SlideShare a Scribd company logo
1 of 59
1521-0103/346/2/318–327$25.00
http://dx.doi.org/10.1124/jpet.113.202994
THE JOURNAL OF PHARMACOLOGY AND
EXPERIMENTAL THERAPEUTICS J Pharmacol Exp Ther
346:318–327, August 2013
Copyright ª 2013 by The American Society for Pharmacology
and Experimental Therapeutics
Selective and Potent Agonists and Antagonists for Investigating
the Role of Mouse Oxytocin Receptors
Marta Busnelli, Elisabetta Bulgheroni, Maurice Manning,
Gunnar Kleinau, and Bice Chini
CNR, Institute of Neuroscience, Milan, Italy (M.B., E.B., B.C.);
Department of Biotechnology and Translational Medicine,
Università degli Studi di Milano, Milan, Italy (M.B.);
Department of Biochemistry and Cancer Biology, University of
Toledo,
Toledo, Ohio (M.M.); and Institute of Experimental Pediatric
Endocrinology, Charité-Universitätsmedizin Berlin, Berlin,
Germany
(G.K.)
Received January 8, 2013; accepted May 29, 2013
ABSTRACT
The neuropeptides oxytocin (OT) and vasopressin (AVP) have
been shown to play a central role in social behaviors; as
a consequence, they have been recognized as potential drugs to
treat neurodevelopmental and psychiatric disorders character -
ized by impaired social interactions. However, despite the basic
and preclinical relevance of mouse strains carrying genetic
alterations in the OT/AVP systems to basic and preclinical
translational neuroscience, the pharmacological profile of
mouse
OT/AVP receptor subtypes has not been fully characterized. To
fill in this gap, we have characterized a number of OT and AVP
agonists and antagonists at three murine OT/AVP receptors
expressed in the nervous system as follows: the oxytocin
(mOTR) and vasopressin V1a (mV1aR) and V1b (mV1bR)
subtypes. These three receptors were transiently expressed in
vitro for binding and intracellular signaling assays, and then
a homology model of the mOTR structure was constructed to
investigate how its molecular features compare with human
and rat OTR orthologs. Our data indicate that the selectivity
profile of the natural ligands, OT and AVP, is conserved in
humans, rats, and mice. Furthermore, we found that the
synthetic peptide [Thr4Gly7]OT (TGOT) is remarkably
selective
for the mOTR and, like the endogenous OT ligand, activates Gq
and Gi and recruits b-arrestins. Finally, we report three
antagonists that exhibit remarkably high affinities and selectiv-
ities at mOTRs. These highly selective pharmacological tools
will contribute to the investigation of the specific physiologic
and pathologic roles of mOTR for the development of selective
OT-based therapeutics.
Introduction
Central oxytocin (OT) and vasopressin (AVP) effects are
mediated by three G-protein-coupled receptors (GPCRs) that
are evolutionarily highly conserved and closely related, with
overall homology varying from 40 to 85%: the vasopressin 1a
receptor (V1aR), the vasopressin 1b receptor (V1bR), and the
OT receptor (OTR) (Barberis et al., 1999; Birnbaumer, 2000;
Zingg and Laporte, 2003). OT and AVP are also structurally
very similar, differing by only two amino acids in most
mammals (Wallis, 2012). Given this high degree of conserva-
tion in both receptors and peptides, the development of
selective agonists and antagonists has proved to be a daunting
task (Manning et al., 2008). Over the last decades, at least
1000 synthetic peptides have been synthesi zed and examined
for their ability to bind to and activate the different OT/AVP
receptor subtypes, an effort that has led to the identification of
a number of subtype-selective analogs for human and rat
subtypes (Manning et al., 2012). However, subtle differences
between receptor sequences in different animal species are
responsible for important changes in the selectivity profile of
some ligands, and so the pharmacological data obtained in one
species cannot be extrapolated tout court to others (Chini and
Manning, 2007).
In addition to their well established systemic physiologic
effects, OT and AVP are potent modulators of social behavior;
consistently, the OT/AVP system is emerging as a relevant
target for the treatment of impaired social functions associ-
ated with neurodevelopmental and psychiatric disorders
(Miller, 2013). Even as OT has been recently reported to
improve cognitive deficits in autistic patients (Modi and
Young, 2012), its use is hampered by several of the following
factors: 1) OT does not cross the blood-brain barrier and is
administered intranasally, with unknown pharmacokinetics;
2) it also binds to and activates the V1aR and V1bR, which are
highly expressed in the brain, where they exert different and
even opposite effects (Pittman and Spencer, 2005); and 3) OT
promotes the coupling of the OTR to different G proteins and
This study was supported by Cariplo Foundation (Grant
2008.2314),
Regione Lombardia (Progetto TerDisMental, ID 16983–Rif.
SAL-50), and
Telethon Foundation (Grant GGP12207).
dx.doi.org/10.1124/jpet.113.202994.
ABBREVIATIONS: AVP, vasopressin; BRET, bioluminescence
resonance energy transfer; ECL, extracellular loop; GPCR, G-
protein-coupled
receptor; HTRF, homogeneous time-resolved fluorescence; ICL,
intracellular loop; IP1, myoinositol 1-phosphate; LVA, linear
vasopressin
antagonist; OT, oxytocin; OTA, oxytocin receptor antagonist;
OTR, oxytocin receptor; PBS, phosphate-buffered saline; RLuc,
Renilla luciferase;
SR49059, (2S)1-[(2R,3S)-1,5-chloro-3-(2-chlorophenyl)-1-(3,4-
dimethoxybenzene-sulfonyl)-3-hydroxy-2,3-dihydro-1H-indole-
2-carbonyl]-pyrroli-
dine-2-carboxamide; t1/2, half-time; TGOT, [Thr
4Gly7]OT; TMH, transmembrane helix; V1aR, vasopressin 1a
receptor; V1bR, vasopressin 1b
receptor; YFP, yellow fluorescent protein.
318
http://dx.doi.org/10.1124/jpet.113.202994
http://dx.doi.org/10.1124/jpet.113.202994
b-arrestins (Busnelli et al., 2012) and, consequently, activates
multiple signaling pathways whose precise roles within the
brain
are currently unknown. The development of more selective,
potent, and therefore, longer-lasting analogs acting on brain is
a priority in the field to understand how (in terms of biologic
mechanisms) and where (in terms of neural specificity) OT
exerts
its effects.
In particular, subtype-selective analogs are crucial for
defining the role of different receptor subtypes in rodent
models currently used to investigate the OT/AVP system in
the processing of socially relevant clues. Studies using trans -
genic mice genetically engineered to eliminate OT, OTR, or
CD38, a protein involved in OT secretion, show that these
animals lose important social behaviors (Ferguson et al., 2000;
Takayanagi et al., 2005; Jin et al., 2007; Higashida et al., 2011).
The deficits in the social paradigm can be fully restored by
a single intracerebroventricular infusion of OT given prior to
the test (Ferguson et al., 2001; Jin et al., 2007; Sala et al.,
2011). Although this effect is consistent with the genetic
alteration in CD38- and OT-null mice, which have a decreased
level of circulating (and centrally released) OT, the efficacy of
OT in restoring social recognition in the OTR-null mice is
particularly intriguing. We have recently shown that the social
behavioral deficits associated with the complete loss of Oxtr
gene expression can be rescued by the activation of cognate
vasopressin receptors, thus suggesting that the OT/AVP brain
systems have overlapping and/or compensatory functions (Sala
et al., 2011).
Another level of complexity in developing selective analogs
derives from the finding that a single GPCR may couple to more
than one G-protein, potentially activating multiple responses.
Interestingly, different ligands show different degrees of in-
trinsic efficacy to different signaling pathways activated by the
same receptor, a phenomenon referred to as “functional se-
lectivity” (Urban et al., 2007; Kenakin, 2011). Because func-
tional selective ligands have been recently described in the OT/
AVP receptor family (in particular for the vasopressin 2
receptor
(Jean-Alphonse et al., 2009), OTR (Reversi et al., 2005; Gravati
et al., 2010; Busnelli et al., 2012), and V1aR (MacKinnon et al.,
2009), the screening of the functional selective properties of
ligands is becoming a crucial issue for the pharmacological
characterization of selective ligands.
The aim of this study was to pharmacologically characterize
a number of OT/AVP analogs at the OT/AVP receptor sub-
types expressed in mouse brain: mOTR, mV1aR, and mV1bR.
We found that [Thr4Gly7]OT (TGOT) (Lowbridge et al., 1977)
has a remarkable selectivity for the mouse OTR through
which, like the endogenous OT ligand, it activates Gq and Gi
and recruits b-arrestins. Furthermore, we identified several
antagonists that exhibit remarkable selectivity profiles at
mOTR. These analogs represent valuable tools to investigate
the specific role of the mOTRs in the brain.
Materials and Methods
Reagents, Constructs, and Peptides. [3H]OT (NET-858, 30–60
Ci/mmol) and [3H]AVP (NET-800, 35–85 Ci/mmol) came from
PerkinElmer Life and Analytical Sciences (Monza, Italy);
coelenter-
azine H from Invitrogen (Milan, Italy); DeepBlueC
coelenterazine
400a (CLz400) from Biotium (Hayward, CA); OT, AVP, TGOT,
and
atosiban from Bachem (Weil am Rhein, Germany); and SR49059
[(2S)1-
[(2R,3S)-1,5-chloro-3-(2-chlorophenyl)-1-(3,4-
dimethoxybenzene-
sulfonyl)-3-hydroxy-2,3-dihydro-1H-indole-2-carbonyl]-
pyrrolidine-2-
carboxamide] from sanofi-aventis (Toulouse, France). All of the
other
peptides used in this study were from the laboratory of M.
Manning of
the University of Toledo (Toledo, OH).
The mouse V1aR cDNA (mV1aR) came from OriGene
(Rockville,
MD); the mouse V1bR (mV1bR) cDNA was a gift from M.A.
Ventura
(U-567; INSERM, Paris, France).
The GFP10-Gg2 cDNA, in which the blue-shifted variant of
Aequorea victoria (GFP10) was fused to Gg2, and the Gb1
cDNA are
described in Gales et al. (2006). The Ga subunit expression
vector
cDNAs came from Missouri S&T cDNA Resource Center
(Rolla, MO).
The expression vector of b-arrestin2 fused at its C terminus to
the
yellow fluorescent protein (b-arrestin2–YFP) (originally
developed in
M. Bouvier’s laboratory) came from Dr. J. Perroy (IGF,
Montpellier,
France), and the expression vector for b-arrestin1–YFP came
from
C. Hoffmann of the University of Wuerzburg (Wuerzburg,
Germany).
The mOTR C-terminally fused to Renilla luciferase (mOTR-
Rluc) was
generated by subcloning the entire coding region of mOTR into
an
Rluc vector (PerkinElmer BioSignal, Inc., Monza, Italy).
Cell Cultures. HEK293 and COS7 cells purchased from the
American Type Culture Collection (Manassas, VA) were grown
in
Dulbecco’s modified Eagle’s medium (Sigma-Aldrich, Milan,
Italy),
supplemented with 10% fetal calf serum and 1% penicillin-
streptomycin (Sigma-Aldrich) in a 10% CO2 humidified
atmosphere
at 37°C.
Transfection. For the ligand binding assays, the COS7 cells
were
transfected by means of electroporation as previously described
(Chini et al., 1995). For the homogeneous time-resolved
fluorescence
(HTRF) and bioluminescence resonance energy transfer (BRET)
assays, HEK293 cells were seeded at a density of 3,100,000
cells/
well in 100-mm plates on the day before transfection. A mix
containing 20 mg of DNA and 60 mg of polyethylenimine (PEI
linear,
MW 25000; Polysciences Europe GmbH, Eppelheim, Germany)
was
prepared with 1 ml of basic medium (without additives such as
serum
or antibiotics) and, after 15 minutes of incubation at room tem-
perature, added directly to cells maintained in 10 ml of
complete
medium containing 10% fetal bovine serum. For the HTRF
experi-
ments, the cells were detached 24 hours after transfection and
seeded
with 100,000 cells/well in white half-area 96-well microplates
(Corning Life Sciences, Amsterdam, The Netherlands). For the
BRET
experiments, the supplemented Dulbecco’s modified Eagle’s
medium
was renewed 24 hours after the transfections, and the cells were
maintained in culture for an additional 24 hours before being
washed
twice, detached, and resuspended with phosphate-buffered
saline
(PBS)-MgCl2 (0.5 mM) at room temperature.
Ligand Binding Assays. The binding assays were performed at
30°C on membranes prepared from COS7 cells as previously
described (Chini et al., 1995). Compound affinities were
determined
by means of competition experiments in which the unlabeled
compound concentrations varied from 10211 to 1025 M, and the
concentration of the radioligand ([3H]OT for mOTR and
[3H]AVP for
mV1aR and mV1bR) was 2–4 � 1029 M. Nonspecific binding
was
determined in the presence of unlabeled OT or AVP (1023 M).
The
ligand binding data were analyzed by means of nonlinear
regression
using Prism version 5 (GraphPad Software, Inc., La Jolla, CA).
The Ki
values were calculated from the experimental IC50 values using
the
Cheng-Prusoff equation for a single population of competitive
sites:
Ki 5 IC50/[1 1 (L/Kd)], where L is the concentration of
radioligand
used in each experiment and the Kd values were as previously
reported: OT Kd 5 0.54 nM for mOTR (Ring et al., 2010), AVP
Kd 5 1.3
nM for mV1aR (Oshikawa et al., 2004), and AVP Kd 5 0.67 nM
for
mV1bR (Serradeil-Le Gal et al., 2007). All of the assays were
performed in triplicate and repeated at least three times.
BRET Assay. The interactions between mOTR and the different
Ga subunits were analyzed by means of BRET2 experiments that
use
RLuc as the donor, the DeepBlueC coelenterazine derivative as
its
substrate, and GFP10 as the acceptor. HEK293 cells were
cotrans-
fected with mOTR-Rluc, GFP10-Gg2, or Gb1, without (-Ga) or
with one
Identification of Selective and Potent Analogs of Mouse OTR
319
of Gaq, Gai1, Gai2, Gai3, Gas, or Gao. Cells were incubated for
2 minutes
with OT, TGOT, and PBS (untreated cells) before the addition
of Rluc
substrate, DeepBlueC; BRET2 was measured immediately after
using
an Infinite F500 reader plate (Tecan, Milan, Italy) that allows
the
sequential integration of light signals detected with two filter
settings
(Rluc, 370–450 nm; GFP10 filter, 510–540 nm). The BRET2
signal was
calculated as the ratio between GFP10 emission and the light
emitted
by Rluc. The positive changes in BRET induced by the ligands
indicated the closest interaction between the donor and the
acceptor
and were expressed on graphs as “BRET ligand effect” using the
formula: (emission GFP10 ligand/emission Rluc ligand) 2
(emission
GFP10 PBS/emission Rluc PBS).
To analyze the kinetics of the mOTR–b-arrestin interactions,
BRET1 experiments that use RLuc as the donor, coelenterazine
H as
its substrate, and YFP as the acceptor were performed. HEK293
cells
were cotransfected with mOTR-Rluc and b-arrestin2–YFP or b-
arrest-
in1–YFP. The transfected cells were distributed in a white 96-
well
microplate (100 mg of proteins per well) (Optiplate;
PerkinElmer Life
and Analytical Sciences) and incubated in the presence or
absence of
ligands. Coelenterazine H was added 8 minutes before the
addition of
the different ligands, and readings were made for 10 minutes
using an
Infinite F500 reader plate (Tecan) and filter set (Rluc filter,
370–480
nm; YFP filter, 520–570 nm). To determine the half-time (t1/2)
of OT-
and other ligand-induced BRET, the data were recorded as the
difference between the ligand-promoted BRET signal and the
average
of the baseline (PBS-treated) BRET signal, and the time at
which the
half-BRET peak was reached was estimated.
Inositol Phosphate Measurements. Myoinositol 1-phosphate
(IP1) accumulation in HEK293 cells transiently transfected with
mOTR, mV1aR, and mV1bR (100,000 cells) was determined in
96-well
half-area microplates (Corning Life Sciences) using the HTRF-
IP-One
Kit (CisBio International, Bagnols-sur-Cèze, France) after 1
hour of
stimulation with increasing concentrations of OT, AVP, and
TGOT at
37°C. The time-resolved fluorescence resonance energy transfer
signals were measured 50 ms after excitation at 620 and 665 nm
using a Tecan Infinite F500 instrument. The IP1 concentrations
were
interpolated from the IP1 standard curve supplied with the kit.
Statistical Analysis. All of the data were analyzed using Graph-
Pad Prism software, version 5. Data from radioligand binding
were
evaluated by the nonlinear, least-squares curve-fitting
procedure.
Concentration-response IP1 curves were analyzed by means of
nonlinear curve fitting using the sigmoidal dose-response
equation.
Parameters errors (Ki and EC50) are all expressed in %CV and
cal-
culated by simultaneous analysis of at least three different
experi-
ments performed in triplicate. Ki comparison has been
performed on
the basis of the F test for the extra sum of squares principle (*P
,
0.05; **P , 0.01; ***P , 0.001). Ligand-induced BRET ratios are
expressed as mean 6 S.E.M and were analyzed with one-way
analysis
of variance followed by Tukey’s post hoc test to determine
statistically
significant differences in treatments (***P , 0.001). The BRET1
kinetics data were normalized by setting the zero time point
immediately after the addition of the ligand, and the data were
analyzed by means of nonlinear least-squares fitting to the one-
phase
exponential association equation.
Homology Modeling of the mOTR Structure. A large number
of GPCR crystal structures in different activity-state-related
con-
formations have been published in recent years (Zhao and Wu,
2012),
most of them cocrystallized with specific ligands (agonists or
antagonists) (Kobilka and Schertler, 2008; Hanson and Stevens,
2009). Therefore, they serve as optimal templates for family A
GPCR
homology modeling (OTRs are members of family A GPCRs)
with the
purpose to study potential details of ligand binding or signal
transduction.
Based on high sequence similarity and overlapping structural
features in the transmembrane helices (TMHs), the b2-
adrenergic
receptor crystal structure in an active conformation was used
here as
a template (Protein Data Bank code 3SN6) (Rasmussen et al.,
2011)
for modeling of the murine OTR. The general modeling
procedure
(sequence alignment, crystal structure preparation for modeling,
side
chain substitutions) was performed as recently described
(Costanzi,
2012).
In addition, extracellular loop (ECL) 2 of the b2-adrenergic
receptor
was partially deleted, because of significant differences in
length and
amino acid composition (biophysical properties) compared with
the
sequence of mOTR ECL2. According to advanced insights from
previous GPCR modeling studies [supplemental material in
Michino
et al. (2009)], only the C-terminal part of the template ECL2
structure,
which includes the highly conserved cysteine bridge to TMH3,
was
kept. Other mOTR ECL2 residues, most likely not involved in
direct
constitution of the ligand binding region, were manually added
as
spacers. The putative general OTR ligand binding region,
located
between the extracellular ends of the TMHs and the ECLs, was
defined
based on similarity to the ligand binding regions of known
GPCR
crystal structures (Deupi and Standfuss, 2011; Kratochwil et al.,
2011;
Wichard et al., 2011; Jacobson and Costanzi, 2012).
The OTR model is constituted by amino acids from positions
Glu36
to Leu344 (the extracellular N terminus and intracellular C
terminus
are not included because of missing structural templates). For
modeling procedures, the Sybyl-X 2.0 version (Tripos
International,
St. Louis, MO) was used. Gaps of missing residues in the loops
of the
template structure were closed manually by adding OTR-
specific
amino acids. Side chains were subjected to conjugate gradient
minimizations (until converging at a termination gradient of 0.1
kcal/mol/Å) and molecular dynamics simulation (2 ns) by fixing
the
backbone of the TMHs. Finally, the model was minimized
without
constraints using the AMBER 7 forcefield. Structure images
were
produced using the PyMOL software (PyMOL Molecular
Graphics
System, version 1.3; Schrödinger, LLC, Portland, OR).
Results
Binding Affinities of Commonly Used OT/AVP Ana-
logs at Mouse Receptors. The ligand binding properties of
commonly used OT/AVP analogs at mOTR, mV1aR, and
mV1bR were determined by competition experiments; calcu-
lated Ki values 6 %CV are reported in Table 1.
With regard to the peptides with agonist activity, the
endogenous OT and AVP ligands had very different selectivity
profiles (Fig. 1, A and B). AVP bound to the three brain-
expressed OT/AVP receptors with almost identical affinity (Ki
values for OTR, V1aR, and V1bR of, respectively, 0.87 nM 6
8% CV, n 5 3; 1.11 nM 6 27% CV, n 5 4; and 0.43 nM 6 12%
CV, n 5 4), whereas OT had a receptor-specific affinity range
that was highest for OTR (Ki 5 0.83 nM 6 17% CV, n 5 4) and
lower for V1aR (Ki 5 20.38 6 26% CV, n 5 5) (P , 0.001
versus mOTR) and V1bR (Ki 5 36.32 nM 6 7% CV, n 5 4) (P ,
0.001 versus mOTR). The dLVT peptide agonist binds with
significantly different Ki values for OTR, V1aR, and V1bR of,
respectively, 0.43 nM 6 20% CV, n 5 5; 3.39 nM 6 28% CV,
n 5 5 (P , 0.001 versus mOTR); and 0.82 nM 6 7% CV, n 5 3
(P , 0.01 versus mOTR) (Fig. 1C). However, we should mention
here that a significant difference in Ki is not sufficient to
define ligand selectivity. As an operational criterion, it has
been proposed that, to be “selective” for a particular subtype,
any ligand should display a Ki at least 2 orders of magni tude
lower than that for the other receptor subtypes (Chini et al.,
2008). On these premises, the only agonist provided with
a good selectivity profile is TGOT, with a Ki of 0.04 nM 6 32%
CV, n 5 5, for OTR and Ki values of .1000 nM for V1aR and
V1bR (Fig. 1D).
Among the Gq OTR antagonists (OTAs) that we analyzed,
whose original synthesis and pharmacological properties are
320 Busnelli et al.
http://www.pdb.org/pdb/explore/explore.do?structureId=3SN6
reviewed in Manning et al. (2008, 2012), atosiban, OTA2, and
OTA3 were selective for mOTR (Fig. 2, A, C, and D), with
affinities in the nanomolar range (Ki 5 1.29 nM 6 46% CV,
n 5 4 for atosiban; Ki 5 0.27 nM 6 25% CV, n 5 4 for OTA2;
and Ki 5 1.24 nM 6 36% CV, n 5 4 for OTA3), and showing Ki
values for V1aR and V1bR that were .1000-fold higher (P ,
0.001). The widely used OTR antagonist OTA1 also had a good
selectivity profile (Fig. 2B), with highest affinity for OTR (Ki 5
0.13 nM 6 42% CV, n 5 5), intermediate for V1aR (Ki 5 34.3
nM 6 33% CV, n 5 4) (P , 0.001), and lowest for V1bR (Ki 5
374 nM 6 20% CV, n 5 5) (P , 0.001).
We finally analyzed three compounds commonly used as
V1aR-selective antagonists: LVA (linear vasopressin antago-
nist), the Manning compound, and the nonpeptidic antagonist
SR49059 [reviewed in Manning et al. (2008, 2012)]. As shown
in Fig. 3, A–C, they bound with different affinities to mouse
OTR, V1aR, and V1bR. The three antagonists bounds with
significantly different Ki values to OTR and V1aR (P , 0.001);
SR49059 also had a significantly different Ki value for V1bR
(P , 0.001 versus mOTR). However, none of them had a good
selectivity profile for mouse OTR, V1aR, and V1bR, as their Ki
affinities for the V1aR were at most 10–20 times lower than
those for OTR (Table 1).
Coupling Properties of Commonly Used OT/AVP
Analogs at Mouse Receptors. As mOTR, mV1aR, and
mV1bR are all coupled to Gq, leading to phospholipase C
activation, inositol phosphate production, and an increase in
intracellular calcium, we assayed the efficacy of the agonists
by drawing up concentration-response curves of IP1 pro-
duction at the three receptor subtypes. Our results indicate
that OT activated mOTR with an EC50 of 4.45 nM 6 31% CV,
n 5 3; and mV1aR and mV1bR with similar, lower, EC50
values (171 nM 6 19% CV, n 5 3; and 87 nM 6 45% CV, n 5 3)
(Fig. 4A), displaying a good selectivity profile. On the contrary,
AVP activates mV1aR, mV1bR, and mOTR with decreasing
potency (Fig. 4B), with calculated EC50 values of, respectively,
0.65 nM 6 89% CV, n 5 3; 6.62 nM 6 32% CV, n 5 3; and 47.9
nM 6 69% CV, n 5 3. There were no significant differences
between OT and AVP Emax in the three receptor subtypes. In
comparison with OT, the highly selective analog TGOT was
characterized by a left-shifted dose-response curve (Fig. 4C), as
expected on the basis of its high binding affinity. The
calculated
EC50 of TGOT for mOTR was 0.18 nM 6 83% CV, n 5 3;
whereas no IP1 production was observed for mV1aR and
mV1bR also at very high peptide doses (.1000 nM), which is in
accordance with its low affinity for these receptor subtypes.
The functional selective properties of TGOT on mOTR
coupling were investigated by means of a BRET2-based assay
in which the energy donor RLuc is fused to the C-terminal end
of mOTR cDNA and GFP10 used as the acceptor is N-
terminally fused to the Gg2 subunit (GFP
10-Gg2). As shown
in Fig. 5, upon OT (1025 M) and TGOT (1025 M) binding,
TABLE 1
Amino acid sequences and affinity values (Ki) of the
investigated ligands
Substitutions and/or modifications of the amino acid sequence
of OT are indicated in boldface type; the superscript numbers
indicate the position of the residue in the peptide
sequence.
Analog Sequence
Ki for:
mOTR mV1a mV1b
nM 6 %CV
OT Cys1, Tyr2, Ile3, Gln4, Asn5, Cys6, Pro7, Leu8, Gly-NH2
9 0.83 6 17 20.38 6 26 36.32 6 7
AVP Cys1, Tyr2, Phe3, Gln4, Asn5, Cys6, Pro7, Arg8, Gly-NH2
9 0.87 6 8 1.11 6 27 0.43 6 12
dLVT dCys1, Tyr2, Ile3, Gln4, Asn5, Cys6, Pro7, Lys8, Gly-
NH2
9 0.43 6 20 3.39 6 28 0.82 6 7
TGOT Cys1, Tyr2, Ile3, Thr4, Asn5, Cys6, Gly7, Leu8, Gly-
NH2
9 0.04 6 32 .1000 .10,000
Atosiban dCys1, DTyr(Et)2, Ile3, Thr4, Asn5, Cys6, Pro7,
Orn8, Gly-NH2
9 1.29 6 46 .1000 .1000
OTA1 d(CH2)5
1, Tyr(Me)2, Ile3, Thr4, Asn5, Cys6, Pro7, Orn8, Tyr-NH2
9 0.13 6 42 34.3 6 33 374.9 6 20
OTA2 desGly-NH2,d(CH2)5
1, Tyr(Me)2, Ile3, Thr4, Asn5, Cys6, Pro7, Orn8 0.27 6 25 232
6 48 .10,000
OTA3 desGly-NH2,d(CH2)5
1, DTyr2, Ile3, Thr4, Asn5, Cys6, Pro7, Orn8 1.24 6 36 .10,000
.10,000
LVA Phenylac1, DTyr(Me)2, Phe3, Gln4, Asn5, Arg6, Pro7,
Arg8 3.90 6 30 0.10 6 19 9.66 6 12
Manning compound d(CH2)5
1, Tyr(Me)2, Phe3, Gln4, Asn5, Cys6, Pro7, Arg8, Gly-NH2
9 42.6 6 18 1.72 6 28 73.87 6 9
SR49059 13.2 6 19 0.94 6 22 97.35 6 12
d, deamino; d(CH2)5, b-mercapto-b,b-
penthamethylenepropionic; desGly-NH2, desglycineamide;
DTyr(Et), O-ethyl-D-tyrosine; Orn, ornithine; Phenylac,
phenylacetyl; Tyr
(Me), O-methyltyrosine.
Fig. 1. Binding properties of OT/AVP agonists at mOTR,
mV1aR, and mV1bR. Competition binding experiments were
performed using increasing
concentrations (from 10211 to 1025 M) of the endogenous
ligands OT (A) and AVP (B) and the synthetic ligands dLVT
(C) and TGOT (D). Ligand binding
was determined on membrane preparations of COS7 cells
transiently transfected with mOTR (black circles), mV1aR (blue
triangles), and mV1bR (red
squares). Specific binding was determined in the presence of 2–
4 � 1029 M [3H]OT for mOTR and [3H]AVP for mV1aR and
mV1bR; nonspecific binding
was determined in the presence of OT or AVP (1023 M). Each
curve is the mean of triplicate determinations of a single
representative experiment.
Identification of Selective and Potent Analogs of Mouse OTR
321
mOTR significantly (P , 0.001 versus PBS) recruits Gq, Gi1,
Gi2, Gi3, and G0, but not Gs, thus confirming its coupling to
the
same G-protein subtypes recruited by OT (Busnelli et al.,
2012).
Finally, to investigate whether TGOT induces b-arrestin
recruitment after receptor activation, we used a “real -time”
BRET1 assay that uses the mOTR-RLuc construct as the
energy donor and b-arrestin2–YFP or b-arrestin1–YFP as the
acceptor (Fig. 6). In cells coexpressing mOTR-Rluc and
b-arrestin2–YFP or b-arrestin1–YFP, OT at a final concen-
tration of 10 mM increased the BRET ratio with t1/2 values of,
respectively, 89.9 6 2.3 seconds (n 5 3) and 101 6 6.1 seconds
(n 5 3); similarly, TGOT at the same concentration increased
the BRET ratio with t1/2 values of, respectively, 124.6 6 4.2
seconds (n 5 3) and 121.9 6 5.1 seconds (n 5 3). Moreover, the
BRET ratio remained stable for at least 10 minutes (Fig. 6),
thus indicating a sustained agonist-induced association
between mOTR and b-arrestins.
Insights into Molecular Differences between OTR
Subtypes Based on a Structural Model. To evaluate
differences in amino acid composition among mouse, rat, and
human OTR subtypes, we first performed an alignment of
their amino acid sequences (Fig. 7). Secondly, we designed a
three-dimensional homology model of a putative mOTR
conformation to analyze the spatial distribution of different
amino acids at corresponding positions and to study structural-
functional features of the mOTR (Fig. 8). The sequence
alignment shows that several not highly conserved amino
acids are distributed over the entire receptor structure, in
particular in the N-terminal region and C-terminal regions
(Fig. 7). In a previous study, it was shown that truncation of
the first 32 residues of the N terminus of the hOTR did not
influence OT binding (Wesley et al., 2002), and the only
residue in the N terminus found to be relevant for high-
affinity OT binding was the conserved arginine at position 34
(Wesley et al., 2002). Variations in the N terminus are thus
unlikely to be involved in determining TGOT high-affinity
binding to the mOTR.
The three-dimensional visualization of the mouse OTR
serpentine domain (TMHs and loops) shows that Val201 in
TMH5 (hOTR: isoleucine, rOTR: valine), Val301 in ECL3
(hOTR: alanine, rOTR: valine) and Ala313 in TMH7 (hOTR:
valine, rOTR: alanine) are located in close spatial proximity to
the putative ligand binding region (Fig. 8). Based on our
model, only Val201 directly participates in the determination
of the ligand pocket properties (such as the shape) and
biophysical parameters. At this position, an isoleucine is
located in the hOTR, which is different in bulkiness and
length compared with the rodent valine.
Alanine at position 159 (hOTR: alanine; rOTR: glycine) and
valine at position 169 (hOTR: alanine; rOTR: alanine), located
at TMH4, are outside the ligand binding pocket. Residue
Phe51 inTMH1 (hOTR: leucine, rOTR: phenylalanine) points
toward the membrane without any intramolecular interaction,
Fig. 2. Binding properties of commonly used OTAs at mOTR,
mV1aR, and mV1bR. Competition binding experiments were
performed using increasing
concentrations (from 10211 to 1025 M) of the Gq antagonists
atosiban (A), OTA1 (B), OTA2 (C), and OTA3 (D). Ligand
binding was determined on
membrane preparations of COS7 cells transiently transfected
with mOTR (black circles), mV1aR (blue triangles), and mV1bR
(red squares). Specific
binding was determined in the presence of 2–4 � 1029 M
[3H]OT for mOTR and [3H]AVP for mV1aR and mV1bR;
nonspecific binding was determined in
the presence of OT or AVP (1023 M). Each curve is the mean of
triplicate determinations of a single representative experiment.
Fig. 3. Binding properties of commonly used V1a antagonists at
mOTR, mV1aR, and mV1bR. Competition binding experiments
were performed using
increasing concentrations (from 10211 to 1025 M) of three
compounds commonly used as selective V1aR antagonists: LVA
(A), the Manning compound
(B), and SR49059 (C). Ligand binding was determined on
membrane preparations of COS7 cells transiently transfected
with mOTR (black circles),
mV1aR (blue triangles), and mV1bR (red squares). Specific
binding was determined in the presence of 2–4 � 1029 M
[3H]OT for mOTR and [3H]AVP for
mV1aR and mV1bR; nonspecific binding was determined in the
presence of OT or AVP (1023 M). Each curve is the mean of
triplicate determinations of
a single representative experiment.
322 Busnelli et al.
and His69 (hOTR: glutamine; rOTR: histidine) is located at
intracellular loop 1 (ICL1). Therefore, they should not have
a direct impact on ligand binding, even though indirect effects
due to changes in intrinsic signaling capacity (e.g., helix
movement flexibility) cannot be excluded.
Discussion
We describe the in vitro pharmacological characterization
of several analogs of mouse OTR, V1a, and V1b receptors, the
three OT/AVP receptor subtypes expressed in mammalian
brain. Peptidic and nonpeptidic OT/AVP analogs have pri-
marily been assayed for their agonistic and antagonistic
activities in in vitro and in vivo assays based on the peripheral
effects of OT/AVP receptors, with OTR activities being
quantified on the basis of myometrial contractility, V1aR
activities on the basis of vasoconstriction, V1bR activities on
the basis of adrenocorticotropin release, and vasopressin 2
receptor activities on the basis of antidiuresis. Much fewer
pharmacological data have been collected concerning the
selective effects of these analogs within the brain, where their
use at very high doses has often led to conflicting or in-
consistent results (Engelmann et al., 1996). A systematic
analysis of the affinity and efficacy of OT/AVP compounds in
selected brain areas would therefore be extremely valuable in
preclinical research. However, this approach is hindered by
a number of technical issues. First of all, OT/AVP receptors
are not highly expressed in brain, and secondly, tritiated OT
and AVP radiotracers have low specific activity. Overcoming
these limitations would require tissue enrichment procedures
to obtain consistent and reproducible results (Elands et al.,
1988), but this would involve the use of a large number of
animals, increase costs, and raise ethical concerns. Conse-
quently, we believe that the in vitro characterization of
transfected receptors represents a preliminary step for the
selection of candidate drugs to be tested in vivo.
Our in vitro results indicate that, as observed in other
animal species, endogenous OT and AVP ligands have
different selectivity profiles for mouse OT/AVP receptors.
AVP binds to the three brain-expressed OT/AVP receptors
with almost identical affinity, but activates the mV1aR,
mV1bR, and mOTR with decreasing potency over 2 orders of
magnitude. On the contrary, OT has a receptor-specific
affinity range that is highest for OTR and lower for V1aR
and V1bR, and this correlates with its potency at the sites of
the three receptor subtypes. Depending on the dose and site of
administration, some of the actions of AVP may be mediated
via the OTR, and OT can bind and activate V1aR and V1bR
expressed in the brain, albeit with lower affinity than AVP
itself, which means that exogenously administered OT
agonists can elicit substantial responses by binding to AVP
receptors. On the other hand, a low AVP dose may also act as
a “competitive antagonist” at the mOTR, and particularly the
Gq-mediated pathway, which is activated with a high EC50.
Fig. 4. Receptor/Gq coupling properties of OT/AVP analogs
determined by means of IP1 inositol phosphate production. IP1
production was measured
using an immune-competitive HTRF-based assay in HEK293
cells transiently transfected with mOTR (black circles), mV1aR
(blue triangles), and
mV1bR (red squares). A total of 100,000 cells were stimulated
for 30 minutes with increasing concentrations (10214 to 1025
M) of OT (A), AVP (B), and
TGOT (C). Each curve is the mean of triplicate determinations
of a single representative experiment.
Fig. 5. TGOT-induced G-protein recruitment determined by
means of a BRET-based assay. BRET2 was measured in
HEK293 cells transiently
transfected with mOTR-RLuc, GFP10-Gg2, and Gb1 in the
absence (-Ga) or presence of the indicated Ga subunits. The
data represent the differences in
BRET signals between the specified BRET partners in the
absence (PBS; empty bars) or presence of OT (1025 M; black
bars) and TGOT (1025 M; red
bars), and are expressed as the mean value 6 S.E.M. of three
independent assays performed in triplicate. One-way analysis of
variance followed by
Tukey’s test was used to determine the statistical differences
between treatments. ***P , 0.001 vs. untreated controls (PBS).
Identification of Selective and Potent Analogs of Mouse OTR
323
Moreover, the fact that AVP is devoid of agonist activity upon
OTR-mediated Gi1, GoA, and GoB activation (Busnelli et al.,
2012) has still undefined pharmacological implications.
In this study, we show that the synthetic peptide TGOT has
remarkable OTR-versus-V1aR/V1bR selectivity in terms of
affinity binding and coupling for mice receptors. TGOT was
originally demonstrated to be highly selective for rat OTR by
means of in vivo bioassays (Lowbridge et al., 1977). However,
this enhanced OTR/V1a selectivity is lost in humans, in whom
the affinities of OT and TGOT to OTR and V1a receptors are
comparable, thus indicating that the use of TGOT does not
have any advantage over OT as far as OTR/V1a selectivity is
concerned (Chini and Manning, 2007). As shown in Table 2,
comparison of OT and TGOT affinities for human, rat, and
mouse OTRs indicates that OT has the same affinity in the
three species, whereas the affinity of TGOT increases by
a factor of 100 going from human to rat to mouse. Concerning
its coupling features, TGOT binding to the OTR led to the
activation of Gq and all the members of the Gi and Go family
exactly as the endogenous OT ligand (Busnelli et al., 2012).
This is particularly relevant in neuronal cells, where it has
been shown that OTR coupling to Gq and Gi/Go results in
opposite effects on cell excitability via inhibition or activation
of potassium channels (Gravati et al., 2010). Furthermore,
Fig. 6. TGOT-mediated b-arrestin1 and b-arrestin2 recruitment.
BRET1 was monitored in HEK293 cells transiently transfecte d
with mOTR-RLuc and
b-arrestin1–YFP or b-arrestin2–YFP. The cells were stimulated
with OT (1025 M; black squares) or TGOT (1025 M; red
circles). Real-time BRET1
measurements were made every 20 seconds for 10 minutes. The
data represent the differences in BRET signals between the
specified BRET partners in
the absence or presence of the OT and TGOT agonist. Each
curve is the mean of triplicate determinations of a single
representative experiment.
Fig. 7. Sequence alignment of human, rat, and mouse OTR. The
sequences of the human, rat, and mouse OTR orthologs are
aligned to each other.
Asterisks indicate potential TMHs numbered from 1 to 7 and
helix 8. Positions of varying amino acids between the OTR
subtypes are highlighted with
black shadows, whereby the three positions of side chain
variations that are in close spatial proximity to the putative
ligand binding pocket are
highlighted with red shadows.
324 Busnelli et al.
TGOT promotes b-arrestin1 and b-arrestin2 recruitment as
efficiently as OT, suggesting similar desensitization and in-
ternalization properties.
Concerning our pharmacological screening for mOTR-
selective antagonists, three peptides were shown to be very
selective: atosiban, OTA2, and OTA3. In our hands, the most
promising antagonist is OTA3, which we found to be highly
selective for mOTR. Unfortunately, among the V1aR antag-
onists tested, we didn’t identify any ligand with a Ki for the
murine V1a receptor subtype at least 2 orders of magnitude
lower than that for the other two receptor subtypes, a condition
previously set as the minimal requirement for a selective
ligand (Chini et al., 2008). The Manning compound, originally
described as a potent V1aR-selective antagonist in rat, also
was found to be not selective in humans (Manning et al., 2012)
or in mice (this study) and can’t be used as a selective V1a
antagonist in these species.
Revealing variations in ligand binding and signaling among
OTR orthologs in in vitro models has an impact on various
aspects of OT/AVP pharmacology, including the identification
of pharmacological tools that can be used in single species of
particular translational interest, such as genetically modified
mouse models. Our study identified analogs that lack
selectivity for human receptors but are highly selective for
mouse OTRs and therefore very valuable for preclinical
studies. However, as we used transfected cells, one major
issue is to verify whether the selectivity profile observed in
transfected cells is maintained in vivo. In this regard, when
used in mice at a dose of 0.0008 ng/animal, OTA3 specifically
blocked the mOTR-mediated rescue of sociability defects in
heterozygous Oxtr1/2 animals, suggesting the validity of this
approach to identify mOTR antagonists (Sala et al., 2013).
TGOT has been previously used in mice mainly in electro-
physiology experiments, which found evidence of its selectiv-
ity for OTR- versus V1a-mediated responses (Huber et al.,
2005; Gozzi et al., 2010). However, in slices, the effective dose
was very similar to the OT doses used, and the half-maximally
effective concentration was only slightly more potent than OT
(Huber et al., 2005). A similar discrepancy between in vitro
and in vivo potencies of TGOT was also observed in social
behavioral rescue experiments recently performed in OTR-
null mice (Sala et al., 2011, 2013). TGOT rescued the social
deficit at a dose of 0.0005 ng/animal in Oxtr1/2 mice, which is
consistent with a selective action of TGOT through OTRs.
However, at a dose of 0.05 ng/animal, TGOT also rescued the
social deficit of Oxtr2/2 mice, suggesting that despite its very
low affinity for the V1a and V1b receptors in vitro, TGOT was
still active on these receptors in vivo (Sala et al., 2013).
Several factors may be responsible for the discrepancy in
TGOT potency observed in vitro and in vivo. Diffusion and
enzymatic degradation may greatly affect peptides’ stability
in tissues. In the brain, the aminopeptidase oxytocinase
hydrolyzes OT, AVP, enkephalins, and other neuropeptides.
The enzyme is present in soluble and membrane-bound forms,
and its distribution varies greatly in different brain regions
(Fernando et al., 2005). In addition, binding affinity and
velocity of catalysis for different substrates could also account
for significant differences in local neuropeptide concentra-
tions and final neurobiological effects.
Finally, studying the pharmacology of receptor orthologs
may contribute to optimizing the design of selective analogs
because subtle differences in ortholog activation can reveal
crucial ligand-receptor interactions involved in binding and
activation processes. As the conservation of the OTR sequence
in the three species is very high (.90%), it should be feasible,
Fig. 8. Structural homology model of the monomeric mouse
OTR. The
structural model (active conformation) of the mOTR (backbone
cartoon,
white) is represented without the N-terminal extracellular part
(Ntt), the
intracellular tail (Ctt), and the middle portion of ICL3. The
potential
ligand binding region is highlighted by an inner surface (green).
This
pocket-like crevice is constituted by specific amino acids (green
lines)
located at the ECLs and the TMHs toward the extracellular site.
This
three-dimensional representation of the OTR is helpful to
identify
potential links between functional differences of the receptor
subspecies
with particular residue variations. Amino acid positions that are
not
conserved between rodent and human OTR are shown as sticks
(red and
brown, labeled). Amino acids Val201 (TMH5), Val301 (ECL3),
and Ala313
(TMH7) (red sticks) are located in close spatial proximity to the
ligand
binding region, whereby only Val201 is a direct determinant of
the main
ligand binding pocket. Alanine at position 159 and Val169
(TMH4) as well
as Phe51 (TMH1) or His69 (ICL1) are outside the putative
ligand binding
site, and side chain variations should not have direct influences
on ligand
binding properties.
TABLE 2
Affinity constants of OT and TGOT for the human, rat, and
mouse OTR
Ki (mean 6 S.D.) for:
Human OTR Rat OTR Mouse OTR
nM
OT 0.79 6 0.22a 1.0 6 0.1b 0.83 6 0.14c
TGOT 6.62 6 1.22d 0.8 6 0.2b 0.04 6 0.01c
a From Chini et al. (1995).
b From Elands et al. (1988).
c From the current study.
d From Chini et al. (1996).
Identification of Selective and Potent Analogs of Mouse OTR
325
in principle, to identify the variable receptor residue(s)
responsible for differences in affinity binding among the
three species. By analyzing an OTR ortholog sequence
alignment (Fig. 7) and by molecular modeling of mOTR (Fig.
8), we explored the distribution and potential relevance of
nonconserved residues to high agonist binding. Most of the
substitutions among the subspecies are located at the N
terminus and in ICL3. So far as is known, these receptor parts
have not directly related to ligand affinity and selectivity in
OTR species (Wesley et al., 2002). Three varying positions are
located in close spatial proximity to the putative ligand
binding region, whereby only the residue at TMH5 directly
participates in the constitution of the ligand binding pocket.
However, at none of these positions is present a residue that is
different in all the three species, suggesting that the increased
TGOT affinity observed in rodent OTR probably does not
result from a single substitution but is more likely due to the
combination of particular variation(s) close to the ligand
binding site and/or at other distinct receptor parts. With
respect to this topic, it will be of future interest to explore the
effect of multiple amino-acid substitutions in OTR subspecies.
In conclusion, our results indicate that the selectivity
profile of OTR/V1a/V1b receptors for the natural OT and
AVP ligands is conserved in humans and rodents (rats and
mice). However, subtle differences between receptor orthologs
are responsible for an increase in the affinity of the synthetic
agonist TGOT for mOTR. We also identified a number of
OTAs characterized by very high selectivity for mOTR. TGOT
and OTAs are therefore valuable molecular tools for in-
vestigating specific mOTR-mediated effects.
Authorship Contributions
Participated in research design: Busnelli, Chini.
Conducted experiments: Busnelli, Bulgheroni, Kleinau.
Contributed reagents: Manning.
Performed data analysis: Busnelli, Bulgheroni, Kleinau, Chini.
Wrote or contributed to the writing of the manuscript: Busnelli,
Manning, Kleinau, Chini.
References
Barberis C, Morin D, Durroux T, Mouillac B, Guillon G, Seyer
R, Hibert M,
and Tribollet E (1999) Molecular pharmacology of AVP and OT
receptors and
therapeutic potential. Drug News Perspect 12:279–292.
Birnbaumer M (2000) Vasopressin receptors. Trends Endocrinol
Metab 11:406–410.
Busnelli M, Saulière A, Manning M, Bouvier M, Galés C, and
Chini B (2012) Func-
tional selective oxytocin-derived agonists discriminate between
individual G pro-
tein family subtypes. J Biol Chem 287:3617–3629.
Chini B and Manning M (2007) Agonist selectivity in the
oxytocin/vasopressin re-
ceptor family: new insights and challenges. Biochem Soc Trans
35:737–741.
Chini B, Manning M, and Guillon G (2008) Affinity and
efficacy of selective agonists
and antagonists for vasopressin and oxytocin receptors: an
“easy guide” to receptor
pharmacology. Prog Brain Res 170:513–517.
Chini B, Mouillac B, Ala Y, Balestre MN, Trumpp-Kallmeyer S,
Hoflack J, Elands J,
Hibert M, Manning M, and Jard S, et al. (1995) Tyr115 is the
key residue for
determining agonist selectivity in the V1a vasopressin receptor.
EMBO J 14:
2176–2182.
Costanzi S (2012) Homology modeling of class A G protein-
coupled receptors. Meth-
ods Mol Biol 857:259–279.
Deupi X and Standfuss J (2011) Structural insights into agonist-
induced activation of
G-protein-coupled receptors. Curr Opin Struct Biol 21:541–551.
Elands J, Barberis C, and Jard S (1988) [3H]-[Thr4,Gly7]OT: a
highly selective ligand
for central and peripheral OT receptors. Am J Physiol 254:E31–
E38.
Engelmann M, Wotjak CT, Neumann I, Ludwig M, and Landgraf
R (1996) Behavioral
consequences of intracerebral vasopressin and oxytocin: focus
on learning and
memory. Neurosci Biobehav Rev 20:341–358.
Ferguson JN, Aldag JM, Insel TR, and Young LJ (2001)
Oxytocin in the medial
amygdala is essential for social recognition in the mouse. J
Neurosci 21:8278–8285.
Ferguson JN, Young LJ, Hearn EF, Matzuk MM, Insel TR, and
Winslow JT (2000)
Social amnesia in mice lacking the oxytocin gene. Nat Genet
25:284–288.
Fernando RN, Larm J, Albiston AL, and Chai SY (2005)
Distribution and cellular
localization of insulin-regulated aminopeptidase in the rat
central nervous system.
J Comp Neurol 487:372–390.
Galés C, Van Durm JJ, Schaak S, Pontier S, Percherancier Y,
Audet M, Paris H,
and Bouvier M (2006) Probing the activation-promoted
structural rearrangements
in preassembled receptor-G protein complexes. Nat Struct Mol
Biol 13:778–786.
Gozzi A, Jain A, Giovannelli A, Bertollini C, Crestan V,
Schwarz AJ, Tsetsenis T,
Ragozzino D, Gross CT, and Bifone A (2010) A neural switch
for active and passive
fear. Neuron 67:656–666.
Gravati M, Busnelli M, Bulgheroni E, Reversi A, Spaiardi P,
Parenti M, Toselli M,
and Chini B (2010) Dual modulation of inward rectifier
potassium currents in
olfactory neuronal cells by promiscuous G protein coupling of
the oxytocin receptor.
J Neurochem 114:1424–1435.
Hanson MA and Stevens RC (2009) Discovery of new GPCR
biology: one receptor
structure at a time. Structure 17:8–14.
Higashida H, Yokoyama S, Munesue T, Kikuchi M, Minabe Y,
and Lopatina O (2011)
CD38 gene knockout juvenile mice: a model of oxytocin signal
defects in autism.
Biol Pharm Bull 34:1369–1372.
Huber D, Veinante P, and Stoop R (2005) Vasopressin and
oxytocin excite distinct
neuronal populations in the central amygdala. Science 308:245–
248.
Jacobson KA and Costanzi S (2012) New insights for drug
design from the X-ray
crystallographic structures of G-protein-coupled receptors. Mol
Pharmacol 82:
361–371.
Jean-Alphonse F, Perkovska S, Frantz MC, Durroux T, Méjean
C, Morin D, Loison S,
Bonnet D, Hibert M, and Mouillac B, et al. (2009) Biased
agonist pharmacocha-
perones of the AVP V2 receptor may treat congenital
nephrogenic diabetes insip-
idus. J Am Soc Nephrol 20:2190–2203.
Jin D, Liu HX, Hirai H, Torashima T, Nagai T, Lopatina O,
Shnayder NA, Yamada K,
Noda M, and Seike T, et al. (2007) CD38 is critical for social
behaviour by regu-
lating oxytocin secretion. Nature 446:41–45.
Kenakin T (2011) Functional selectivity and biased receptor
signaling. J Pharmacol
Exp Ther 336:296–302.
Kobilka B and Schertler GF (2008) New G-protein-coupled
receptor crystal struc-
tures: insights and limitations. Trends Pharmacol Sci 29:79–83.
Kratochwil NA, Gatti-McArthur S, Hoener MC, Lindemann L,
Christ AD, Green LG,
Guba W, Martin RE, Malherbe P, and Porter RH, et al. (2011) G
protein-coupled
receptor transmembrane binding pockets and their applications
in GPCR research
and drug discovery: a survey. Curr Top Med Chem 11:1902–
1924.
Lowbridge J, Manning M, Haldar J, and Sawyer WH (1977)
Synthesis and some
pharmacological properties of [4-threonine, 7-glycine]oxytocin,
[1-(L-2-hydroxy-3-
mercaptopropanoic acid), 4-threonine, 7-glycine]oxytocin
(hydroxyl[Thr4, Gly7]
oxytocin), and [7-glycine]oxytocin: peptides with high
oxytocic-antidiuretic selec-
tivity. J Med Chem 20:120–123.
MacKinnon AC, Tufail-Hanif U, Wheatley M, Rossi AG,
Haslett C, Seckl M,
and Sethi T (2009) Targeting V1A-vasopressin receptors with
[Arg6, D-Trp7,9,
NmePhe8]-substance P (6-11) identifies a strategy to develop
novel anti-cancer
therapies. Br J Pharmacol 156:36–47.
Manning M, Misicka A, Olma A, Bankowski K, Stoev S, Chini
B, Durroux T, Mouillac
B, Corbani M, and Guillon G (2012) Oxytocin and vasopressin
agonists and
antagonists as research tools and potential therapeutics. J
Neuroendocrinol 24:
609–628.
Manning M, Stoev S, Chini B, Durroux T, Mouillac B, and
Guillon G (2008) Peptide
and non-peptide agonists and antagonists for the vasopressin
and oxytocin V1a,
V1b, V2 and OT receptors: research tools and potential
therapeutic agents. Prog
Brain Res 170:473–512.
Michino M, Abola E, Brooks CL, 3rd, Dixon JS, Moult J, and
Stevens RC; GPCR Dock
2008 participants (2009) Community-wide assessment of GPCR
structure model-
ling and ligand docking: GPCR Dock 2008. Nat Rev Drug
Discov 8:455–463.
Miller G (2013) Neuroscience. The promise and perils of
oxytocin. Science 339:
267–269.
Modi ME and Young LJ (2012) The oxytocin system in drug
discovery for autism:
animal models and novel therapeutic strategies. Horm Behav
61:340–350.
Oshikawa S, Tanoue A, Koshimizu TA, Kitagawa Y, and
Tsujimoto G (2004) Vaso-
pressin stimulates insulin release from islet cells through V1b
receptors: a com-
bined pharmacological/knockout approach. Mol Pharmacol
65:623–629.
Pittman QJ and Spencer SJ (2005) Neurohypophysial peptides:
gatekeepers in the
amygdala. Trends Endocrinol Metab 16:343–344.
Rasmussen SG, DeVree BT, Zou Y, Kruse AC, Chung KY,
Kobilka TS, Thian FS,
Chae PS, Pardon E, and Calinski D, et al. (2011) Crystal
structure of the b2
adrenergic receptor-Gs protein complex. Nature 477:549–555.
Reversi A, Rimoldi V, Marrocco T, Cassoni P, Bussolati G,
Parenti M, and Chini B
(2005) The oxytocin receptor antagonist atosiban inhibits cell
growth via a “biased
agonist” mechanism. J Biol Chem 280:16311–16318.
Ring RH, Schechter LE, Leonard SK, Dwyer JM, Platt BJ, Graf
R, Grauer S, Puli-
cicchio C, Resnick L, and Rahman Z, et al. (2010) Receptor and
behavioral phar-
macology of WAY-267464, a non-peptide oxytocin receptor
agonist. Neuropharmacology
58:69–77.
Sala M, Braida D, Donzelli A, Martucci R, Busnelli M,
Bulgheroni E, Rubino T,
Parolaro D, Nishimori K, and Chini B (2013) Mice
heterozygous for the oxytocin
receptor gene (Oxtr(1/-)) show impaired social behaviour but
not increased ag-
gression or cognitive inflexibility: evidence of a selective
haploinsufficiency gene
effect. J Neuroendocrinol 25:107–118.
Sala M, Braida D, Lentini D, Busnelli M, Bulgheroni E,
Capurro V, Finardi A,
Donzelli A, Pattini L, and Rubino T, et al. (2011)
Pharmacologic rescue of impaired
cognitive flexibility, social deficits, increased aggression, and
seizure susceptibility
in oxytocin receptor null mice: a neurobehavioral model of
autism. Biol Psychiatry
69:875–882.
Serradeil-Le Gal C, Raufaste D, Derick S, Blankenstein J, Allen
J, Pouzet B, Pascal
M, Wagnon J, and Ventura MA (2007) Biological
characterization of rodent and
human vasopressin V1b receptors using SSR-149415, a
nonpeptide V1b receptor
ligand. Am J Physiol Regul Integr Comp Physiol 293:R938–
R949.
Takayanagi Y, Yoshida M, Bielsky IF, Ross HE, Kawamata M,
Onaka T, Yanagisawa
T, Kimura T, Matzuk MM, and Young LJ, et al. (2005)
Pervasive social deficits, but
326 Busnelli et al.
normal parturition, in oxytocin receptor-deficient mice. Proc
Natl Acad Sci USA
102:16096–16101.
Urban JD, Clarke WP, von Zastrow M, Nichols DE, Kobilka B,
Weinstein H, Javitch
JA, Roth BL, Christopoulos A, and Sexton PM, et al. (2007)
Functional selectivity
and classical concepts of quantitative pharmacology. J
Pharmacol Exp Ther 320:
1–13.
Wallis M (2012) Molecular evolution of the neurohypophysial
hormone precursors in
mammals: comparative genomics reveals novel mammalian
oxytocin and vaso-
pressin analogues. Gen Comp Endocrinol 179:313–318.
Wesley VJ, Hawtin SR, Howard HC, and Wheatley M (2002)
Agonist-specific, high-
affinity binding epitopes are contributed by an arginine in the
N-terminus of the
human oxytocin receptor. Biochemistr y 41:5086–5092.
Wichard JD, Ter Laak A, Krause G, Heinrich N, Kühne R, and
Kleinau G (2011)
Chemogenomic analysis of G-protein coupled receptors and
their ligands deciphers
locks and keys governing diverse aspects of signalling. PLoS
ONE 6:e16811.
Zhao Q and Wu BL (2012) Ice breaking in GPCR structural
biology. Acta Pharmacol
Sin 33:324–334.
Zingg HH and Laporte SA (2003) The oxytocin receptor. Trends
Endocrinol Metab 14:
222–227.
Address correspondence to: Dr. Bice Chini, CNR, Institute of
Neuroscience,
Via Vanvitelli 32, 20129 Milano, Italy. E-mail:
[email protected]
Identification of Selective and Potent Analogs of Mouse OTR
327
mailto:[email protected]
PK-PD Primer Homework
Max 4pages only answers
PK-Pharmacology Primer
Instructions for the PK problems:
Please answer the questions below as best as you can by
examining the outcomes of the pharmacokinetic models
provided in the Excel file “CLRE-238 SP21 PK-PD
Homework.xlxs.” In many cases, you will need to find the
requested value of parameters by trial and error, changing some
of the values in bold red in the worksheets and looking at the
resulting plots. You do not need to perform any calculations.
All the answers are very short. Do not be intimidated by the
Excel worksheets; this is a lot easier than it appears at first
glance.
General comment: Because you are expected to obtain your
answers by visually examining the pharmacokinetic plots in the
associated Excel file, your quantitative answers will be graded
as correct if they were reasonably close to the exact answer.
There is no requirement to be precise.
Problem A:
The antibiotic genericomycin (not a real one) is administered by
iv bolus to hospitalized patients. The antibiotic has an
elimination half-life of 4.0 h in people. Previous experience has
shown that to be effective at reducing the bacterial burden its
plasma concentration must equal or higher than 5 µg/ml (Ceff).
Question 1: Using the “Intravenous” tab of the Excel file,
determine the duration of action of the following iv bolus doses
and enter the values in the table below.
Hint: You can eyeball the time values from the chart or read
them from the list of Cp vs time for the Plot (blue numbers)
Answer 1:
Dose
(mg)
Duration of Action
(h)
140
4
280
8
560
12
1120
16
Question 2: What is the effect of doubling the dose on the
duration of action?
Answer 2: it adds the duration of action by 4 hours
Question 3: What would be the necessary iv bolus dose to
establish plasma concentration 5 µg/ml at 24 h? Is this dose
realistic? Assume genericomycin costs $10/mg.
Answer 3:
Problem B:
You have created a new company, NovoAntibiot, Inc. with the
intention of creating a new antibiotic with much better
properties than genericomycin. Your clinical advisors suggest
that the new antibiotic should be administered by iv bolus once
a day with a dose of at most 560 mg per person (your new
therapeutic product profile).
Question 4: What elimination half-life should you design in
your new antibiotic to achieve a plasma concentration of at least
5 µg/ml 24 h after a 560 mg iv bolus dose?
Answer 4: 40
Welcome to drug discovery! After working for 2 years and
spending $15 M from your Angel and series A investors you
were unable to increase the half-life of your new antibiotics. All
of them still have halflives around 4 h. But you got lucky, some
of your new compounds have higher potency. Drug candidate
NABT 813, is much more potent than genericomycin. In vitro
experiments and animal models suggest that the minimum
effective concentration (Ceff) in humans will be 1 µg/ml.
Question 5: What iv bolus dose of NABT 813 will treat the
patients for 24h if its elimination half-life is 4.0 h and its Ceff
is 1 µg/ml? Is it realistic? (compare with your Answer 3
above).)
Answer 5: 140mg
Problem C
Unfortunately, your new antibiotic NABT 813 decreased the
number and body weight of rat pups in formal GLP repro-tox
studies in rats. The compound cannot be used in women who
might not know whether they are pregnant at the time they are
admitted to the hospital with a systemic infection. This is a
crushing blow and your investors abandon you. The new drug
discovery program is terminated.
However, your PK consultant suggests that you might be able to
formulate genericomycin for subcutaneous administration by
creating an insoluble salt and adding some excipients that will
retard its absorption, allowing (perhaps) once a day dosing.
Your intellectual property attorney states that you might be able
to obtain patent protection for the new formulation if you
demonstrate substantial advantages over the generic iv
formulation. You apply for and get an SBIR grant to
demonstrate proof of (PK) concept.
Question 6: In the “Subcutaneous” tab of the Excel file, set the
elimination half-life of genericomycin to 4.0 h, its Ceff at 5
µg/ml, and its dose to 280 mg. Examine the effect of changing
the absorption half-life of genericomycin from your new
formulations by replacing the value of t1/2 (abs) and complete
the table below.
Answer 6:
t1/2 abs (h)
Tmax (h)
Cmax (µg/ml)
0.05
0.25
18
1
2.5
13
3
5
8.5
6
7
5.9
10
9
4.2
Hint: You can eyeball the above values from the chart or read
them from the list of Cp vs time for the Plots (blue numbers).
No need to be very precise.
Question 7: Why does the PK profile for absorption with a very
short half-life (0.05 h) look like the ivbolus profile? (This never
happens in real life….)
Answer 7: Clearance is more rapid and half-lives are shorter
Question 8: What is the effect of increasing the absorption half-
life on Tmax and Cmax?
Answer 8: increasing tmax increases absorption while
increasing cmax decreases absorption
Question 9: Is there any value of absorption half-life that will
allow you to have plasma concentration higher or equal to Ceff
at 24 h when dosing sc 280 mg?
Answer 9: yes
Question 10: What is the duration of action of genericomycin at
a dose of 280 mg if it is administered in the formulation with
absorption half-life of 10 hours?
Answer 10: 4.3
Problem X (bonus):
Your microbiology consultant tells you that an analogue of
genericomycin with longer eliminationhalflife was developed in
the Soviet Union in the 1960’s, but it never reached the West
and was forgotten. Its name was bolshoimitsina and had an
elimination half-life in humans of 7.8 h, and Ceff also 5 µg/ml.
You obtain a sample of bolshoimitsina via a website vendor in
Bulgaria and find that it also has a sc absorption half-life of
10.0 h in your preferred formulation.
Question 11: What sc dose of formulated bolshoimitsina will
provide a plasma concentration equal to Ceff 5 µg/ml at 24 h?
Answer 11: 9.43mg
Question 12: At the dose of bolshoimitsina you found in Answer
11, when does the drug start having its effect in the patient?
Answer 12: 0.5hrs
PK-Pharmacology Primer
Homework with Answers
Student Name:
Instructions for the Pharmacology problems:
Please extract the requested information from the papers
provided. You do not need to read the papers in detail. This is
an exercise in identifying the required information quickly
while checking its validity. Your answers will be very short, a
value (with units!) or a few words.
Problem P:
Look at the provided paper by Busnelli et al., “Selective and
Potent Agonists and Antagonists for Investigating the Role of
Mouse Oxytocin Receptors”, and answer the following
questions:
Question 31: What was the Ki value for compound “LVA” at
the mOTR (mouse oxytocin receptor)?
Answer 31:
Question 32: Does LVA have higher or lower affinity than “OT”
(oxytocin) for the mOTR?
Answer 32:
Look in Materials and Methods, section on Ligand Binding
Assays.
Question 33: How were the Ki values obtained: Schild analysis
or from IC50 values?
Answer 33:
Question 34: What compound was the radioligand for measuring
binding to the mOTR, and at what concentration was used?
Answer 34:
Question 35: What was the Kd value for the radioligand at the
mOTR?
Answer 35:
Look in the main body of the paper.
Question 36: Which figure shows the competition binding
experiments for compound “OTA1”?
Answer 36:
Question 37: Looking at said figure, would you say that OTA1
has higher affinity for the mV1aR (mouse vasopressin type 1a
receptor) than for the mOTR?
Answer 37:
Problem Q:
Look at the provided paper by Ichinose et al., “Development of
a Highly Potent Analogue and a Long-Acting Analogue of
Oxytocin for the Treatment of Social Impairment-Like
Behaviors”, and answer the following questions:
Question 38: What was the EC50 at the hOTR (human oxytocin
receptor) of compound 4?
Answer 38:
Question 39: What was the efficacy of compound 4?
Answer 39:
Question 40: Of the new compounds prepared in this paper
(Compounds 1 to 6), which one was more potent at the hOTR?
Which one was the most potent at the hV1aR?
Answer 40:
Look in Results and Discussion, section on Agonist-Induced
Increase in Intracellular Ca2+ Concentrations.
Question 41: Which second messenger was measured to
determine receptor activation?
Answer 41:
Look at Table 2, data on Ki at the hOTR and EC50 at the hOTR
Question 42: Would you suspect that there might be
considerable “receptor reserve” for activation of the hOTR by
OT? What about carbetocin?
Answer 42:
Page 1 of 5
PK-PD Primer Homework
Max 4pages only answers
PK-Pharmacology Primer
Instructions for the PK problems:
Please answer the questions below as best as you can by
examining the outcomes of the pharmacokinetic models
provided in the Excel file “CLRE-238 SP21 PK-PD
Homework.xlxs.” In many cases, you will need to find the
requested value of parameters by trial and error, changing some
of the values in bold red in the worksheets and looking at the
resulting plots. You do not need to perform any calculations.
All the answers are very short. Do not be intimidated by the
Excel worksheets; this is a lot easier than it appears at first
glance.
General comment: Because you are expected to obtain your
answers by visually examining the pharmacokinetic plots in the
associated Excel file, your quantitative answers will be graded
as correct if they were reasonably close to the exact answer.
There is no requirement to be precise.
Problem A:
The antibiotic genericomycin (not a real one) is administered by
iv bolus to hospitalized patients. The antibiotic has an
elimination half-life of 4.0 h in people. Previous experience has
shown that to be effective at reducing the bacterial burden its
plasma concentration must equal or higher than 5 µg/ml (Ceff).
Question 1: Using the “Intravenous” tab of the Excel file,
determine the duration of action of the following iv bolus doses
and enter the values in the table below.
Hint: You can eyeball the time values from the chart or read
them from the list of Cp vs time for the Plot (blue numbers)
Answer 1:
Dose
(mg)
Duration of Action
(h)
140
4
280
8
560
12
1120
16
Question 2: What is the effect of doubling the dose on the
duration of action?
Answer 2: it adds the duration of action by 4 hours
Question 3: What would be the necessary iv bolus dose to
establish plasma concentration 5 µg/ml at 24 h? Is this dose
realistic? Assume genericomycin costs $10/mg.
Answer 3:
Problem B:
You have created a new company, NovoAntibiot, Inc. with the
intention of creating a new antibiotic with much better
properties than genericomycin. Your clinical advisors suggest
that the new antibiotic should be administered by iv bolus once
a day with a dose of at most 560 mg per person (your new
therapeutic product profile).
Question 4: What elimination half-life should you design in
your new antibiotic to achieve a plasma concentration of at least
5 µg/ml 24 h after a 560 mg iv bolus dose?
Answer 4: 40
Welcome to drug discovery! After working for 2 years and
spending $15 M from your Angel and series A investors you
were unable to increase the half-life of your new antibiotics. All
of them still have halflives around 4 h. But you got lucky, some
of your new compounds have higher potency. Drug candidate
NABT 813, is much more potent than genericomycin. In vitro
experiments and animal models suggest that the minimum
effective concentration (Ceff) in humans will be 1 µg/ml.
Question 5: What iv bolus dose of NABT 813 will treat the
patients for 24h if its elimination half-life is 4.0 h and its Ceff
is 1 µg/ml? Is it realistic? (compare with your Answer 3
above).)
Answer 5: 140mg
Problem C
Unfortunately, your new antibiotic NABT 813 decreased the
number and body weight of rat pups in formal GLP repro-tox
studies in rats. The compound cannot be used in women who
might not know whether they are pregnant at the time they are
admitted to the hospital with a systemic infection. This is a
crushing blow and your investors abandon you. The new drug
discovery program is terminated.
However, your PK consultant suggests that you might be able to
formulate genericomycin for subcutaneous administration by
creating an insoluble salt and adding some excipients that will
retard its absorption, allowing (perhaps) once a day dosing.
Your intellectual property attorney states that you might be able
to obtain patent protection for the new formulation if you
demonstrate substantial advantages over the generic iv
formulation. You apply for and get an SBIR grant to
demonstrate proof of (PK) concept.
Question 6: In the “Subcutaneous” tab of the Excel file, set the
elimination half-life of genericomycin to 4.0 h, its Ceff at 5
µg/ml, and its dose to 280 mg. Examine the effect of changing
the absorption half-life of genericomycin from your new
formulations by replacing the value of t1/2 (abs) and complete
the table below.
Answer 6:
t1/2 abs (h)
Tmax (h)
Cmax (µg/ml)
0.05
0.25
18
1
2.5
13
3
5
8.5
6
7
5.9
10
9
4.2
Hint: You can eyeball the above values from the chart or read
them from the list of Cp vs time for the Plots (blue numbers).
No need to be very precise.
Question 7: Why does the PK profile for absorption with a very
short half-life (0.05 h) look like the ivbolus profile? (This never
happens in real life….)
Answer 7: Clearance is more rapid and half-lives are shorter
Question 8: What is the effect of increasing the absorption half-
life on Tmax and Cmax?
Answer 8: increasing tmax increases absorption while
increasing cmax decreases absorption
Question 9: Is there any value of absorption half-life that will
allow you to have plasma concentration higher or equal to Ceff
at 24 h when dosing sc 280 mg?
Answer 9: yes
Question 10: What is the duration of action of genericomycin at
a dose of 280 mg if it is administered in the formulation with
absorption half-life of 10 hours?
Answer 10: 4.3
Problem X (bonus):
Your microbiology consultant tells you that an analogue of
genericomycin with longer eliminationhalflife was developed in
the Soviet Union in the 1960’s, but it never reached the West
and was forgotten. Its name was bolshoimitsina and had an
elimination half-life in humans of 7.8 h, and Ceff also 5 µg/ml.
You obtain a sample of bolshoimitsina via a website vendor in
Bulgaria and find that it also has a sc absorption half-life of
10.0 h in your preferred formulation.
Question 11: What sc dose of formulated bolshoimitsina will
provide a plasma concentration equal to Ceff 5 µg/ml at 24 h?
Answer 11: 9.43mg
Question 12: At the dose of bolshoimitsina you found in Answer
11, when does the drug start having its effect in the patient?
Answer 12: 0.5hrs
PK-Pharmacology Primer
Homework with Answers
Student Name:
Instructions for the Pharmacology problems:
Please extract the requested information from the papers
provided. You do not need to read the papers in detail. This is
an exercise in identifying the required information quickly
while checking its validity. Your answers will be very short, a
value (with units!) or a few words.
Problem P:
Look at the provided paper by Busnelli et al., “Selective and
Potent Agonists and Antagonists for Investigating the Role of
Mouse Oxytocin Receptors”, and answer the following
questions:
Question 31: What was the Ki value for compound “LVA” at
the mOTR (mouse oxytocin receptor)?
Answer 31:
Question 32: Does LVA have higher or lower affinity than “OT”
(oxytocin) for the mOTR?
Answer 32:
Look in Materials and Methods, section on Ligand Binding
Assays.
Question 33: How were the Ki values obtained: Schild analysis
or from IC50 values?
Answer 33:
Question 34: What compound was the radioligand for measuring
binding to the mOTR, and at what concentration was used?
Answer 34:
Question 35: What was the Kd value for the radioligand at the
mOTR?
Answer 35:
Look in the main body of the paper.
Question 36: Which figure shows the competition binding
experiments for compound “OTA1”?
Answer 36:
Question 37: Looking at said figure, would you say that OTA1
has higher affinity for the mV1aR (mouse vasopressin type 1a
receptor) than for the mOTR?
Answer 37:
Problem Q:
Look at the provided paper by Ichinose et al., “Development of
a Highly Potent Analogue and a Long-Acting Analogue of
Oxytocin for the Treatment of Social Impairment-Like
Behaviors”, and answer the following questions:
Question 38: What was the EC50 at the hOTR (human oxytocin
receptor) of compound 4?
Answer 38:
Question 39: What was the efficacy of compound 4?
Answer 39:
Question 40: Of the new compounds prepared in this paper
(Compounds 1 to 6), which one was more potent at the hOTR?
Which one was the most potent at the hV1aR?
Answer 40:
Look in Results and Discussion, section on Agonist-Induced
Increase in Intracellular Ca2+ Concentrations.
Question 41: Which second messenger was measured to
determine receptor activation?
Answer 41:
Look at Table 2, data on Ki at the hOTR and EC50 at the hOTR
Question 42: Would you suspect that there might be
considerable “receptor reserve” for activation of the hOTR by
OT? What about carbetocin?
Answer 42:
Page 1 of 5
IntravenousThese equations are for reference only. You do not
have to use them to complete your homework. But they help to
understand the results!Plasma concentration (Cp) for one
compartment model with iv bolus dosing and first order
eliminationCp= (D/Vd)*exp(-ke*t)Equation for extension of
duration of actionUsing the model in this worksheet, please
answer the questions in the handout Ci is the plasma
concentration at a given time, assumed to be Ci>CeffPlease do
not change any of the calculationsDelta teff is for how much
longer the drug will be active (above Ceff) for a given half-
lifeYou only have to change some of the values of some of the
parameters in bold redThis equation can be derived from the
Cp(t) equation aboveThis plot describes the plasma
concentration (Cp) of an ideal drug after administration of an
intravenous bolus(There are no drugs with this simple
pharmacokinetic behavior, but it is a simple illustrative
example)The drug is considered active in the patient if its
plasma concentration is superior to a threshold value "Ceff"
determined from previous clinical
experienceParameterAbbreviationUnitsYour ValuesElimination
half-lifet1/2 (elim)h4.0Same plot, with Y axis set to maximum
Cp 10 ug/mlDoseDmg/personActive plasma concentration
thresholdCeffug/ml1.0Don't touch these cellsElimination
rateke1/h0.173Apparent volume of
distributionVdL/person14Threshold plotTime (h)Ceff
(ug/ml)01.0251.0Don't touch these cellsPlot Time (h)Cp
(ug/ml)00.00.10.00.250.00.50.010.01.50.020.030.040.050.060.0
80.0100.0120.0140.0160.0180.0200.0220.0240.000260.0280.03
00.0
PK Intravenous Administration
Cp (ug/ml) 0 0.1 0.25 0.5 1 1.5 2 3 4 5
6 8 10 12 14 16 18 20 22 24 26 28
30 0 0 0 0 0 0 0 0 0 0 0
0 0 0 0 0 0 0 0 0 0 0 0
Ceff (ug/ml) 0 25 1 1
Time (h)
Cp (ug/ml)
PK Intravenous Administration
Cp (ug/ml) 0 0.1 0.25 0.5 1 1.5 2 3 4 5
6 8 10 12 14 16 18 20 22 24 26 28
30 0 0 0 0 0 0 0 0 0 0 0
0 0 0 0 0 0 0 0 0 0 0 0
Ceff (ug/ml) 0 25 1 1
Time (h)
Cp (ug/ml)
SubcutaneousCLRE-238Applied Translational ResearchThese
equations are for reference only. You do not have to use them to
complete your homework. But they help to understand the
results!Spring 2021PK-PD Primer HomeworkPlasma
concentration (Cp) for one compartment model with sc bolus
dosing and first order eliminationDate3/30/21Claudio D.
Schteingart Ph.D.C(t) is Cp the plasma concentration as a
function of timef is the bioavailability (scale 0 to 1) of the drug
in the sc spaceUsing the model in this worksheet, please answer
the questions in the handout ka is the absorption rate, ke is the
elimination ratePlease do not change any of the calculationsD is
the dose, Vd is the apparent volume of distributionYou only
have to change some of the values of some of the parameters in
bold redEquation for extension of duration of actionCi is the
plasma concentration at a given time, assumed to be
Ci>CeffDelta teff is for how much longer the drug will be
active (above Ceff) for a given half-lifeThis equation can be
used during the elimination phase of the above curveThis plot
describes the plasma concentration (Cp) of an ideal drug after
administration of a subcutaneous bolusThe drug is considered
active in the patient if its plasma concentration is superior to a
threshold value "Ceff" determined from previous clini cal
experienceIf the absorption half-life equals the elimination half-
life the math fails (division by zero). If needed, enter a slightly
different value, e.g. 3.99 or 4.01 instead of 4.00Same plot, with
Y axis set to maximum Cp 10
ug/mlParameterAbbreviati onUnitsYour ValuesAbsorption half-
lifet1/2 (abs)h24.00Elimination half-lifet1/2
(elim)h5.0DoseDmg/person9.43Active plasma concentration
thresholdCeffug/ml5.0Don't touch these cellsSubcutaneous
bioavailability 0 to 1f1Apparent volume of
distributionVdL/person14Absorption rateka1/h0.029Elimination
rateke1/h0.139Multiplication factorfDka/(Vd(Ka-ke))ug/ml-
0Threshold plotTime (h)Ceff (ug/ml)05.0265.0Don't touch these
cellsPlotTime (h)Cp
(ug/ml)00.000.10.000.250.000.50.0110.021.50.0320.032.50.043
0.053.50.0540.0650.0660.0770.0890.09110.09130.09150.09170.
09190.09210.09230.08240.08250.08260.08270.08
PK Subcutaneous Administration
Cp (ug/ml) 0 0.1 0.25 0.5 1 1.5 2 2.5 3
3.5 4 5 6 7 9 11 13 15 17 19 21
23 24 25 26 27 0 1.9291349543759174E-3
4.7627511742739078E-3 9.3290009959562988E-3
1.7899518669098738E-2 2.5764263897523468E-2
3.297239017841809E-2 3.9569732249353361E-2
4.5599028724170154E-2 5.1100129814037637E-2
5.6110191132563512E-2 6.4793416734735515E-2
7.1898633268821469E-2 7.764303677600605E-2
8.5779467479190435E-2 9.0433868498689868E-2
9.2534238683107894E-2 9.2779107146231402E-2
9.1693364761172624E-2 8.967056151835319E-2
8.7004947768308957E-2 8.3915743577409357E-2
8.2264905141199454E-2 8.056551884472278E-2
7.8831334871670267E-2 7.7074110918482291E-2 Ceff
(ug/ml) 0 26 5 5
Time (h)
Cp (ug/ml)
PK Subcutaneous Administration
Cp (ug/ml) 0 0.1 0.25 0.5 1 1.5 2 2.5 3
3.5 4 5 6 7 9 11 13 15 17 19 21
23 24 25 26 27 0 1.9291349543759174E-3
4.7627511742739078E-3 9.3290009959562988E-3
1.7899518669098738E-2 2.5764263897523468E-2
3.297239017841809E-2 3.9569732249353361E-2
4.5599028724170154E-2 5.1100129814037637E-2
5.6110191132563512E-2 6.4793416734735515E-2
7.1898633268821469E-2 7.764303677600605E-2
8.5779467479190435E-2 9.0433868498689868E-2
9.2534238683107894E-2 9.2779107146231402E-2
9.1693364761172624E-2 8.967056151835319E-2
8.7004947768308957E-2 8.3915743577409357E-2
8.2264905141199454E-2 8.056551884472278E-2
7.8831334871670267E-2 7.7074110918482291E-2 Ceff
(ug/ml) 0 26 5 5
Time (h)
Cp (ug/ml)

More Related Content

Similar to 1521-01033462318–327$25.00 httpdx.doi.org10.1124jpet.1

Basic Formal Ontology (BFO) and Disease
 Basic Formal Ontology (BFO) and Disease Basic Formal Ontology (BFO) and Disease
Basic Formal Ontology (BFO) and DiseaseBarry Smith
 
Omics technology: an important tool in mechanism studies of Chinese herbal fo...
Omics technology: an important tool in mechanism studies of Chinese herbal fo...Omics technology: an important tool in mechanism studies of Chinese herbal fo...
Omics technology: an important tool in mechanism studies of Chinese herbal fo...LucyPi1
 
PUTATIVE DRUG TARGET IDENTIFICATION FOR SEPTIC ARTHRITIS THROUGH DATA MINING ...
PUTATIVE DRUG TARGET IDENTIFICATION FOR SEPTIC ARTHRITIS THROUGH DATA MINING ...PUTATIVE DRUG TARGET IDENTIFICATION FOR SEPTIC ARTHRITIS THROUGH DATA MINING ...
PUTATIVE DRUG TARGET IDENTIFICATION FOR SEPTIC ARTHRITIS THROUGH DATA MINING ...Jing Zang
 
Jpcr mauro luisetto et al amyotrophic lateral sclerosis and endogenous esoge...
Jpcr mauro luisetto  et al amyotrophic lateral sclerosis and endogenous esoge...Jpcr mauro luisetto  et al amyotrophic lateral sclerosis and endogenous esoge...
Jpcr mauro luisetto et al amyotrophic lateral sclerosis and endogenous esoge...M. Luisetto Pharm.D.Spec. Pharmacology
 
clinical._pharmacology._ESSAY
clinical._pharmacology._ESSAYclinical._pharmacology._ESSAY
clinical._pharmacology._ESSAYDimitrios Brachos
 
Genomics and proteomics in drug discovery and development
Genomics and proteomics in drug discovery and developmentGenomics and proteomics in drug discovery and development
Genomics and proteomics in drug discovery and developmentSuchittaU
 
Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6
Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6
Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6Imhotep Virtual Medical School
 
SARCOSTEMMA ACIDUM
SARCOSTEMMA ACIDUMSARCOSTEMMA ACIDUM
SARCOSTEMMA ACIDUMrahees555
 
cloning. Second, it is sensitive. Activities canbe detected
cloning. Second, it is sensitive. Activities canbe detected cloning. Second, it is sensitive. Activities canbe detected
cloning. Second, it is sensitive. Activities canbe detected WilheminaRossi174
 
The mammalian target of rapamycin a cell key modulator of ageing
The mammalian target of rapamycin a cell key modulator of ageingThe mammalian target of rapamycin a cell key modulator of ageing
The mammalian target of rapamycin a cell key modulator of ageingXavier Viñals Gonzalez
 
Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...
Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...
Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...M. Luisetto Pharm.D.Spec. Pharmacology
 
Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...
Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...
Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...Sean Ekins
 

Similar to 1521-01033462318–327$25.00 httpdx.doi.org10.1124jpet.1 (20)

ISX9 - Karthik Gopalakrishnan
ISX9 - Karthik GopalakrishnanISX9 - Karthik Gopalakrishnan
ISX9 - Karthik Gopalakrishnan
 
Basic Formal Ontology (BFO) and Disease
 Basic Formal Ontology (BFO) and Disease Basic Formal Ontology (BFO) and Disease
Basic Formal Ontology (BFO) and Disease
 
Effect of Environmental Chemical Exposures on Epigenetics of Diseases: A Syst...
Effect of Environmental Chemical Exposures on Epigenetics of Diseases: A Syst...Effect of Environmental Chemical Exposures on Epigenetics of Diseases: A Syst...
Effect of Environmental Chemical Exposures on Epigenetics of Diseases: A Syst...
 
Omics technology: an important tool in mechanism studies of Chinese herbal fo...
Omics technology: an important tool in mechanism studies of Chinese herbal fo...Omics technology: an important tool in mechanism studies of Chinese herbal fo...
Omics technology: an important tool in mechanism studies of Chinese herbal fo...
 
Marfia_et_al-2014-Glia
Marfia_et_al-2014-GliaMarfia_et_al-2014-Glia
Marfia_et_al-2014-Glia
 
Geomics proteomics
Geomics proteomicsGeomics proteomics
Geomics proteomics
 
PUTATIVE DRUG TARGET IDENTIFICATION FOR SEPTIC ARTHRITIS THROUGH DATA MINING ...
PUTATIVE DRUG TARGET IDENTIFICATION FOR SEPTIC ARTHRITIS THROUGH DATA MINING ...PUTATIVE DRUG TARGET IDENTIFICATION FOR SEPTIC ARTHRITIS THROUGH DATA MINING ...
PUTATIVE DRUG TARGET IDENTIFICATION FOR SEPTIC ARTHRITIS THROUGH DATA MINING ...
 
Jpcr mauro luisetto et al amyotrophic lateral sclerosis and endogenous esoge...
Jpcr mauro luisetto  et al amyotrophic lateral sclerosis and endogenous esoge...Jpcr mauro luisetto  et al amyotrophic lateral sclerosis and endogenous esoge...
Jpcr mauro luisetto et al amyotrophic lateral sclerosis and endogenous esoge...
 
clinical._pharmacology._ESSAY
clinical._pharmacology._ESSAYclinical._pharmacology._ESSAY
clinical._pharmacology._ESSAY
 
Genomics and proteomics in drug discovery and development
Genomics and proteomics in drug discovery and developmentGenomics and proteomics in drug discovery and development
Genomics and proteomics in drug discovery and development
 
Proteomics
ProteomicsProteomics
Proteomics
 
Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6
Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6
Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6
 
neu%2E2012%2E2782
neu%2E2012%2E2782neu%2E2012%2E2782
neu%2E2012%2E2782
 
SARCOSTEMMA ACIDUM
SARCOSTEMMA ACIDUMSARCOSTEMMA ACIDUM
SARCOSTEMMA ACIDUM
 
cloning. Second, it is sensitive. Activities canbe detected
cloning. Second, it is sensitive. Activities canbe detected cloning. Second, it is sensitive. Activities canbe detected
cloning. Second, it is sensitive. Activities canbe detected
 
The mammalian target of rapamycin a cell key modulator of ageing
The mammalian target of rapamycin a cell key modulator of ageingThe mammalian target of rapamycin a cell key modulator of ageing
The mammalian target of rapamycin a cell key modulator of ageing
 
Mazalouskas_2015
Mazalouskas_2015Mazalouskas_2015
Mazalouskas_2015
 
Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...
Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...
Luisetto m et al amyotropyc lateral sclerosis and endogenous -esogenous toxic...
 
Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...
Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...
Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...
 
Herrmann STAT3 2008
Herrmann STAT3 2008Herrmann STAT3 2008
Herrmann STAT3 2008
 

More from MatthewTennant613

Assignment Application Adoption of New Technology SystemsAs a nu.docx
Assignment Application Adoption of New Technology SystemsAs a nu.docxAssignment Application Adoption of New Technology SystemsAs a nu.docx
Assignment Application Adoption of New Technology SystemsAs a nu.docxMatthewTennant613
 
Assignment Accreditation and Quality EnhancementThe purpose of ac.docx
Assignment Accreditation and Quality EnhancementThe purpose of ac.docxAssignment Accreditation and Quality EnhancementThe purpose of ac.docx
Assignment Accreditation and Quality EnhancementThe purpose of ac.docxMatthewTennant613
 
ASSIGNMENT AOperationsManagement- Y.docx
ASSIGNMENT AOperationsManagement- Y.docxASSIGNMENT AOperationsManagement- Y.docx
ASSIGNMENT AOperationsManagement- Y.docxMatthewTennant613
 
Assignment Adaptive ResponseAs an advanced practice nurse, you wi.docx
Assignment Adaptive ResponseAs an advanced practice nurse, you wi.docxAssignment Adaptive ResponseAs an advanced practice nurse, you wi.docx
Assignment Adaptive ResponseAs an advanced practice nurse, you wi.docxMatthewTennant613
 
Assignment 5 Senior Seminar Project Due Week 10 and worth 200 poi.docx
Assignment 5 Senior Seminar Project Due Week 10 and worth 200 poi.docxAssignment 5 Senior Seminar Project Due Week 10 and worth 200 poi.docx
Assignment 5 Senior Seminar Project Due Week 10 and worth 200 poi.docxMatthewTennant613
 
Assignment 5 Federal Contracting Activities and Contract Types Du.docx
Assignment 5 Federal Contracting Activities and Contract Types Du.docxAssignment 5 Federal Contracting Activities and Contract Types Du.docx
Assignment 5 Federal Contracting Activities and Contract Types Du.docxMatthewTennant613
 
Assignment 5 CrowdsourcingDue 06102017 At 1159 PMCrowdso.docx
Assignment 5 CrowdsourcingDue 06102017 At 1159 PMCrowdso.docxAssignment 5 CrowdsourcingDue 06102017 At 1159 PMCrowdso.docx
Assignment 5 CrowdsourcingDue 06102017 At 1159 PMCrowdso.docxMatthewTennant613
 
Assignment 4What are the power motivators of police leaders Expla.docx
Assignment 4What are the power motivators of police leaders Expla.docxAssignment 4What are the power motivators of police leaders Expla.docx
Assignment 4What are the power motivators of police leaders Expla.docxMatthewTennant613
 
Assignment 4Project ProgressDue Week 9 and worth 200 points.docx
Assignment 4Project ProgressDue Week 9 and worth 200 points.docxAssignment 4Project ProgressDue Week 9 and worth 200 points.docx
Assignment 4Project ProgressDue Week 9 and worth 200 points.docxMatthewTennant613
 
Assignment 4 PresentationChoose any federal statute that is curre.docx
Assignment 4 PresentationChoose any federal statute that is curre.docxAssignment 4 PresentationChoose any federal statute that is curre.docx
Assignment 4 PresentationChoose any federal statute that is curre.docxMatthewTennant613
 
Assignment 4 The Perfect ManagerWrite a one to two (1–2) page pap.docx
Assignment 4 The Perfect ManagerWrite a one to two (1–2) page pap.docxAssignment 4 The Perfect ManagerWrite a one to two (1–2) page pap.docx
Assignment 4 The Perfect ManagerWrite a one to two (1–2) page pap.docxMatthewTennant613
 
Assignment 4 Presentation Choose any federal statute that is cu.docx
Assignment 4 Presentation Choose any federal statute that is cu.docxAssignment 4 Presentation Choose any federal statute that is cu.docx
Assignment 4 Presentation Choose any federal statute that is cu.docxMatthewTennant613
 
Assignment 4 Inmates Rights and Special CircumstancesDue Week 8 a.docx
Assignment 4 Inmates Rights and Special CircumstancesDue Week 8 a.docxAssignment 4 Inmates Rights and Special CircumstancesDue Week 8 a.docx
Assignment 4 Inmates Rights and Special CircumstancesDue Week 8 a.docxMatthewTennant613
 
Assignment 4 Part D Your Marketing Plan – Video Presentation.docx
Assignment 4 Part D Your Marketing Plan – Video Presentation.docxAssignment 4 Part D Your Marketing Plan – Video Presentation.docx
Assignment 4 Part D Your Marketing Plan – Video Presentation.docxMatthewTennant613
 
Assignment 4 DUE Friday 72117 @ 1100amTurn in a written respon.docx
Assignment 4 DUE Friday 72117 @ 1100amTurn in a written respon.docxAssignment 4 DUE Friday 72117 @ 1100amTurn in a written respon.docx
Assignment 4 DUE Friday 72117 @ 1100amTurn in a written respon.docxMatthewTennant613
 
Assignment 4 Database Modeling and NormalizationImagine that yo.docx
Assignment 4 Database Modeling and NormalizationImagine that yo.docxAssignment 4 Database Modeling and NormalizationImagine that yo.docx
Assignment 4 Database Modeling and NormalizationImagine that yo.docxMatthewTennant613
 
Assignment 3 Inductive and Deductive ArgumentsIn this assignment,.docx
Assignment 3 Inductive and Deductive ArgumentsIn this assignment,.docxAssignment 3 Inductive and Deductive ArgumentsIn this assignment,.docx
Assignment 3 Inductive and Deductive ArgumentsIn this assignment,.docxMatthewTennant613
 
Assignment 3 Wireless WorldWith the fast-moving technology, the w.docx
Assignment 3 Wireless WorldWith the fast-moving technology, the w.docxAssignment 3 Wireless WorldWith the fast-moving technology, the w.docx
Assignment 3 Wireless WorldWith the fast-moving technology, the w.docxMatthewTennant613
 
Assignment 3 Web Design Usability Guide PresentationBefore you .docx
Assignment 3 Web Design Usability Guide PresentationBefore you .docxAssignment 3 Web Design Usability Guide PresentationBefore you .docx
Assignment 3 Web Design Usability Guide PresentationBefore you .docxMatthewTennant613
 
Assignment 3 Understanding the Prevalence of Community PolicingAs.docx
Assignment 3 Understanding the Prevalence of Community PolicingAs.docxAssignment 3 Understanding the Prevalence of Community PolicingAs.docx
Assignment 3 Understanding the Prevalence of Community PolicingAs.docxMatthewTennant613
 

More from MatthewTennant613 (20)

Assignment Application Adoption of New Technology SystemsAs a nu.docx
Assignment Application Adoption of New Technology SystemsAs a nu.docxAssignment Application Adoption of New Technology SystemsAs a nu.docx
Assignment Application Adoption of New Technology SystemsAs a nu.docx
 
Assignment Accreditation and Quality EnhancementThe purpose of ac.docx
Assignment Accreditation and Quality EnhancementThe purpose of ac.docxAssignment Accreditation and Quality EnhancementThe purpose of ac.docx
Assignment Accreditation and Quality EnhancementThe purpose of ac.docx
 
ASSIGNMENT AOperationsManagement- Y.docx
ASSIGNMENT AOperationsManagement- Y.docxASSIGNMENT AOperationsManagement- Y.docx
ASSIGNMENT AOperationsManagement- Y.docx
 
Assignment Adaptive ResponseAs an advanced practice nurse, you wi.docx
Assignment Adaptive ResponseAs an advanced practice nurse, you wi.docxAssignment Adaptive ResponseAs an advanced practice nurse, you wi.docx
Assignment Adaptive ResponseAs an advanced practice nurse, you wi.docx
 
Assignment 5 Senior Seminar Project Due Week 10 and worth 200 poi.docx
Assignment 5 Senior Seminar Project Due Week 10 and worth 200 poi.docxAssignment 5 Senior Seminar Project Due Week 10 and worth 200 poi.docx
Assignment 5 Senior Seminar Project Due Week 10 and worth 200 poi.docx
 
Assignment 5 Federal Contracting Activities and Contract Types Du.docx
Assignment 5 Federal Contracting Activities and Contract Types Du.docxAssignment 5 Federal Contracting Activities and Contract Types Du.docx
Assignment 5 Federal Contracting Activities and Contract Types Du.docx
 
Assignment 5 CrowdsourcingDue 06102017 At 1159 PMCrowdso.docx
Assignment 5 CrowdsourcingDue 06102017 At 1159 PMCrowdso.docxAssignment 5 CrowdsourcingDue 06102017 At 1159 PMCrowdso.docx
Assignment 5 CrowdsourcingDue 06102017 At 1159 PMCrowdso.docx
 
Assignment 4What are the power motivators of police leaders Expla.docx
Assignment 4What are the power motivators of police leaders Expla.docxAssignment 4What are the power motivators of police leaders Expla.docx
Assignment 4What are the power motivators of police leaders Expla.docx
 
Assignment 4Project ProgressDue Week 9 and worth 200 points.docx
Assignment 4Project ProgressDue Week 9 and worth 200 points.docxAssignment 4Project ProgressDue Week 9 and worth 200 points.docx
Assignment 4Project ProgressDue Week 9 and worth 200 points.docx
 
Assignment 4 PresentationChoose any federal statute that is curre.docx
Assignment 4 PresentationChoose any federal statute that is curre.docxAssignment 4 PresentationChoose any federal statute that is curre.docx
Assignment 4 PresentationChoose any federal statute that is curre.docx
 
Assignment 4 The Perfect ManagerWrite a one to two (1–2) page pap.docx
Assignment 4 The Perfect ManagerWrite a one to two (1–2) page pap.docxAssignment 4 The Perfect ManagerWrite a one to two (1–2) page pap.docx
Assignment 4 The Perfect ManagerWrite a one to two (1–2) page pap.docx
 
Assignment 4 Presentation Choose any federal statute that is cu.docx
Assignment 4 Presentation Choose any federal statute that is cu.docxAssignment 4 Presentation Choose any federal statute that is cu.docx
Assignment 4 Presentation Choose any federal statute that is cu.docx
 
Assignment 4 Inmates Rights and Special CircumstancesDue Week 8 a.docx
Assignment 4 Inmates Rights and Special CircumstancesDue Week 8 a.docxAssignment 4 Inmates Rights and Special CircumstancesDue Week 8 a.docx
Assignment 4 Inmates Rights and Special CircumstancesDue Week 8 a.docx
 
Assignment 4 Part D Your Marketing Plan – Video Presentation.docx
Assignment 4 Part D Your Marketing Plan – Video Presentation.docxAssignment 4 Part D Your Marketing Plan – Video Presentation.docx
Assignment 4 Part D Your Marketing Plan – Video Presentation.docx
 
Assignment 4 DUE Friday 72117 @ 1100amTurn in a written respon.docx
Assignment 4 DUE Friday 72117 @ 1100amTurn in a written respon.docxAssignment 4 DUE Friday 72117 @ 1100amTurn in a written respon.docx
Assignment 4 DUE Friday 72117 @ 1100amTurn in a written respon.docx
 
Assignment 4 Database Modeling and NormalizationImagine that yo.docx
Assignment 4 Database Modeling and NormalizationImagine that yo.docxAssignment 4 Database Modeling and NormalizationImagine that yo.docx
Assignment 4 Database Modeling and NormalizationImagine that yo.docx
 
Assignment 3 Inductive and Deductive ArgumentsIn this assignment,.docx
Assignment 3 Inductive and Deductive ArgumentsIn this assignment,.docxAssignment 3 Inductive and Deductive ArgumentsIn this assignment,.docx
Assignment 3 Inductive and Deductive ArgumentsIn this assignment,.docx
 
Assignment 3 Wireless WorldWith the fast-moving technology, the w.docx
Assignment 3 Wireless WorldWith the fast-moving technology, the w.docxAssignment 3 Wireless WorldWith the fast-moving technology, the w.docx
Assignment 3 Wireless WorldWith the fast-moving technology, the w.docx
 
Assignment 3 Web Design Usability Guide PresentationBefore you .docx
Assignment 3 Web Design Usability Guide PresentationBefore you .docxAssignment 3 Web Design Usability Guide PresentationBefore you .docx
Assignment 3 Web Design Usability Guide PresentationBefore you .docx
 
Assignment 3 Understanding the Prevalence of Community PolicingAs.docx
Assignment 3 Understanding the Prevalence of Community PolicingAs.docxAssignment 3 Understanding the Prevalence of Community PolicingAs.docx
Assignment 3 Understanding the Prevalence of Community PolicingAs.docx
 

Recently uploaded

POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptxPOINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptxSayali Powar
 
Arihant handbook biology for class 11 .pdf
Arihant handbook biology for class 11 .pdfArihant handbook biology for class 11 .pdf
Arihant handbook biology for class 11 .pdfchloefrazer622
 
Beyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global ImpactBeyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global ImpactPECB
 
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxSOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxiammrhaywood
 
Organic Name Reactions for the students and aspirants of Chemistry12th.pptx
Organic Name Reactions  for the students and aspirants of Chemistry12th.pptxOrganic Name Reactions  for the students and aspirants of Chemistry12th.pptx
Organic Name Reactions for the students and aspirants of Chemistry12th.pptxVS Mahajan Coaching Centre
 
Student login on Anyboli platform.helpin
Student login on Anyboli platform.helpinStudent login on Anyboli platform.helpin
Student login on Anyboli platform.helpinRaunakKeshri1
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13Steve Thomason
 
Disha NEET Physics Guide for classes 11 and 12.pdf
Disha NEET Physics Guide for classes 11 and 12.pdfDisha NEET Physics Guide for classes 11 and 12.pdf
Disha NEET Physics Guide for classes 11 and 12.pdfchloefrazer622
 
A Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy ReformA Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy ReformChameera Dedduwage
 
1029 - Danh muc Sach Giao Khoa 10 . pdf
1029 -  Danh muc Sach Giao Khoa 10 . pdf1029 -  Danh muc Sach Giao Khoa 10 . pdf
1029 - Danh muc Sach Giao Khoa 10 . pdfQucHHunhnh
 
Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)eniolaolutunde
 
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdfBASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdfSoniaTolstoy
 
The byproduct of sericulture in different industries.pptx
The byproduct of sericulture in different industries.pptxThe byproduct of sericulture in different industries.pptx
The byproduct of sericulture in different industries.pptxShobhayan Kirtania
 
Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityGeoBlogs
 
1029-Danh muc Sach Giao Khoa khoi 6.pdf
1029-Danh muc Sach Giao Khoa khoi  6.pdf1029-Danh muc Sach Giao Khoa khoi  6.pdf
1029-Danh muc Sach Giao Khoa khoi 6.pdfQucHHunhnh
 
Separation of Lanthanides/ Lanthanides and Actinides
Separation of Lanthanides/ Lanthanides and ActinidesSeparation of Lanthanides/ Lanthanides and Actinides
Separation of Lanthanides/ Lanthanides and ActinidesFatimaKhan178732
 
Q4-W6-Restating Informational Text Grade 3
Q4-W6-Restating Informational Text Grade 3Q4-W6-Restating Informational Text Grade 3
Q4-W6-Restating Informational Text Grade 3JemimahLaneBuaron
 
Advanced Views - Calendar View in Odoo 17
Advanced Views - Calendar View in Odoo 17Advanced Views - Calendar View in Odoo 17
Advanced Views - Calendar View in Odoo 17Celine George
 

Recently uploaded (20)

POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptxPOINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
 
Arihant handbook biology for class 11 .pdf
Arihant handbook biology for class 11 .pdfArihant handbook biology for class 11 .pdf
Arihant handbook biology for class 11 .pdf
 
Beyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global ImpactBeyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global Impact
 
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxSOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
 
Organic Name Reactions for the students and aspirants of Chemistry12th.pptx
Organic Name Reactions  for the students and aspirants of Chemistry12th.pptxOrganic Name Reactions  for the students and aspirants of Chemistry12th.pptx
Organic Name Reactions for the students and aspirants of Chemistry12th.pptx
 
Student login on Anyboli platform.helpin
Student login on Anyboli platform.helpinStudent login on Anyboli platform.helpin
Student login on Anyboli platform.helpin
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13
 
Disha NEET Physics Guide for classes 11 and 12.pdf
Disha NEET Physics Guide for classes 11 and 12.pdfDisha NEET Physics Guide for classes 11 and 12.pdf
Disha NEET Physics Guide for classes 11 and 12.pdf
 
A Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy ReformA Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy Reform
 
1029 - Danh muc Sach Giao Khoa 10 . pdf
1029 -  Danh muc Sach Giao Khoa 10 . pdf1029 -  Danh muc Sach Giao Khoa 10 . pdf
1029 - Danh muc Sach Giao Khoa 10 . pdf
 
Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)
 
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdfBASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
 
Advance Mobile Application Development class 07
Advance Mobile Application Development class 07Advance Mobile Application Development class 07
Advance Mobile Application Development class 07
 
The byproduct of sericulture in different industries.pptx
The byproduct of sericulture in different industries.pptxThe byproduct of sericulture in different industries.pptx
The byproduct of sericulture in different industries.pptx
 
INDIA QUIZ 2024 RLAC DELHI UNIVERSITY.pptx
INDIA QUIZ 2024 RLAC DELHI UNIVERSITY.pptxINDIA QUIZ 2024 RLAC DELHI UNIVERSITY.pptx
INDIA QUIZ 2024 RLAC DELHI UNIVERSITY.pptx
 
Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activity
 
1029-Danh muc Sach Giao Khoa khoi 6.pdf
1029-Danh muc Sach Giao Khoa khoi  6.pdf1029-Danh muc Sach Giao Khoa khoi  6.pdf
1029-Danh muc Sach Giao Khoa khoi 6.pdf
 
Separation of Lanthanides/ Lanthanides and Actinides
Separation of Lanthanides/ Lanthanides and ActinidesSeparation of Lanthanides/ Lanthanides and Actinides
Separation of Lanthanides/ Lanthanides and Actinides
 
Q4-W6-Restating Informational Text Grade 3
Q4-W6-Restating Informational Text Grade 3Q4-W6-Restating Informational Text Grade 3
Q4-W6-Restating Informational Text Grade 3
 
Advanced Views - Calendar View in Odoo 17
Advanced Views - Calendar View in Odoo 17Advanced Views - Calendar View in Odoo 17
Advanced Views - Calendar View in Odoo 17
 

1521-01033462318–327$25.00 httpdx.doi.org10.1124jpet.1

  • 1. 1521-0103/346/2/318–327$25.00 http://dx.doi.org/10.1124/jpet.113.202994 THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS J Pharmacol Exp Ther 346:318–327, August 2013 Copyright ª 2013 by The American Society for Pharmacology and Experimental Therapeutics Selective and Potent Agonists and Antagonists for Investigating the Role of Mouse Oxytocin Receptors Marta Busnelli, Elisabetta Bulgheroni, Maurice Manning, Gunnar Kleinau, and Bice Chini CNR, Institute of Neuroscience, Milan, Italy (M.B., E.B., B.C.); Department of Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy (M.B.); Department of Biochemistry and Cancer Biology, University of Toledo, Toledo, Ohio (M.M.); and Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, Berlin, Germany (G.K.) Received January 8, 2013; accepted May 29, 2013 ABSTRACT The neuropeptides oxytocin (OT) and vasopressin (AVP) have been shown to play a central role in social behaviors; as a consequence, they have been recognized as potential drugs to treat neurodevelopmental and psychiatric disorders character - ized by impaired social interactions. However, despite the basic and preclinical relevance of mouse strains carrying genetic
  • 2. alterations in the OT/AVP systems to basic and preclinical translational neuroscience, the pharmacological profile of mouse OT/AVP receptor subtypes has not been fully characterized. To fill in this gap, we have characterized a number of OT and AVP agonists and antagonists at three murine OT/AVP receptors expressed in the nervous system as follows: the oxytocin (mOTR) and vasopressin V1a (mV1aR) and V1b (mV1bR) subtypes. These three receptors were transiently expressed in vitro for binding and intracellular signaling assays, and then a homology model of the mOTR structure was constructed to investigate how its molecular features compare with human and rat OTR orthologs. Our data indicate that the selectivity profile of the natural ligands, OT and AVP, is conserved in humans, rats, and mice. Furthermore, we found that the synthetic peptide [Thr4Gly7]OT (TGOT) is remarkably selective for the mOTR and, like the endogenous OT ligand, activates Gq and Gi and recruits b-arrestins. Finally, we report three antagonists that exhibit remarkably high affinities and selectiv- ities at mOTRs. These highly selective pharmacological tools will contribute to the investigation of the specific physiologic and pathologic roles of mOTR for the development of selective OT-based therapeutics. Introduction Central oxytocin (OT) and vasopressin (AVP) effects are mediated by three G-protein-coupled receptors (GPCRs) that are evolutionarily highly conserved and closely related, with overall homology varying from 40 to 85%: the vasopressin 1a receptor (V1aR), the vasopressin 1b receptor (V1bR), and the OT receptor (OTR) (Barberis et al., 1999; Birnbaumer, 2000; Zingg and Laporte, 2003). OT and AVP are also structurally very similar, differing by only two amino acids in most
  • 3. mammals (Wallis, 2012). Given this high degree of conserva- tion in both receptors and peptides, the development of selective agonists and antagonists has proved to be a daunting task (Manning et al., 2008). Over the last decades, at least 1000 synthetic peptides have been synthesi zed and examined for their ability to bind to and activate the different OT/AVP receptor subtypes, an effort that has led to the identification of a number of subtype-selective analogs for human and rat subtypes (Manning et al., 2012). However, subtle differences between receptor sequences in different animal species are responsible for important changes in the selectivity profile of some ligands, and so the pharmacological data obtained in one species cannot be extrapolated tout court to others (Chini and Manning, 2007). In addition to their well established systemic physiologic effects, OT and AVP are potent modulators of social behavior; consistently, the OT/AVP system is emerging as a relevant target for the treatment of impaired social functions associ- ated with neurodevelopmental and psychiatric disorders (Miller, 2013). Even as OT has been recently reported to improve cognitive deficits in autistic patients (Modi and Young, 2012), its use is hampered by several of the following factors: 1) OT does not cross the blood-brain barrier and is administered intranasally, with unknown pharmacokinetics; 2) it also binds to and activates the V1aR and V1bR, which are highly expressed in the brain, where they exert different and even opposite effects (Pittman and Spencer, 2005); and 3) OT promotes the coupling of the OTR to different G proteins and This study was supported by Cariplo Foundation (Grant 2008.2314), Regione Lombardia (Progetto TerDisMental, ID 16983–Rif. SAL-50), and Telethon Foundation (Grant GGP12207).
  • 4. dx.doi.org/10.1124/jpet.113.202994. ABBREVIATIONS: AVP, vasopressin; BRET, bioluminescence resonance energy transfer; ECL, extracellular loop; GPCR, G- protein-coupled receptor; HTRF, homogeneous time-resolved fluorescence; ICL, intracellular loop; IP1, myoinositol 1-phosphate; LVA, linear vasopressin antagonist; OT, oxytocin; OTA, oxytocin receptor antagonist; OTR, oxytocin receptor; PBS, phosphate-buffered saline; RLuc, Renilla luciferase; SR49059, (2S)1-[(2R,3S)-1,5-chloro-3-(2-chlorophenyl)-1-(3,4- dimethoxybenzene-sulfonyl)-3-hydroxy-2,3-dihydro-1H-indole- 2-carbonyl]-pyrroli- dine-2-carboxamide; t1/2, half-time; TGOT, [Thr 4Gly7]OT; TMH, transmembrane helix; V1aR, vasopressin 1a receptor; V1bR, vasopressin 1b receptor; YFP, yellow fluorescent protein. 318 http://dx.doi.org/10.1124/jpet.113.202994 http://dx.doi.org/10.1124/jpet.113.202994 b-arrestins (Busnelli et al., 2012) and, consequently, activates multiple signaling pathways whose precise roles within the brain are currently unknown. The development of more selective, potent, and therefore, longer-lasting analogs acting on brain is a priority in the field to understand how (in terms of biologic mechanisms) and where (in terms of neural specificity) OT exerts its effects.
  • 5. In particular, subtype-selective analogs are crucial for defining the role of different receptor subtypes in rodent models currently used to investigate the OT/AVP system in the processing of socially relevant clues. Studies using trans - genic mice genetically engineered to eliminate OT, OTR, or CD38, a protein involved in OT secretion, show that these animals lose important social behaviors (Ferguson et al., 2000; Takayanagi et al., 2005; Jin et al., 2007; Higashida et al., 2011). The deficits in the social paradigm can be fully restored by a single intracerebroventricular infusion of OT given prior to the test (Ferguson et al., 2001; Jin et al., 2007; Sala et al., 2011). Although this effect is consistent with the genetic alteration in CD38- and OT-null mice, which have a decreased level of circulating (and centrally released) OT, the efficacy of OT in restoring social recognition in the OTR-null mice is particularly intriguing. We have recently shown that the social behavioral deficits associated with the complete loss of Oxtr gene expression can be rescued by the activation of cognate vasopressin receptors, thus suggesting that the OT/AVP brain systems have overlapping and/or compensatory functions (Sala et al., 2011). Another level of complexity in developing selective analogs derives from the finding that a single GPCR may couple to more than one G-protein, potentially activating multiple responses. Interestingly, different ligands show different degrees of in- trinsic efficacy to different signaling pathways activated by the same receptor, a phenomenon referred to as “functional se- lectivity” (Urban et al., 2007; Kenakin, 2011). Because func- tional selective ligands have been recently described in the OT/ AVP receptor family (in particular for the vasopressin 2 receptor (Jean-Alphonse et al., 2009), OTR (Reversi et al., 2005; Gravati et al., 2010; Busnelli et al., 2012), and V1aR (MacKinnon et al., 2009), the screening of the functional selective properties of
  • 6. ligands is becoming a crucial issue for the pharmacological characterization of selective ligands. The aim of this study was to pharmacologically characterize a number of OT/AVP analogs at the OT/AVP receptor sub- types expressed in mouse brain: mOTR, mV1aR, and mV1bR. We found that [Thr4Gly7]OT (TGOT) (Lowbridge et al., 1977) has a remarkable selectivity for the mouse OTR through which, like the endogenous OT ligand, it activates Gq and Gi and recruits b-arrestins. Furthermore, we identified several antagonists that exhibit remarkable selectivity profiles at mOTR. These analogs represent valuable tools to investigate the specific role of the mOTRs in the brain. Materials and Methods Reagents, Constructs, and Peptides. [3H]OT (NET-858, 30–60 Ci/mmol) and [3H]AVP (NET-800, 35–85 Ci/mmol) came from PerkinElmer Life and Analytical Sciences (Monza, Italy); coelenter- azine H from Invitrogen (Milan, Italy); DeepBlueC coelenterazine 400a (CLz400) from Biotium (Hayward, CA); OT, AVP, TGOT, and atosiban from Bachem (Weil am Rhein, Germany); and SR49059 [(2S)1- [(2R,3S)-1,5-chloro-3-(2-chlorophenyl)-1-(3,4- dimethoxybenzene- sulfonyl)-3-hydroxy-2,3-dihydro-1H-indole-2-carbonyl]- pyrrolidine-2- carboxamide] from sanofi-aventis (Toulouse, France). All of the other peptides used in this study were from the laboratory of M. Manning of the University of Toledo (Toledo, OH).
  • 7. The mouse V1aR cDNA (mV1aR) came from OriGene (Rockville, MD); the mouse V1bR (mV1bR) cDNA was a gift from M.A. Ventura (U-567; INSERM, Paris, France). The GFP10-Gg2 cDNA, in which the blue-shifted variant of Aequorea victoria (GFP10) was fused to Gg2, and the Gb1 cDNA are described in Gales et al. (2006). The Ga subunit expression vector cDNAs came from Missouri S&T cDNA Resource Center (Rolla, MO). The expression vector of b-arrestin2 fused at its C terminus to the yellow fluorescent protein (b-arrestin2–YFP) (originally developed in M. Bouvier’s laboratory) came from Dr. J. Perroy (IGF, Montpellier, France), and the expression vector for b-arrestin1–YFP came from C. Hoffmann of the University of Wuerzburg (Wuerzburg, Germany). The mOTR C-terminally fused to Renilla luciferase (mOTR- Rluc) was generated by subcloning the entire coding region of mOTR into an Rluc vector (PerkinElmer BioSignal, Inc., Monza, Italy). Cell Cultures. HEK293 and COS7 cells purchased from the American Type Culture Collection (Manassas, VA) were grown in Dulbecco’s modified Eagle’s medium (Sigma-Aldrich, Milan, Italy), supplemented with 10% fetal calf serum and 1% penicillin-
  • 8. streptomycin (Sigma-Aldrich) in a 10% CO2 humidified atmosphere at 37°C. Transfection. For the ligand binding assays, the COS7 cells were transfected by means of electroporation as previously described (Chini et al., 1995). For the homogeneous time-resolved fluorescence (HTRF) and bioluminescence resonance energy transfer (BRET) assays, HEK293 cells were seeded at a density of 3,100,000 cells/ well in 100-mm plates on the day before transfection. A mix containing 20 mg of DNA and 60 mg of polyethylenimine (PEI linear, MW 25000; Polysciences Europe GmbH, Eppelheim, Germany) was prepared with 1 ml of basic medium (without additives such as serum or antibiotics) and, after 15 minutes of incubation at room tem- perature, added directly to cells maintained in 10 ml of complete medium containing 10% fetal bovine serum. For the HTRF experi- ments, the cells were detached 24 hours after transfection and seeded with 100,000 cells/well in white half-area 96-well microplates (Corning Life Sciences, Amsterdam, The Netherlands). For the BRET experiments, the supplemented Dulbecco’s modified Eagle’s medium was renewed 24 hours after the transfections, and the cells were maintained in culture for an additional 24 hours before being washed twice, detached, and resuspended with phosphate-buffered saline
  • 9. (PBS)-MgCl2 (0.5 mM) at room temperature. Ligand Binding Assays. The binding assays were performed at 30°C on membranes prepared from COS7 cells as previously described (Chini et al., 1995). Compound affinities were determined by means of competition experiments in which the unlabeled compound concentrations varied from 10211 to 1025 M, and the concentration of the radioligand ([3H]OT for mOTR and [3H]AVP for mV1aR and mV1bR) was 2–4 � 1029 M. Nonspecific binding was determined in the presence of unlabeled OT or AVP (1023 M). The ligand binding data were analyzed by means of nonlinear regression using Prism version 5 (GraphPad Software, Inc., La Jolla, CA). The Ki values were calculated from the experimental IC50 values using the Cheng-Prusoff equation for a single population of competitive sites: Ki 5 IC50/[1 1 (L/Kd)], where L is the concentration of radioligand used in each experiment and the Kd values were as previously reported: OT Kd 5 0.54 nM for mOTR (Ring et al., 2010), AVP Kd 5 1.3 nM for mV1aR (Oshikawa et al., 2004), and AVP Kd 5 0.67 nM for mV1bR (Serradeil-Le Gal et al., 2007). All of the assays were performed in triplicate and repeated at least three times. BRET Assay. The interactions between mOTR and the different Ga subunits were analyzed by means of BRET2 experiments that use RLuc as the donor, the DeepBlueC coelenterazine derivative as
  • 10. its substrate, and GFP10 as the acceptor. HEK293 cells were cotrans- fected with mOTR-Rluc, GFP10-Gg2, or Gb1, without (-Ga) or with one Identification of Selective and Potent Analogs of Mouse OTR 319 of Gaq, Gai1, Gai2, Gai3, Gas, or Gao. Cells were incubated for 2 minutes with OT, TGOT, and PBS (untreated cells) before the addition of Rluc substrate, DeepBlueC; BRET2 was measured immediately after using an Infinite F500 reader plate (Tecan, Milan, Italy) that allows the sequential integration of light signals detected with two filter settings (Rluc, 370–450 nm; GFP10 filter, 510–540 nm). The BRET2 signal was calculated as the ratio between GFP10 emission and the light emitted by Rluc. The positive changes in BRET induced by the ligands indicated the closest interaction between the donor and the acceptor and were expressed on graphs as “BRET ligand effect” using the formula: (emission GFP10 ligand/emission Rluc ligand) 2 (emission GFP10 PBS/emission Rluc PBS). To analyze the kinetics of the mOTR–b-arrestin interactions, BRET1 experiments that use RLuc as the donor, coelenterazine H as
  • 11. its substrate, and YFP as the acceptor were performed. HEK293 cells were cotransfected with mOTR-Rluc and b-arrestin2–YFP or b- arrest- in1–YFP. The transfected cells were distributed in a white 96- well microplate (100 mg of proteins per well) (Optiplate; PerkinElmer Life and Analytical Sciences) and incubated in the presence or absence of ligands. Coelenterazine H was added 8 minutes before the addition of the different ligands, and readings were made for 10 minutes using an Infinite F500 reader plate (Tecan) and filter set (Rluc filter, 370–480 nm; YFP filter, 520–570 nm). To determine the half-time (t1/2) of OT- and other ligand-induced BRET, the data were recorded as the difference between the ligand-promoted BRET signal and the average of the baseline (PBS-treated) BRET signal, and the time at which the half-BRET peak was reached was estimated. Inositol Phosphate Measurements. Myoinositol 1-phosphate (IP1) accumulation in HEK293 cells transiently transfected with mOTR, mV1aR, and mV1bR (100,000 cells) was determined in 96-well half-area microplates (Corning Life Sciences) using the HTRF- IP-One Kit (CisBio International, Bagnols-sur-Cèze, France) after 1 hour of stimulation with increasing concentrations of OT, AVP, and TGOT at 37°C. The time-resolved fluorescence resonance energy transfer
  • 12. signals were measured 50 ms after excitation at 620 and 665 nm using a Tecan Infinite F500 instrument. The IP1 concentrations were interpolated from the IP1 standard curve supplied with the kit. Statistical Analysis. All of the data were analyzed using Graph- Pad Prism software, version 5. Data from radioligand binding were evaluated by the nonlinear, least-squares curve-fitting procedure. Concentration-response IP1 curves were analyzed by means of nonlinear curve fitting using the sigmoidal dose-response equation. Parameters errors (Ki and EC50) are all expressed in %CV and cal- culated by simultaneous analysis of at least three different experi- ments performed in triplicate. Ki comparison has been performed on the basis of the F test for the extra sum of squares principle (*P , 0.05; **P , 0.01; ***P , 0.001). Ligand-induced BRET ratios are expressed as mean 6 S.E.M and were analyzed with one-way analysis of variance followed by Tukey’s post hoc test to determine statistically significant differences in treatments (***P , 0.001). The BRET1 kinetics data were normalized by setting the zero time point immediately after the addition of the ligand, and the data were analyzed by means of nonlinear least-squares fitting to the one- phase exponential association equation. Homology Modeling of the mOTR Structure. A large number of GPCR crystal structures in different activity-state-related
  • 13. con- formations have been published in recent years (Zhao and Wu, 2012), most of them cocrystallized with specific ligands (agonists or antagonists) (Kobilka and Schertler, 2008; Hanson and Stevens, 2009). Therefore, they serve as optimal templates for family A GPCR homology modeling (OTRs are members of family A GPCRs) with the purpose to study potential details of ligand binding or signal transduction. Based on high sequence similarity and overlapping structural features in the transmembrane helices (TMHs), the b2- adrenergic receptor crystal structure in an active conformation was used here as a template (Protein Data Bank code 3SN6) (Rasmussen et al., 2011) for modeling of the murine OTR. The general modeling procedure (sequence alignment, crystal structure preparation for modeling, side chain substitutions) was performed as recently described (Costanzi, 2012). In addition, extracellular loop (ECL) 2 of the b2-adrenergic receptor was partially deleted, because of significant differences in length and amino acid composition (biophysical properties) compared with the sequence of mOTR ECL2. According to advanced insights from previous GPCR modeling studies [supplemental material in
  • 14. Michino et al. (2009)], only the C-terminal part of the template ECL2 structure, which includes the highly conserved cysteine bridge to TMH3, was kept. Other mOTR ECL2 residues, most likely not involved in direct constitution of the ligand binding region, were manually added as spacers. The putative general OTR ligand binding region, located between the extracellular ends of the TMHs and the ECLs, was defined based on similarity to the ligand binding regions of known GPCR crystal structures (Deupi and Standfuss, 2011; Kratochwil et al., 2011; Wichard et al., 2011; Jacobson and Costanzi, 2012). The OTR model is constituted by amino acids from positions Glu36 to Leu344 (the extracellular N terminus and intracellular C terminus are not included because of missing structural templates). For modeling procedures, the Sybyl-X 2.0 version (Tripos International, St. Louis, MO) was used. Gaps of missing residues in the loops of the template structure were closed manually by adding OTR- specific amino acids. Side chains were subjected to conjugate gradient minimizations (until converging at a termination gradient of 0.1 kcal/mol/Å) and molecular dynamics simulation (2 ns) by fixing the backbone of the TMHs. Finally, the model was minimized without
  • 15. constraints using the AMBER 7 forcefield. Structure images were produced using the PyMOL software (PyMOL Molecular Graphics System, version 1.3; Schrödinger, LLC, Portland, OR). Results Binding Affinities of Commonly Used OT/AVP Ana- logs at Mouse Receptors. The ligand binding properties of commonly used OT/AVP analogs at mOTR, mV1aR, and mV1bR were determined by competition experiments; calcu- lated Ki values 6 %CV are reported in Table 1. With regard to the peptides with agonist activity, the endogenous OT and AVP ligands had very different selectivity profiles (Fig. 1, A and B). AVP bound to the three brain- expressed OT/AVP receptors with almost identical affinity (Ki values for OTR, V1aR, and V1bR of, respectively, 0.87 nM 6 8% CV, n 5 3; 1.11 nM 6 27% CV, n 5 4; and 0.43 nM 6 12% CV, n 5 4), whereas OT had a receptor-specific affinity range that was highest for OTR (Ki 5 0.83 nM 6 17% CV, n 5 4) and lower for V1aR (Ki 5 20.38 6 26% CV, n 5 5) (P , 0.001 versus mOTR) and V1bR (Ki 5 36.32 nM 6 7% CV, n 5 4) (P , 0.001 versus mOTR). The dLVT peptide agonist binds with significantly different Ki values for OTR, V1aR, and V1bR of, respectively, 0.43 nM 6 20% CV, n 5 5; 3.39 nM 6 28% CV, n 5 5 (P , 0.001 versus mOTR); and 0.82 nM 6 7% CV, n 5 3 (P , 0.01 versus mOTR) (Fig. 1C). However, we should mention here that a significant difference in Ki is not sufficient to define ligand selectivity. As an operational criterion, it has been proposed that, to be “selective” for a particular subtype, any ligand should display a Ki at least 2 orders of magni tude lower than that for the other receptor subtypes (Chini et al., 2008). On these premises, the only agonist provided with a good selectivity profile is TGOT, with a Ki of 0.04 nM 6 32%
  • 16. CV, n 5 5, for OTR and Ki values of .1000 nM for V1aR and V1bR (Fig. 1D). Among the Gq OTR antagonists (OTAs) that we analyzed, whose original synthesis and pharmacological properties are 320 Busnelli et al. http://www.pdb.org/pdb/explore/explore.do?structureId=3SN6 reviewed in Manning et al. (2008, 2012), atosiban, OTA2, and OTA3 were selective for mOTR (Fig. 2, A, C, and D), with affinities in the nanomolar range (Ki 5 1.29 nM 6 46% CV, n 5 4 for atosiban; Ki 5 0.27 nM 6 25% CV, n 5 4 for OTA2; and Ki 5 1.24 nM 6 36% CV, n 5 4 for OTA3), and showing Ki values for V1aR and V1bR that were .1000-fold higher (P , 0.001). The widely used OTR antagonist OTA1 also had a good selectivity profile (Fig. 2B), with highest affinity for OTR (Ki 5 0.13 nM 6 42% CV, n 5 5), intermediate for V1aR (Ki 5 34.3 nM 6 33% CV, n 5 4) (P , 0.001), and lowest for V1bR (Ki 5 374 nM 6 20% CV, n 5 5) (P , 0.001). We finally analyzed three compounds commonly used as V1aR-selective antagonists: LVA (linear vasopressin antago- nist), the Manning compound, and the nonpeptidic antagonist SR49059 [reviewed in Manning et al. (2008, 2012)]. As shown in Fig. 3, A–C, they bound with different affinities to mouse OTR, V1aR, and V1bR. The three antagonists bounds with significantly different Ki values to OTR and V1aR (P , 0.001); SR49059 also had a significantly different Ki value for V1bR (P , 0.001 versus mOTR). However, none of them had a good selectivity profile for mouse OTR, V1aR, and V1bR, as their Ki affinities for the V1aR were at most 10–20 times lower than those for OTR (Table 1). Coupling Properties of Commonly Used OT/AVP
  • 17. Analogs at Mouse Receptors. As mOTR, mV1aR, and mV1bR are all coupled to Gq, leading to phospholipase C activation, inositol phosphate production, and an increase in intracellular calcium, we assayed the efficacy of the agonists by drawing up concentration-response curves of IP1 pro- duction at the three receptor subtypes. Our results indicate that OT activated mOTR with an EC50 of 4.45 nM 6 31% CV, n 5 3; and mV1aR and mV1bR with similar, lower, EC50 values (171 nM 6 19% CV, n 5 3; and 87 nM 6 45% CV, n 5 3) (Fig. 4A), displaying a good selectivity profile. On the contrary, AVP activates mV1aR, mV1bR, and mOTR with decreasing potency (Fig. 4B), with calculated EC50 values of, respectively, 0.65 nM 6 89% CV, n 5 3; 6.62 nM 6 32% CV, n 5 3; and 47.9 nM 6 69% CV, n 5 3. There were no significant differences between OT and AVP Emax in the three receptor subtypes. In comparison with OT, the highly selective analog TGOT was characterized by a left-shifted dose-response curve (Fig. 4C), as expected on the basis of its high binding affinity. The calculated EC50 of TGOT for mOTR was 0.18 nM 6 83% CV, n 5 3; whereas no IP1 production was observed for mV1aR and mV1bR also at very high peptide doses (.1000 nM), which is in accordance with its low affinity for these receptor subtypes. The functional selective properties of TGOT on mOTR coupling were investigated by means of a BRET2-based assay in which the energy donor RLuc is fused to the C-terminal end of mOTR cDNA and GFP10 used as the acceptor is N- terminally fused to the Gg2 subunit (GFP 10-Gg2). As shown in Fig. 5, upon OT (1025 M) and TGOT (1025 M) binding, TABLE 1
  • 18. Amino acid sequences and affinity values (Ki) of the investigated ligands Substitutions and/or modifications of the amino acid sequence of OT are indicated in boldface type; the superscript numbers indicate the position of the residue in the peptide sequence. Analog Sequence Ki for: mOTR mV1a mV1b nM 6 %CV OT Cys1, Tyr2, Ile3, Gln4, Asn5, Cys6, Pro7, Leu8, Gly-NH2 9 0.83 6 17 20.38 6 26 36.32 6 7 AVP Cys1, Tyr2, Phe3, Gln4, Asn5, Cys6, Pro7, Arg8, Gly-NH2 9 0.87 6 8 1.11 6 27 0.43 6 12 dLVT dCys1, Tyr2, Ile3, Gln4, Asn5, Cys6, Pro7, Lys8, Gly- NH2 9 0.43 6 20 3.39 6 28 0.82 6 7 TGOT Cys1, Tyr2, Ile3, Thr4, Asn5, Cys6, Gly7, Leu8, Gly- NH2 9 0.04 6 32 .1000 .10,000 Atosiban dCys1, DTyr(Et)2, Ile3, Thr4, Asn5, Cys6, Pro7, Orn8, Gly-NH2 9 1.29 6 46 .1000 .1000 OTA1 d(CH2)5 1, Tyr(Me)2, Ile3, Thr4, Asn5, Cys6, Pro7, Orn8, Tyr-NH2 9 0.13 6 42 34.3 6 33 374.9 6 20
  • 19. OTA2 desGly-NH2,d(CH2)5 1, Tyr(Me)2, Ile3, Thr4, Asn5, Cys6, Pro7, Orn8 0.27 6 25 232 6 48 .10,000 OTA3 desGly-NH2,d(CH2)5 1, DTyr2, Ile3, Thr4, Asn5, Cys6, Pro7, Orn8 1.24 6 36 .10,000 .10,000 LVA Phenylac1, DTyr(Me)2, Phe3, Gln4, Asn5, Arg6, Pro7, Arg8 3.90 6 30 0.10 6 19 9.66 6 12 Manning compound d(CH2)5 1, Tyr(Me)2, Phe3, Gln4, Asn5, Cys6, Pro7, Arg8, Gly-NH2 9 42.6 6 18 1.72 6 28 73.87 6 9 SR49059 13.2 6 19 0.94 6 22 97.35 6 12 d, deamino; d(CH2)5, b-mercapto-b,b- penthamethylenepropionic; desGly-NH2, desglycineamide; DTyr(Et), O-ethyl-D-tyrosine; Orn, ornithine; Phenylac, phenylacetyl; Tyr (Me), O-methyltyrosine. Fig. 1. Binding properties of OT/AVP agonists at mOTR, mV1aR, and mV1bR. Competition binding experiments were performed using increasing concentrations (from 10211 to 1025 M) of the endogenous ligands OT (A) and AVP (B) and the synthetic ligands dLVT (C) and TGOT (D). Ligand binding was determined on membrane preparations of COS7 cells transiently transfected with mOTR (black circles), mV1aR (blue triangles), and mV1bR (red squares). Specific binding was determined in the presence of 2– 4 � 1029 M [3H]OT for mOTR and [3H]AVP for mV1aR and mV1bR; nonspecific binding was determined in the presence of OT or AVP (1023 M). Each
  • 20. curve is the mean of triplicate determinations of a single representative experiment. Identification of Selective and Potent Analogs of Mouse OTR 321 mOTR significantly (P , 0.001 versus PBS) recruits Gq, Gi1, Gi2, Gi3, and G0, but not Gs, thus confirming its coupling to the same G-protein subtypes recruited by OT (Busnelli et al., 2012). Finally, to investigate whether TGOT induces b-arrestin recruitment after receptor activation, we used a “real -time” BRET1 assay that uses the mOTR-RLuc construct as the energy donor and b-arrestin2–YFP or b-arrestin1–YFP as the acceptor (Fig. 6). In cells coexpressing mOTR-Rluc and b-arrestin2–YFP or b-arrestin1–YFP, OT at a final concen- tration of 10 mM increased the BRET ratio with t1/2 values of, respectively, 89.9 6 2.3 seconds (n 5 3) and 101 6 6.1 seconds (n 5 3); similarly, TGOT at the same concentration increased the BRET ratio with t1/2 values of, respectively, 124.6 6 4.2 seconds (n 5 3) and 121.9 6 5.1 seconds (n 5 3). Moreover, the BRET ratio remained stable for at least 10 minutes (Fig. 6), thus indicating a sustained agonist-induced association between mOTR and b-arrestins. Insights into Molecular Differences between OTR Subtypes Based on a Structural Model. To evaluate differences in amino acid composition among mouse, rat, and human OTR subtypes, we first performed an alignment of their amino acid sequences (Fig. 7). Secondly, we designed a three-dimensional homology model of a putative mOTR conformation to analyze the spatial distribution of different
  • 21. amino acids at corresponding positions and to study structural- functional features of the mOTR (Fig. 8). The sequence alignment shows that several not highly conserved amino acids are distributed over the entire receptor structure, in particular in the N-terminal region and C-terminal regions (Fig. 7). In a previous study, it was shown that truncation of the first 32 residues of the N terminus of the hOTR did not influence OT binding (Wesley et al., 2002), and the only residue in the N terminus found to be relevant for high- affinity OT binding was the conserved arginine at position 34 (Wesley et al., 2002). Variations in the N terminus are thus unlikely to be involved in determining TGOT high-affinity binding to the mOTR. The three-dimensional visualization of the mouse OTR serpentine domain (TMHs and loops) shows that Val201 in TMH5 (hOTR: isoleucine, rOTR: valine), Val301 in ECL3 (hOTR: alanine, rOTR: valine) and Ala313 in TMH7 (hOTR: valine, rOTR: alanine) are located in close spatial proximity to the putative ligand binding region (Fig. 8). Based on our model, only Val201 directly participates in the determination of the ligand pocket properties (such as the shape) and biophysical parameters. At this position, an isoleucine is located in the hOTR, which is different in bulkiness and length compared with the rodent valine. Alanine at position 159 (hOTR: alanine; rOTR: glycine) and valine at position 169 (hOTR: alanine; rOTR: alanine), located at TMH4, are outside the ligand binding pocket. Residue Phe51 inTMH1 (hOTR: leucine, rOTR: phenylalanine) points toward the membrane without any intramolecular interaction, Fig. 2. Binding properties of commonly used OTAs at mOTR, mV1aR, and mV1bR. Competition binding experiments were performed using increasing
  • 22. concentrations (from 10211 to 1025 M) of the Gq antagonists atosiban (A), OTA1 (B), OTA2 (C), and OTA3 (D). Ligand binding was determined on membrane preparations of COS7 cells transiently transfected with mOTR (black circles), mV1aR (blue triangles), and mV1bR (red squares). Specific binding was determined in the presence of 2–4 � 1029 M [3H]OT for mOTR and [3H]AVP for mV1aR and mV1bR; nonspecific binding was determined in the presence of OT or AVP (1023 M). Each curve is the mean of triplicate determinations of a single representative experiment. Fig. 3. Binding properties of commonly used V1a antagonists at mOTR, mV1aR, and mV1bR. Competition binding experiments were performed using increasing concentrations (from 10211 to 1025 M) of three compounds commonly used as selective V1aR antagonists: LVA (A), the Manning compound (B), and SR49059 (C). Ligand binding was determined on membrane preparations of COS7 cells transiently transfected with mOTR (black circles), mV1aR (blue triangles), and mV1bR (red squares). Specific binding was determined in the presence of 2–4 � 1029 M [3H]OT for mOTR and [3H]AVP for mV1aR and mV1bR; nonspecific binding was determined in the presence of OT or AVP (1023 M). Each curve is the mean of triplicate determinations of a single representative experiment. 322 Busnelli et al. and His69 (hOTR: glutamine; rOTR: histidine) is located at intracellular loop 1 (ICL1). Therefore, they should not have a direct impact on ligand binding, even though indirect effects
  • 23. due to changes in intrinsic signaling capacity (e.g., helix movement flexibility) cannot be excluded. Discussion We describe the in vitro pharmacological characterization of several analogs of mouse OTR, V1a, and V1b receptors, the three OT/AVP receptor subtypes expressed in mammalian brain. Peptidic and nonpeptidic OT/AVP analogs have pri- marily been assayed for their agonistic and antagonistic activities in in vitro and in vivo assays based on the peripheral effects of OT/AVP receptors, with OTR activities being quantified on the basis of myometrial contractility, V1aR activities on the basis of vasoconstriction, V1bR activities on the basis of adrenocorticotropin release, and vasopressin 2 receptor activities on the basis of antidiuresis. Much fewer pharmacological data have been collected concerning the selective effects of these analogs within the brain, where their use at very high doses has often led to conflicting or in- consistent results (Engelmann et al., 1996). A systematic analysis of the affinity and efficacy of OT/AVP compounds in selected brain areas would therefore be extremely valuable in preclinical research. However, this approach is hindered by a number of technical issues. First of all, OT/AVP receptors are not highly expressed in brain, and secondly, tritiated OT and AVP radiotracers have low specific activity. Overcoming these limitations would require tissue enrichment procedures to obtain consistent and reproducible results (Elands et al., 1988), but this would involve the use of a large number of animals, increase costs, and raise ethical concerns. Conse- quently, we believe that the in vitro characterization of transfected receptors represents a preliminary step for the selection of candidate drugs to be tested in vivo. Our in vitro results indicate that, as observed in other
  • 24. animal species, endogenous OT and AVP ligands have different selectivity profiles for mouse OT/AVP receptors. AVP binds to the three brain-expressed OT/AVP receptors with almost identical affinity, but activates the mV1aR, mV1bR, and mOTR with decreasing potency over 2 orders of magnitude. On the contrary, OT has a receptor-specific affinity range that is highest for OTR and lower for V1aR and V1bR, and this correlates with its potency at the sites of the three receptor subtypes. Depending on the dose and site of administration, some of the actions of AVP may be mediated via the OTR, and OT can bind and activate V1aR and V1bR expressed in the brain, albeit with lower affinity than AVP itself, which means that exogenously administered OT agonists can elicit substantial responses by binding to AVP receptors. On the other hand, a low AVP dose may also act as a “competitive antagonist” at the mOTR, and particularly the Gq-mediated pathway, which is activated with a high EC50. Fig. 4. Receptor/Gq coupling properties of OT/AVP analogs determined by means of IP1 inositol phosphate production. IP1 production was measured using an immune-competitive HTRF-based assay in HEK293 cells transiently transfected with mOTR (black circles), mV1aR (blue triangles), and mV1bR (red squares). A total of 100,000 cells were stimulated for 30 minutes with increasing concentrations (10214 to 1025 M) of OT (A), AVP (B), and TGOT (C). Each curve is the mean of triplicate determinations of a single representative experiment. Fig. 5. TGOT-induced G-protein recruitment determined by means of a BRET-based assay. BRET2 was measured in HEK293 cells transiently transfected with mOTR-RLuc, GFP10-Gg2, and Gb1 in the absence (-Ga) or presence of the indicated Ga subunits. The data represent the differences in
  • 25. BRET signals between the specified BRET partners in the absence (PBS; empty bars) or presence of OT (1025 M; black bars) and TGOT (1025 M; red bars), and are expressed as the mean value 6 S.E.M. of three independent assays performed in triplicate. One-way analysis of variance followed by Tukey’s test was used to determine the statistical differences between treatments. ***P , 0.001 vs. untreated controls (PBS). Identification of Selective and Potent Analogs of Mouse OTR 323 Moreover, the fact that AVP is devoid of agonist activity upon OTR-mediated Gi1, GoA, and GoB activation (Busnelli et al., 2012) has still undefined pharmacological implications. In this study, we show that the synthetic peptide TGOT has remarkable OTR-versus-V1aR/V1bR selectivity in terms of affinity binding and coupling for mice receptors. TGOT was originally demonstrated to be highly selective for rat OTR by means of in vivo bioassays (Lowbridge et al., 1977). However, this enhanced OTR/V1a selectivity is lost in humans, in whom the affinities of OT and TGOT to OTR and V1a receptors are comparable, thus indicating that the use of TGOT does not have any advantage over OT as far as OTR/V1a selectivity is concerned (Chini and Manning, 2007). As shown in Table 2, comparison of OT and TGOT affinities for human, rat, and mouse OTRs indicates that OT has the same affinity in the three species, whereas the affinity of TGOT increases by a factor of 100 going from human to rat to mouse. Concerning its coupling features, TGOT binding to the OTR led to the activation of Gq and all the members of the Gi and Go family exactly as the endogenous OT ligand (Busnelli et al., 2012).
  • 26. This is particularly relevant in neuronal cells, where it has been shown that OTR coupling to Gq and Gi/Go results in opposite effects on cell excitability via inhibition or activation of potassium channels (Gravati et al., 2010). Furthermore, Fig. 6. TGOT-mediated b-arrestin1 and b-arrestin2 recruitment. BRET1 was monitored in HEK293 cells transiently transfecte d with mOTR-RLuc and b-arrestin1–YFP or b-arrestin2–YFP. The cells were stimulated with OT (1025 M; black squares) or TGOT (1025 M; red circles). Real-time BRET1 measurements were made every 20 seconds for 10 minutes. The data represent the differences in BRET signals between the specified BRET partners in the absence or presence of the OT and TGOT agonist. Each curve is the mean of triplicate determinations of a single representative experiment. Fig. 7. Sequence alignment of human, rat, and mouse OTR. The sequences of the human, rat, and mouse OTR orthologs are aligned to each other. Asterisks indicate potential TMHs numbered from 1 to 7 and helix 8. Positions of varying amino acids between the OTR subtypes are highlighted with black shadows, whereby the three positions of side chain variations that are in close spatial proximity to the putative ligand binding pocket are highlighted with red shadows. 324 Busnelli et al. TGOT promotes b-arrestin1 and b-arrestin2 recruitment as efficiently as OT, suggesting similar desensitization and in-
  • 27. ternalization properties. Concerning our pharmacological screening for mOTR- selective antagonists, three peptides were shown to be very selective: atosiban, OTA2, and OTA3. In our hands, the most promising antagonist is OTA3, which we found to be highly selective for mOTR. Unfortunately, among the V1aR antag- onists tested, we didn’t identify any ligand with a Ki for the murine V1a receptor subtype at least 2 orders of magnitude lower than that for the other two receptor subtypes, a condition previously set as the minimal requirement for a selective ligand (Chini et al., 2008). The Manning compound, originally described as a potent V1aR-selective antagonist in rat, also was found to be not selective in humans (Manning et al., 2012) or in mice (this study) and can’t be used as a selective V1a antagonist in these species. Revealing variations in ligand binding and signaling among OTR orthologs in in vitro models has an impact on various aspects of OT/AVP pharmacology, including the identification of pharmacological tools that can be used in single species of particular translational interest, such as genetically modified mouse models. Our study identified analogs that lack selectivity for human receptors but are highly selective for mouse OTRs and therefore very valuable for preclinical studies. However, as we used transfected cells, one major issue is to verify whether the selectivity profile observed in transfected cells is maintained in vivo. In this regard, when used in mice at a dose of 0.0008 ng/animal, OTA3 specifically blocked the mOTR-mediated rescue of sociability defects in heterozygous Oxtr1/2 animals, suggesting the validity of this approach to identify mOTR antagonists (Sala et al., 2013). TGOT has been previously used in mice mainly in electro- physiology experiments, which found evidence of its selectiv- ity for OTR- versus V1a-mediated responses (Huber et al., 2005; Gozzi et al., 2010). However, in slices, the effective dose
  • 28. was very similar to the OT doses used, and the half-maximally effective concentration was only slightly more potent than OT (Huber et al., 2005). A similar discrepancy between in vitro and in vivo potencies of TGOT was also observed in social behavioral rescue experiments recently performed in OTR- null mice (Sala et al., 2011, 2013). TGOT rescued the social deficit at a dose of 0.0005 ng/animal in Oxtr1/2 mice, which is consistent with a selective action of TGOT through OTRs. However, at a dose of 0.05 ng/animal, TGOT also rescued the social deficit of Oxtr2/2 mice, suggesting that despite its very low affinity for the V1a and V1b receptors in vitro, TGOT was still active on these receptors in vivo (Sala et al., 2013). Several factors may be responsible for the discrepancy in TGOT potency observed in vitro and in vivo. Diffusion and enzymatic degradation may greatly affect peptides’ stability in tissues. In the brain, the aminopeptidase oxytocinase hydrolyzes OT, AVP, enkephalins, and other neuropeptides. The enzyme is present in soluble and membrane-bound forms, and its distribution varies greatly in different brain regions (Fernando et al., 2005). In addition, binding affinity and velocity of catalysis for different substrates could also account for significant differences in local neuropeptide concentra- tions and final neurobiological effects. Finally, studying the pharmacology of receptor orthologs may contribute to optimizing the design of selective analogs because subtle differences in ortholog activation can reveal crucial ligand-receptor interactions involved in binding and activation processes. As the conservation of the OTR sequence in the three species is very high (.90%), it should be feasible, Fig. 8. Structural homology model of the monomeric mouse OTR. The structural model (active conformation) of the mOTR (backbone cartoon, white) is represented without the N-terminal extracellular part
  • 29. (Ntt), the intracellular tail (Ctt), and the middle portion of ICL3. The potential ligand binding region is highlighted by an inner surface (green). This pocket-like crevice is constituted by specific amino acids (green lines) located at the ECLs and the TMHs toward the extracellular site. This three-dimensional representation of the OTR is helpful to identify potential links between functional differences of the receptor subspecies with particular residue variations. Amino acid positions that are not conserved between rodent and human OTR are shown as sticks (red and brown, labeled). Amino acids Val201 (TMH5), Val301 (ECL3), and Ala313 (TMH7) (red sticks) are located in close spatial proximity to the ligand binding region, whereby only Val201 is a direct determinant of the main ligand binding pocket. Alanine at position 159 and Val169 (TMH4) as well as Phe51 (TMH1) or His69 (ICL1) are outside the putative ligand binding site, and side chain variations should not have direct influences on ligand binding properties. TABLE 2 Affinity constants of OT and TGOT for the human, rat, and mouse OTR Ki (mean 6 S.D.) for:
  • 30. Human OTR Rat OTR Mouse OTR nM OT 0.79 6 0.22a 1.0 6 0.1b 0.83 6 0.14c TGOT 6.62 6 1.22d 0.8 6 0.2b 0.04 6 0.01c a From Chini et al. (1995). b From Elands et al. (1988). c From the current study. d From Chini et al. (1996). Identification of Selective and Potent Analogs of Mouse OTR 325 in principle, to identify the variable receptor residue(s) responsible for differences in affinity binding among the three species. By analyzing an OTR ortholog sequence alignment (Fig. 7) and by molecular modeling of mOTR (Fig. 8), we explored the distribution and potential relevance of nonconserved residues to high agonist binding. Most of the substitutions among the subspecies are located at the N terminus and in ICL3. So far as is known, these receptor parts have not directly related to ligand affinity and selectivity in OTR species (Wesley et al., 2002). Three varying positions are located in close spatial proximity to the putative ligand binding region, whereby only the residue at TMH5 directly participates in the constitution of the ligand binding pocket. However, at none of these positions is present a residue that is different in all the three species, suggesting that the increased TGOT affinity observed in rodent OTR probably does not result from a single substitution but is more likely due to the
  • 31. combination of particular variation(s) close to the ligand binding site and/or at other distinct receptor parts. With respect to this topic, it will be of future interest to explore the effect of multiple amino-acid substitutions in OTR subspecies. In conclusion, our results indicate that the selectivity profile of OTR/V1a/V1b receptors for the natural OT and AVP ligands is conserved in humans and rodents (rats and mice). However, subtle differences between receptor orthologs are responsible for an increase in the affinity of the synthetic agonist TGOT for mOTR. We also identified a number of OTAs characterized by very high selectivity for mOTR. TGOT and OTAs are therefore valuable molecular tools for in- vestigating specific mOTR-mediated effects. Authorship Contributions Participated in research design: Busnelli, Chini. Conducted experiments: Busnelli, Bulgheroni, Kleinau. Contributed reagents: Manning. Performed data analysis: Busnelli, Bulgheroni, Kleinau, Chini. Wrote or contributed to the writing of the manuscript: Busnelli, Manning, Kleinau, Chini. References Barberis C, Morin D, Durroux T, Mouillac B, Guillon G, Seyer R, Hibert M, and Tribollet E (1999) Molecular pharmacology of AVP and OT receptors and therapeutic potential. Drug News Perspect 12:279–292. Birnbaumer M (2000) Vasopressin receptors. Trends Endocrinol Metab 11:406–410. Busnelli M, Saulière A, Manning M, Bouvier M, Galés C, and
  • 32. Chini B (2012) Func- tional selective oxytocin-derived agonists discriminate between individual G pro- tein family subtypes. J Biol Chem 287:3617–3629. Chini B and Manning M (2007) Agonist selectivity in the oxytocin/vasopressin re- ceptor family: new insights and challenges. Biochem Soc Trans 35:737–741. Chini B, Manning M, and Guillon G (2008) Affinity and efficacy of selective agonists and antagonists for vasopressin and oxytocin receptors: an “easy guide” to receptor pharmacology. Prog Brain Res 170:513–517. Chini B, Mouillac B, Ala Y, Balestre MN, Trumpp-Kallmeyer S, Hoflack J, Elands J, Hibert M, Manning M, and Jard S, et al. (1995) Tyr115 is the key residue for determining agonist selectivity in the V1a vasopressin receptor. EMBO J 14: 2176–2182. Costanzi S (2012) Homology modeling of class A G protein- coupled receptors. Meth- ods Mol Biol 857:259–279. Deupi X and Standfuss J (2011) Structural insights into agonist- induced activation of G-protein-coupled receptors. Curr Opin Struct Biol 21:541–551. Elands J, Barberis C, and Jard S (1988) [3H]-[Thr4,Gly7]OT: a highly selective ligand for central and peripheral OT receptors. Am J Physiol 254:E31– E38.
  • 33. Engelmann M, Wotjak CT, Neumann I, Ludwig M, and Landgraf R (1996) Behavioral consequences of intracerebral vasopressin and oxytocin: focus on learning and memory. Neurosci Biobehav Rev 20:341–358. Ferguson JN, Aldag JM, Insel TR, and Young LJ (2001) Oxytocin in the medial amygdala is essential for social recognition in the mouse. J Neurosci 21:8278–8285. Ferguson JN, Young LJ, Hearn EF, Matzuk MM, Insel TR, and Winslow JT (2000) Social amnesia in mice lacking the oxytocin gene. Nat Genet 25:284–288. Fernando RN, Larm J, Albiston AL, and Chai SY (2005) Distribution and cellular localization of insulin-regulated aminopeptidase in the rat central nervous system. J Comp Neurol 487:372–390. Galés C, Van Durm JJ, Schaak S, Pontier S, Percherancier Y, Audet M, Paris H, and Bouvier M (2006) Probing the activation-promoted structural rearrangements in preassembled receptor-G protein complexes. Nat Struct Mol Biol 13:778–786. Gozzi A, Jain A, Giovannelli A, Bertollini C, Crestan V, Schwarz AJ, Tsetsenis T, Ragozzino D, Gross CT, and Bifone A (2010) A neural switch for active and passive fear. Neuron 67:656–666.
  • 34. Gravati M, Busnelli M, Bulgheroni E, Reversi A, Spaiardi P, Parenti M, Toselli M, and Chini B (2010) Dual modulation of inward rectifier potassium currents in olfactory neuronal cells by promiscuous G protein coupling of the oxytocin receptor. J Neurochem 114:1424–1435. Hanson MA and Stevens RC (2009) Discovery of new GPCR biology: one receptor structure at a time. Structure 17:8–14. Higashida H, Yokoyama S, Munesue T, Kikuchi M, Minabe Y, and Lopatina O (2011) CD38 gene knockout juvenile mice: a model of oxytocin signal defects in autism. Biol Pharm Bull 34:1369–1372. Huber D, Veinante P, and Stoop R (2005) Vasopressin and oxytocin excite distinct neuronal populations in the central amygdala. Science 308:245– 248. Jacobson KA and Costanzi S (2012) New insights for drug design from the X-ray crystallographic structures of G-protein-coupled receptors. Mol Pharmacol 82: 361–371. Jean-Alphonse F, Perkovska S, Frantz MC, Durroux T, Méjean C, Morin D, Loison S, Bonnet D, Hibert M, and Mouillac B, et al. (2009) Biased agonist pharmacocha- perones of the AVP V2 receptor may treat congenital nephrogenic diabetes insip- idus. J Am Soc Nephrol 20:2190–2203.
  • 35. Jin D, Liu HX, Hirai H, Torashima T, Nagai T, Lopatina O, Shnayder NA, Yamada K, Noda M, and Seike T, et al. (2007) CD38 is critical for social behaviour by regu- lating oxytocin secretion. Nature 446:41–45. Kenakin T (2011) Functional selectivity and biased receptor signaling. J Pharmacol Exp Ther 336:296–302. Kobilka B and Schertler GF (2008) New G-protein-coupled receptor crystal struc- tures: insights and limitations. Trends Pharmacol Sci 29:79–83. Kratochwil NA, Gatti-McArthur S, Hoener MC, Lindemann L, Christ AD, Green LG, Guba W, Martin RE, Malherbe P, and Porter RH, et al. (2011) G protein-coupled receptor transmembrane binding pockets and their applications in GPCR research and drug discovery: a survey. Curr Top Med Chem 11:1902– 1924. Lowbridge J, Manning M, Haldar J, and Sawyer WH (1977) Synthesis and some pharmacological properties of [4-threonine, 7-glycine]oxytocin, [1-(L-2-hydroxy-3- mercaptopropanoic acid), 4-threonine, 7-glycine]oxytocin (hydroxyl[Thr4, Gly7] oxytocin), and [7-glycine]oxytocin: peptides with high oxytocic-antidiuretic selec- tivity. J Med Chem 20:120–123. MacKinnon AC, Tufail-Hanif U, Wheatley M, Rossi AG, Haslett C, Seckl M,
  • 36. and Sethi T (2009) Targeting V1A-vasopressin receptors with [Arg6, D-Trp7,9, NmePhe8]-substance P (6-11) identifies a strategy to develop novel anti-cancer therapies. Br J Pharmacol 156:36–47. Manning M, Misicka A, Olma A, Bankowski K, Stoev S, Chini B, Durroux T, Mouillac B, Corbani M, and Guillon G (2012) Oxytocin and vasopressin agonists and antagonists as research tools and potential therapeutics. J Neuroendocrinol 24: 609–628. Manning M, Stoev S, Chini B, Durroux T, Mouillac B, and Guillon G (2008) Peptide and non-peptide agonists and antagonists for the vasopressin and oxytocin V1a, V1b, V2 and OT receptors: research tools and potential therapeutic agents. Prog Brain Res 170:473–512. Michino M, Abola E, Brooks CL, 3rd, Dixon JS, Moult J, and Stevens RC; GPCR Dock 2008 participants (2009) Community-wide assessment of GPCR structure model- ling and ligand docking: GPCR Dock 2008. Nat Rev Drug Discov 8:455–463. Miller G (2013) Neuroscience. The promise and perils of oxytocin. Science 339: 267–269. Modi ME and Young LJ (2012) The oxytocin system in drug discovery for autism: animal models and novel therapeutic strategies. Horm Behav
  • 37. 61:340–350. Oshikawa S, Tanoue A, Koshimizu TA, Kitagawa Y, and Tsujimoto G (2004) Vaso- pressin stimulates insulin release from islet cells through V1b receptors: a com- bined pharmacological/knockout approach. Mol Pharmacol 65:623–629. Pittman QJ and Spencer SJ (2005) Neurohypophysial peptides: gatekeepers in the amygdala. Trends Endocrinol Metab 16:343–344. Rasmussen SG, DeVree BT, Zou Y, Kruse AC, Chung KY, Kobilka TS, Thian FS, Chae PS, Pardon E, and Calinski D, et al. (2011) Crystal structure of the b2 adrenergic receptor-Gs protein complex. Nature 477:549–555. Reversi A, Rimoldi V, Marrocco T, Cassoni P, Bussolati G, Parenti M, and Chini B (2005) The oxytocin receptor antagonist atosiban inhibits cell growth via a “biased agonist” mechanism. J Biol Chem 280:16311–16318. Ring RH, Schechter LE, Leonard SK, Dwyer JM, Platt BJ, Graf R, Grauer S, Puli- cicchio C, Resnick L, and Rahman Z, et al. (2010) Receptor and behavioral phar- macology of WAY-267464, a non-peptide oxytocin receptor agonist. Neuropharmacology 58:69–77. Sala M, Braida D, Donzelli A, Martucci R, Busnelli M, Bulgheroni E, Rubino T, Parolaro D, Nishimori K, and Chini B (2013) Mice
  • 38. heterozygous for the oxytocin receptor gene (Oxtr(1/-)) show impaired social behaviour but not increased ag- gression or cognitive inflexibility: evidence of a selective haploinsufficiency gene effect. J Neuroendocrinol 25:107–118. Sala M, Braida D, Lentini D, Busnelli M, Bulgheroni E, Capurro V, Finardi A, Donzelli A, Pattini L, and Rubino T, et al. (2011) Pharmacologic rescue of impaired cognitive flexibility, social deficits, increased aggression, and seizure susceptibility in oxytocin receptor null mice: a neurobehavioral model of autism. Biol Psychiatry 69:875–882. Serradeil-Le Gal C, Raufaste D, Derick S, Blankenstein J, Allen J, Pouzet B, Pascal M, Wagnon J, and Ventura MA (2007) Biological characterization of rodent and human vasopressin V1b receptors using SSR-149415, a nonpeptide V1b receptor ligand. Am J Physiol Regul Integr Comp Physiol 293:R938– R949. Takayanagi Y, Yoshida M, Bielsky IF, Ross HE, Kawamata M, Onaka T, Yanagisawa T, Kimura T, Matzuk MM, and Young LJ, et al. (2005) Pervasive social deficits, but 326 Busnelli et al. normal parturition, in oxytocin receptor-deficient mice. Proc
  • 39. Natl Acad Sci USA 102:16096–16101. Urban JD, Clarke WP, von Zastrow M, Nichols DE, Kobilka B, Weinstein H, Javitch JA, Roth BL, Christopoulos A, and Sexton PM, et al. (2007) Functional selectivity and classical concepts of quantitative pharmacology. J Pharmacol Exp Ther 320: 1–13. Wallis M (2012) Molecular evolution of the neurohypophysial hormone precursors in mammals: comparative genomics reveals novel mammalian oxytocin and vaso- pressin analogues. Gen Comp Endocrinol 179:313–318. Wesley VJ, Hawtin SR, Howard HC, and Wheatley M (2002) Agonist-specific, high- affinity binding epitopes are contributed by an arginine in the N-terminus of the human oxytocin receptor. Biochemistr y 41:5086–5092. Wichard JD, Ter Laak A, Krause G, Heinrich N, Kühne R, and Kleinau G (2011) Chemogenomic analysis of G-protein coupled receptors and their ligands deciphers locks and keys governing diverse aspects of signalling. PLoS ONE 6:e16811. Zhao Q and Wu BL (2012) Ice breaking in GPCR structural biology. Acta Pharmacol Sin 33:324–334. Zingg HH and Laporte SA (2003) The oxytocin receptor. Trends Endocrinol Metab 14:
  • 40. 222–227. Address correspondence to: Dr. Bice Chini, CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milano, Italy. E-mail: [email protected] Identification of Selective and Potent Analogs of Mouse OTR 327 mailto:[email protected] PK-PD Primer Homework Max 4pages only answers PK-Pharmacology Primer Instructions for the PK problems: Please answer the questions below as best as you can by examining the outcomes of the pharmacokinetic models provided in the Excel file “CLRE-238 SP21 PK-PD Homework.xlxs.” In many cases, you will need to find the requested value of parameters by trial and error, changing some of the values in bold red in the worksheets and looking at the resulting plots. You do not need to perform any calculations. All the answers are very short. Do not be intimidated by the Excel worksheets; this is a lot easier than it appears at first glance. General comment: Because you are expected to obtain your answers by visually examining the pharmacokinetic plots in the associated Excel file, your quantitative answers will be graded as correct if they were reasonably close to the exact answer. There is no requirement to be precise. Problem A: The antibiotic genericomycin (not a real one) is administered by iv bolus to hospitalized patients. The antibiotic has an elimination half-life of 4.0 h in people. Previous experience has
  • 41. shown that to be effective at reducing the bacterial burden its plasma concentration must equal or higher than 5 µg/ml (Ceff). Question 1: Using the “Intravenous” tab of the Excel file, determine the duration of action of the following iv bolus doses and enter the values in the table below. Hint: You can eyeball the time values from the chart or read them from the list of Cp vs time for the Plot (blue numbers) Answer 1: Dose (mg) Duration of Action (h) 140 4 280 8 560 12 1120 16 Question 2: What is the effect of doubling the dose on the duration of action? Answer 2: it adds the duration of action by 4 hours Question 3: What would be the necessary iv bolus dose to establish plasma concentration 5 µg/ml at 24 h? Is this dose realistic? Assume genericomycin costs $10/mg. Answer 3: Problem B: You have created a new company, NovoAntibiot, Inc. with the intention of creating a new antibiotic with much better properties than genericomycin. Your clinical advisors suggest
  • 42. that the new antibiotic should be administered by iv bolus once a day with a dose of at most 560 mg per person (your new therapeutic product profile). Question 4: What elimination half-life should you design in your new antibiotic to achieve a plasma concentration of at least 5 µg/ml 24 h after a 560 mg iv bolus dose? Answer 4: 40 Welcome to drug discovery! After working for 2 years and spending $15 M from your Angel and series A investors you were unable to increase the half-life of your new antibiotics. All of them still have halflives around 4 h. But you got lucky, some of your new compounds have higher potency. Drug candidate NABT 813, is much more potent than genericomycin. In vitro experiments and animal models suggest that the minimum effective concentration (Ceff) in humans will be 1 µg/ml. Question 5: What iv bolus dose of NABT 813 will treat the patients for 24h if its elimination half-life is 4.0 h and its Ceff is 1 µg/ml? Is it realistic? (compare with your Answer 3 above).) Answer 5: 140mg Problem C Unfortunately, your new antibiotic NABT 813 decreased the number and body weight of rat pups in formal GLP repro-tox studies in rats. The compound cannot be used in women who might not know whether they are pregnant at the time they are admitted to the hospital with a systemic infection. This is a crushing blow and your investors abandon you. The new drug discovery program is terminated.
  • 43. However, your PK consultant suggests that you might be able to formulate genericomycin for subcutaneous administration by creating an insoluble salt and adding some excipients that will retard its absorption, allowing (perhaps) once a day dosing. Your intellectual property attorney states that you might be able to obtain patent protection for the new formulation if you demonstrate substantial advantages over the generic iv formulation. You apply for and get an SBIR grant to demonstrate proof of (PK) concept. Question 6: In the “Subcutaneous” tab of the Excel file, set the elimination half-life of genericomycin to 4.0 h, its Ceff at 5 µg/ml, and its dose to 280 mg. Examine the effect of changing the absorption half-life of genericomycin from your new formulations by replacing the value of t1/2 (abs) and complete the table below. Answer 6: t1/2 abs (h) Tmax (h) Cmax (µg/ml) 0.05 0.25 18 1 2.5 13 3 5 8.5 6 7 5.9
  • 44. 10 9 4.2 Hint: You can eyeball the above values from the chart or read them from the list of Cp vs time for the Plots (blue numbers). No need to be very precise. Question 7: Why does the PK profile for absorption with a very short half-life (0.05 h) look like the ivbolus profile? (This never happens in real life….) Answer 7: Clearance is more rapid and half-lives are shorter Question 8: What is the effect of increasing the absorption half- life on Tmax and Cmax? Answer 8: increasing tmax increases absorption while increasing cmax decreases absorption Question 9: Is there any value of absorption half-life that will allow you to have plasma concentration higher or equal to Ceff at 24 h when dosing sc 280 mg? Answer 9: yes Question 10: What is the duration of action of genericomycin at a dose of 280 mg if it is administered in the formulation with absorption half-life of 10 hours? Answer 10: 4.3 Problem X (bonus): Your microbiology consultant tells you that an analogue of genericomycin with longer eliminationhalflife was developed in the Soviet Union in the 1960’s, but it never reached the West and was forgotten. Its name was bolshoimitsina and had an elimination half-life in humans of 7.8 h, and Ceff also 5 µg/ml.
  • 45. You obtain a sample of bolshoimitsina via a website vendor in Bulgaria and find that it also has a sc absorption half-life of 10.0 h in your preferred formulation. Question 11: What sc dose of formulated bolshoimitsina will provide a plasma concentration equal to Ceff 5 µg/ml at 24 h? Answer 11: 9.43mg Question 12: At the dose of bolshoimitsina you found in Answer 11, when does the drug start having its effect in the patient? Answer 12: 0.5hrs PK-Pharmacology Primer Homework with Answers Student Name: Instructions for the Pharmacology problems: Please extract the requested information from the papers provided. You do not need to read the papers in detail. This is an exercise in identifying the required information quickly while checking its validity. Your answers will be very short, a value (with units!) or a few words. Problem P: Look at the provided paper by Busnelli et al., “Selective and Potent Agonists and Antagonists for Investigating the Role of Mouse Oxytocin Receptors”, and answer the following questions: Question 31: What was the Ki value for compound “LVA” at the mOTR (mouse oxytocin receptor)? Answer 31: Question 32: Does LVA have higher or lower affinity than “OT” (oxytocin) for the mOTR? Answer 32:
  • 46. Look in Materials and Methods, section on Ligand Binding Assays. Question 33: How were the Ki values obtained: Schild analysis or from IC50 values? Answer 33: Question 34: What compound was the radioligand for measuring binding to the mOTR, and at what concentration was used? Answer 34: Question 35: What was the Kd value for the radioligand at the mOTR? Answer 35: Look in the main body of the paper. Question 36: Which figure shows the competition binding experiments for compound “OTA1”? Answer 36: Question 37: Looking at said figure, would you say that OTA1 has higher affinity for the mV1aR (mouse vasopressin type 1a receptor) than for the mOTR? Answer 37: Problem Q: Look at the provided paper by Ichinose et al., “Development of a Highly Potent Analogue and a Long-Acting Analogue of Oxytocin for the Treatment of Social Impairment-Like Behaviors”, and answer the following questions: Question 38: What was the EC50 at the hOTR (human oxytocin receptor) of compound 4? Answer 38: Question 39: What was the efficacy of compound 4?
  • 47. Answer 39: Question 40: Of the new compounds prepared in this paper (Compounds 1 to 6), which one was more potent at the hOTR? Which one was the most potent at the hV1aR? Answer 40: Look in Results and Discussion, section on Agonist-Induced Increase in Intracellular Ca2+ Concentrations. Question 41: Which second messenger was measured to determine receptor activation? Answer 41: Look at Table 2, data on Ki at the hOTR and EC50 at the hOTR Question 42: Would you suspect that there might be considerable “receptor reserve” for activation of the hOTR by OT? What about carbetocin? Answer 42: Page 1 of 5 PK-PD Primer Homework Max 4pages only answers PK-Pharmacology Primer Instructions for the PK problems: Please answer the questions below as best as you can by examining the outcomes of the pharmacokinetic models provided in the Excel file “CLRE-238 SP21 PK-PD Homework.xlxs.” In many cases, you will need to find the requested value of parameters by trial and error, changing some of the values in bold red in the worksheets and looking at the
  • 48. resulting plots. You do not need to perform any calculations. All the answers are very short. Do not be intimidated by the Excel worksheets; this is a lot easier than it appears at first glance. General comment: Because you are expected to obtain your answers by visually examining the pharmacokinetic plots in the associated Excel file, your quantitative answers will be graded as correct if they were reasonably close to the exact answer. There is no requirement to be precise. Problem A: The antibiotic genericomycin (not a real one) is administered by iv bolus to hospitalized patients. The antibiotic has an elimination half-life of 4.0 h in people. Previous experience has shown that to be effective at reducing the bacterial burden its plasma concentration must equal or higher than 5 µg/ml (Ceff). Question 1: Using the “Intravenous” tab of the Excel file, determine the duration of action of the following iv bolus doses and enter the values in the table below. Hint: You can eyeball the time values from the chart or read them from the list of Cp vs time for the Plot (blue numbers) Answer 1: Dose (mg) Duration of Action (h) 140 4 280 8 560 12 1120 16
  • 49. Question 2: What is the effect of doubling the dose on the duration of action? Answer 2: it adds the duration of action by 4 hours Question 3: What would be the necessary iv bolus dose to establish plasma concentration 5 µg/ml at 24 h? Is this dose realistic? Assume genericomycin costs $10/mg. Answer 3: Problem B: You have created a new company, NovoAntibiot, Inc. with the intention of creating a new antibiotic with much better properties than genericomycin. Your clinical advisors suggest that the new antibiotic should be administered by iv bolus once a day with a dose of at most 560 mg per person (your new therapeutic product profile). Question 4: What elimination half-life should you design in your new antibiotic to achieve a plasma concentration of at least 5 µg/ml 24 h after a 560 mg iv bolus dose? Answer 4: 40 Welcome to drug discovery! After working for 2 years and spending $15 M from your Angel and series A investors you were unable to increase the half-life of your new antibiotics. All of them still have halflives around 4 h. But you got lucky, some of your new compounds have higher potency. Drug candidate NABT 813, is much more potent than genericomycin. In vitro experiments and animal models suggest that the minimum effective concentration (Ceff) in humans will be 1 µg/ml. Question 5: What iv bolus dose of NABT 813 will treat the patients for 24h if its elimination half-life is 4.0 h and its Ceff is 1 µg/ml? Is it realistic? (compare with your Answer 3 above).)
  • 50. Answer 5: 140mg Problem C Unfortunately, your new antibiotic NABT 813 decreased the number and body weight of rat pups in formal GLP repro-tox studies in rats. The compound cannot be used in women who might not know whether they are pregnant at the time they are admitted to the hospital with a systemic infection. This is a crushing blow and your investors abandon you. The new drug discovery program is terminated. However, your PK consultant suggests that you might be able to formulate genericomycin for subcutaneous administration by creating an insoluble salt and adding some excipients that will retard its absorption, allowing (perhaps) once a day dosing. Your intellectual property attorney states that you might be able to obtain patent protection for the new formulation if you demonstrate substantial advantages over the generic iv formulation. You apply for and get an SBIR grant to demonstrate proof of (PK) concept. Question 6: In the “Subcutaneous” tab of the Excel file, set the elimination half-life of genericomycin to 4.0 h, its Ceff at 5 µg/ml, and its dose to 280 mg. Examine the effect of changing the absorption half-life of genericomycin from your new formulations by replacing the value of t1/2 (abs) and complete the table below. Answer 6: t1/2 abs (h)
  • 51. Tmax (h) Cmax (µg/ml) 0.05 0.25 18 1 2.5 13 3 5 8.5 6 7 5.9 10 9 4.2 Hint: You can eyeball the above values from the chart or read them from the list of Cp vs time for the Plots (blue numbers). No need to be very precise. Question 7: Why does the PK profile for absorption with a very short half-life (0.05 h) look like the ivbolus profile? (This never happens in real life….) Answer 7: Clearance is more rapid and half-lives are shorter Question 8: What is the effect of increasing the absorption half- life on Tmax and Cmax? Answer 8: increasing tmax increases absorption while increasing cmax decreases absorption Question 9: Is there any value of absorption half-life that will allow you to have plasma concentration higher or equal to Ceff
  • 52. at 24 h when dosing sc 280 mg? Answer 9: yes Question 10: What is the duration of action of genericomycin at a dose of 280 mg if it is administered in the formulation with absorption half-life of 10 hours? Answer 10: 4.3 Problem X (bonus): Your microbiology consultant tells you that an analogue of genericomycin with longer eliminationhalflife was developed in the Soviet Union in the 1960’s, but it never reached the West and was forgotten. Its name was bolshoimitsina and had an elimination half-life in humans of 7.8 h, and Ceff also 5 µg/ml. You obtain a sample of bolshoimitsina via a website vendor in Bulgaria and find that it also has a sc absorption half-life of 10.0 h in your preferred formulation. Question 11: What sc dose of formulated bolshoimitsina will provide a plasma concentration equal to Ceff 5 µg/ml at 24 h? Answer 11: 9.43mg Question 12: At the dose of bolshoimitsina you found in Answer 11, when does the drug start having its effect in the patient? Answer 12: 0.5hrs PK-Pharmacology Primer Homework with Answers Student Name: Instructions for the Pharmacology problems: Please extract the requested information from the papers provided. You do not need to read the papers in detail. This is an exercise in identifying the required information quickly while checking its validity. Your answers will be very short, a value (with units!) or a few words.
  • 53. Problem P: Look at the provided paper by Busnelli et al., “Selective and Potent Agonists and Antagonists for Investigating the Role of Mouse Oxytocin Receptors”, and answer the following questions: Question 31: What was the Ki value for compound “LVA” at the mOTR (mouse oxytocin receptor)? Answer 31: Question 32: Does LVA have higher or lower affinity than “OT” (oxytocin) for the mOTR? Answer 32: Look in Materials and Methods, section on Ligand Binding Assays. Question 33: How were the Ki values obtained: Schild analysis or from IC50 values? Answer 33: Question 34: What compound was the radioligand for measuring binding to the mOTR, and at what concentration was used? Answer 34: Question 35: What was the Kd value for the radioligand at the mOTR? Answer 35: Look in the main body of the paper. Question 36: Which figure shows the competition binding experiments for compound “OTA1”? Answer 36: Question 37: Looking at said figure, would you say that OTA1 has higher affinity for the mV1aR (mouse vasopressin type 1a
  • 54. receptor) than for the mOTR? Answer 37: Problem Q: Look at the provided paper by Ichinose et al., “Development of a Highly Potent Analogue and a Long-Acting Analogue of Oxytocin for the Treatment of Social Impairment-Like Behaviors”, and answer the following questions: Question 38: What was the EC50 at the hOTR (human oxytocin receptor) of compound 4? Answer 38: Question 39: What was the efficacy of compound 4? Answer 39: Question 40: Of the new compounds prepared in this paper (Compounds 1 to 6), which one was more potent at the hOTR? Which one was the most potent at the hV1aR? Answer 40: Look in Results and Discussion, section on Agonist-Induced Increase in Intracellular Ca2+ Concentrations. Question 41: Which second messenger was measured to determine receptor activation? Answer 41: Look at Table 2, data on Ki at the hOTR and EC50 at the hOTR Question 42: Would you suspect that there might be considerable “receptor reserve” for activation of the hOTR by OT? What about carbetocin? Answer 42:
  • 55. Page 1 of 5 IntravenousThese equations are for reference only. You do not have to use them to complete your homework. But they help to understand the results!Plasma concentration (Cp) for one compartment model with iv bolus dosing and first order eliminationCp= (D/Vd)*exp(-ke*t)Equation for extension of duration of actionUsing the model in this worksheet, please answer the questions in the handout Ci is the plasma concentration at a given time, assumed to be Ci>CeffPlease do not change any of the calculationsDelta teff is for how much longer the drug will be active (above Ceff) for a given half- lifeYou only have to change some of the values of some of the parameters in bold redThis equation can be derived from the Cp(t) equation aboveThis plot describes the plasma concentration (Cp) of an ideal drug after administration of an intravenous bolus(There are no drugs with this simple pharmacokinetic behavior, but it is a simple illustrative example)The drug is considered active in the patient if its plasma concentration is superior to a threshold value "Ceff" determined from previous clinical experienceParameterAbbreviationUnitsYour ValuesElimination half-lifet1/2 (elim)h4.0Same plot, with Y axis set to maximum Cp 10 ug/mlDoseDmg/personActive plasma concentration thresholdCeffug/ml1.0Don't touch these cellsElimination rateke1/h0.173Apparent volume of distributionVdL/person14Threshold plotTime (h)Ceff (ug/ml)01.0251.0Don't touch these cellsPlot Time (h)Cp (ug/ml)00.00.10.00.250.00.50.010.01.50.020.030.040.050.060.0 80.0100.0120.0140.0160.0180.0200.0220.0240.000260.0280.03 00.0 PK Intravenous Administration Cp (ug/ml) 0 0.1 0.25 0.5 1 1.5 2 3 4 5 6 8 10 12 14 16 18 20 22 24 26 28
  • 56. 30 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 Ceff (ug/ml) 0 25 1 1 Time (h) Cp (ug/ml) PK Intravenous Administration Cp (ug/ml) 0 0.1 0.25 0.5 1 1.5 2 3 4 5 6 8 10 12 14 16 18 20 22 24 26 28 30 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 Ceff (ug/ml) 0 25 1 1 Time (h) Cp (ug/ml) SubcutaneousCLRE-238Applied Translational ResearchThese equations are for reference only. You do not have to use them to complete your homework. But they help to understand the results!Spring 2021PK-PD Primer HomeworkPlasma concentration (Cp) for one compartment model with sc bolus dosing and first order eliminationDate3/30/21Claudio D. Schteingart Ph.D.C(t) is Cp the plasma concentration as a function of timef is the bioavailability (scale 0 to 1) of the drug in the sc spaceUsing the model in this worksheet, please answer the questions in the handout ka is the absorption rate, ke is the
  • 57. elimination ratePlease do not change any of the calculationsD is the dose, Vd is the apparent volume of distributionYou only have to change some of the values of some of the parameters in bold redEquation for extension of duration of actionCi is the plasma concentration at a given time, assumed to be Ci>CeffDelta teff is for how much longer the drug will be active (above Ceff) for a given half-lifeThis equation can be used during the elimination phase of the above curveThis plot describes the plasma concentration (Cp) of an ideal drug after administration of a subcutaneous bolusThe drug is considered active in the patient if its plasma concentration is superior to a threshold value "Ceff" determined from previous clini cal experienceIf the absorption half-life equals the elimination half- life the math fails (division by zero). If needed, enter a slightly different value, e.g. 3.99 or 4.01 instead of 4.00Same plot, with Y axis set to maximum Cp 10 ug/mlParameterAbbreviati onUnitsYour ValuesAbsorption half- lifet1/2 (abs)h24.00Elimination half-lifet1/2 (elim)h5.0DoseDmg/person9.43Active plasma concentration thresholdCeffug/ml5.0Don't touch these cellsSubcutaneous bioavailability 0 to 1f1Apparent volume of distributionVdL/person14Absorption rateka1/h0.029Elimination rateke1/h0.139Multiplication factorfDka/(Vd(Ka-ke))ug/ml- 0Threshold plotTime (h)Ceff (ug/ml)05.0265.0Don't touch these cellsPlotTime (h)Cp (ug/ml)00.000.10.000.250.000.50.0110.021.50.0320.032.50.043 0.053.50.0540.0650.0660.0770.0890.09110.09130.09150.09170. 09190.09210.09230.08240.08250.08260.08270.08 PK Subcutaneous Administration Cp (ug/ml) 0 0.1 0.25 0.5 1 1.5 2 2.5 3 3.5 4 5 6 7 9 11 13 15 17 19 21 23 24 25 26 27 0 1.9291349543759174E-3 4.7627511742739078E-3 9.3290009959562988E-3 1.7899518669098738E-2 2.5764263897523468E-2 3.297239017841809E-2 3.9569732249353361E-2
  • 58. 4.5599028724170154E-2 5.1100129814037637E-2 5.6110191132563512E-2 6.4793416734735515E-2 7.1898633268821469E-2 7.764303677600605E-2 8.5779467479190435E-2 9.0433868498689868E-2 9.2534238683107894E-2 9.2779107146231402E-2 9.1693364761172624E-2 8.967056151835319E-2 8.7004947768308957E-2 8.3915743577409357E-2 8.2264905141199454E-2 8.056551884472278E-2 7.8831334871670267E-2 7.7074110918482291E-2 Ceff (ug/ml) 0 26 5 5 Time (h) Cp (ug/ml) PK Subcutaneous Administration Cp (ug/ml) 0 0.1 0.25 0.5 1 1.5 2 2.5 3 3.5 4 5 6 7 9 11 13 15 17 19 21 23 24 25 26 27 0 1.9291349543759174E-3 4.7627511742739078E-3 9.3290009959562988E-3 1.7899518669098738E-2 2.5764263897523468E-2 3.297239017841809E-2 3.9569732249353361E-2 4.5599028724170154E-2 5.1100129814037637E-2 5.6110191132563512E-2 6.4793416734735515E-2 7.1898633268821469E-2 7.764303677600605E-2 8.5779467479190435E-2 9.0433868498689868E-2 9.2534238683107894E-2 9.2779107146231402E-2 9.1693364761172624E-2 8.967056151835319E-2 8.7004947768308957E-2 8.3915743577409357E-2 8.2264905141199454E-2 8.056551884472278E-2 7.8831334871670267E-2 7.7074110918482291E-2 Ceff (ug/ml) 0 26 5 5