Biol Chem 273:29462–
29468.
De Keyser Y, Auzan C, Lenne F, Beldjord C, Thibonnier M, Bertagna X and Clauser
E (1994) Molecular cloning, sequencing, and functional expression of a cDNA
encoding the human V3-pituitary vasopressin receptor. FEBS Lett 356:215–220.
Gorbulev V, Büchner H, Akhundova A and Fahrenholz F (1993) Molecular cloning
and functional characterization of V2[8-lysine]vasopressin and oxytocin receptors
from a pig kidney cell line. Eur J Biochem 215:1–7.
Henderson R, Baldwin JM, Ceska TA, Zemlin F, Beckmann E and Downing KH
(1990) Model for the structure of bacteriorhodopsin based on high-resolution
electron cryo-microscopy. J Mol Biol 213:899 –929.
Hibert M, Hoflack J, Trumpp-Kallmeyer S, Mouillac B, Chini B, Mahé E, Cotte N,
Jard S, Manning M and Barberis C (1999) Functional architecture of vasopressin/
oxytocin receptors. J Recept Signal Transduct Res 19:589 –596.
Howl J and Wheatley M (1996) Molecular recognition of peptide and non-peptide
ligands by the extracellular domains of neurohypophysial hormone receptors.
Biochem J 317:577–582.
Hutchins AM, Phillips PA, Venter DJ, Burrell LM and Johnston CI (1995) Molecular
cloning and sequencing of the gene encoding a sheep arginine vasopressin type 1a
receptor. Biochem Biophys Acta 1263:266 –270.
Ji TH, Grossmann M and Ji I (1998) G protein-coupled receptors. J Biol Chem
273:17299 –17302.
Jones TA, Zou JY, Cowan SW and Kjeldgaard M (1991) Improved methods for
building protein models in electron density maps and the location of errors in these
models. Acta Crystallogr A 47:110 –119.
Kimura T, Tanizawa O, Mori K, Brownstein MJ and Okayama H (1992) Structure
and expression of a human oxytocin receptor. Nature (Lond) 356:526 –529.
Levitt M (1992) Accurate modeling of protein conformation by automatic segment
matching. J Mol Biol 226:507–533.
Lolait SJ, O’Carroll AM, McBride OW, Konig M, Morel A and Brownstein MJ (1992)
Cloning and characterization of a vasopressin V2 receptor and possible link to
nephrogenic diabetes insipidus. Nature (Lond) 357:336 –339.
Mahlmann S, Meyerhof W, Hausmann H, Heierhorst J, Schönrock C, Zwiers H,
Lederis K and Richter D (1994) Structure, function, and phylogeny of
[Arg8]vasotocin receptors from teleost fish and toad. Proc Natl Acad Sci USA
91:1342–1345.
Manning M, Cheng LL, Stoev S, Sawyer WH, Tribollet E, Barberis C, Wo NC and
Chan WY (1995) Novel potent and selective antagonists and radioiodinated ligands for oxytocin and vasopressin receptors, in Neurohypophysis: Recent Progress
of Vasopressin and Oxytocin Research (Saito T, Kurokawa K and Yoshida S eds) pp
21–38, Elsevier, Amsterdam.
Molecular Model of Vasopressin Receptor