ArticlePDF Available

Taar1-Mediated Modulation Of Presynaptic Dopaminergic Neurotransmission: Role Of D2 Dopamine Autoreceptors.

Authors:

Figures

Content may be subject to copyright.
Taar1-mediated modulation of presynaptic dopaminergic
neurotransmission: Role of D2 dopamine autoreceptors
D. Leo
a
, L. Mus
a
, S. Espinoza
a
, M.C. Hoener
b
, T.D. Sotnikova
a
, R.R. Gainetdinov
a
,
c
,
d
,
*
a
Department of Neuroscience and Brain Technology, Istituto Italiano di Tecnologia, 16163, Genova, Italy
b
Neuroscience Research, Pharmaceuticals Division, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
c
Skolkovo Institute of Science and Technology, Skolkovo, 143025, Moscow Region, Russia
d
Faculty of Biology and Soil Science, St. Petersburg State University, 199034, St. Petersburg, Russia
article info
Article history:
Received 7 January 2014
Received in revised form
12 February 2014
Accepted 12 February 2014
Keywords:
Trace amine-associate receptor 1 (TAAR1)
Dopamine (DA)
Fast scan cyclic voltammetry (FSCV)
Dopamine receptor 2 (D2R)
Schizophrenia
Neuropsychiatric disorders
abstract
Trace Amine-Associated Receptor 1 (TAAR1) is a G protein-coupled receptor (GPCR) expressed in several
mammalian brain areas and activated by trace amines(TAs). TAs role is unknown; however, discovery
of their receptors provided an opportunity to investigate their functions. In vivo evidence has indicated
an inhibitory inuence of TAAR1 on dopamine (DA) neurotransmission, presumably via modulation of
dopamine transporter (DAT) or interaction with the D2 DA receptor and/or activation of inwardly
rectifying K
þ
channels. To elucidate the mechanisms of TAAR1-dependent modulation, we used TAAR1
knockout mice (TAAR1-KO), a TAAR1 agonist (RO5166017) and a TAAR1 antagonist (EPPTB) in a set of
neurochemical experiments. Analysis of the tissue content of TAAR1-KO revealed increased level of the
DA metabolite homovanillic acid (HVA), and in vivo microdialysis showed increased extracellular DA in
the nucleus accumbens (NAcc) of TAAR1-KO. In fast scan cyclic voltammetry (FSCV) experiments, the
evoked DA release was higher in the TAAR1-KO NAcc. Furthermore, the agonist RO5166017 induced a
decrease in the DA release in wild-type that could be prevented by the application of the TAAR1
antagonist EPPTB. No alterations in DA clearance, which are mediated by the DAT, were observed. To
evaluate the interaction between TAAR1 and D2 autoreceptors, we tested the autoreceptor-mediated
dynamics. Only in wild type mice, the TAAR1 agonist was able to potentiate quinpirole-induced inhib-
itory effect on DA release. Furthermore, the short-term plasticity of DA release following paired pulses
was decreased in TAAR1-KO, indicating less autoinhibition of D2 autoreceptors. These observations
suggest a close interaction between TAAR1 and the D2 autoreceptor regulation.
Ó2014 Elsevier Ltd. All rights reserved.
1. Introduction
Octopamine (OCT), tyramine (TYR) and
b
-phenethylamine (
b
-
PEA), as well as several other non-catechol amines, are metabolites
of aromatic amino acids and are known as trace amines (TAs). TAs
are a family of endogenous compounds with strong structural
similarity to the classical monoamine neurotransmitters and are
present in mammalian tissues at low (nanomolar) concentrations
(Berry, 2004; Grandy, 2007; Lindemann and Hoener, 2005). The
endogenous levels of these compounds are at least two orders of
magnitude below those of classical monoamine neurotransmitters
such as dopamine (DA), noradrenaline (NE) and serotonin (5HT).
TAs are found in many species; in invertebrates, tyramine and
octopamine are well-characterized neurotransmitters that modu-
late movement, feeding, metabolism, muscular tone and other
functions (Axelrod and Saavedra, 1977; Cooper and Venton, 2009).
Trace amines are also produced in bacteria, fungi, and plant cells
and can be found in some food products, most notably in chocolate,
cheese and red wine (Branchek and Blackburn, 2003). Despite be-
ing known for more than a century, the role played by TAs in
mammalian, and particularly human, physiology is still enigmatic.
However, it has been noted that levels of TAs are altered in a variety
of human disorders ranging from schizophrenia, Parkinsons dis-
ease, attention decit hyperactivity disorder (ADHD), and
Abbreviations: TAAR1, Trace Amine-Associated Receptor 1; DA, dopamine; DAT,
dopamine transporter; EPPTB, (N-(3-ethoxy-phenyl)-4-pyrrolidin-1-yl-3-
triuoromethyl-benzamide; HVA, homovanillic acid; FSCV, fast scan cyclic vol-
tammetry; NAcc, nucleus accumbens; DStr, dorsal striatum; D2, dopamine receptor
2.
*Corresponding author. Department of Neuroscience and Brain Technology,
Istituto Italiano di Tecnologia, Genova 16163, Italy. Tel.: þ39 010 71781516.
E-mail addresses: damiana.leo@iit.it (D. Leo), liudmila.mus@iit.it (L. Mus),
stefano.espinoza@iit.it (S. Espinoza), marius.hoener@roche.com (M.C. Hoener),
tatiana.sotnikova@iit.it (T.D. Sotnikova), raul.gainetdinov@iit.it (R.R. Gainetdinov).
Contents lists available at ScienceDirect
Neuropharmacology
journal homepage: www.elsevier.com/locate/neuropharm
http://dx.doi.org/10.1016/j.neuropharm.2014.02.007
0028-3908/Ó2014 Elsevier Ltd. All rights reserved.
Neuropharmacology 81 (2014) 283e291
Tourettes syndrome to migraines and drug addiction (Boulton,
1980; Sandler et al., 1980). For decades, TAs were considered to
be false neurotransmittersthat were able to modulate the
signaling of monoamines by displacing them from storage vesicles
and/or by acting on the plasma membrane transporters in an
amphetamine-like manner (Berry, 2004; Parker and Cubeddu,
1988). Interestingly, the rate of synthesis of TAs was found to be
comparable with that of classic monoamines, suggesting that the
low levels of TAs in brain tissue are most likely determined by the
extremely fast rate of metabolism and/or the inability of TAs to be
stored in vesicles as classical neurotransmitters (Grandy, 2007;
Sotnikova et al., 2009).
However, in 2001, a family of novel mammalian G protein-
coupled receptors (GPCRs) were characterized with some mem-
bers of this family showing a high afnity for TAs (Borowsky et al.,
2001). This family of newly discovered receptors was later re-
named the Trace Amine-Associated Receptors (TAARs) family
(Maguire et al., 2009; Lindemann et al., 2005). The TAAR family
includes 6 functional members in humans (TAAR1-9 including 3
members encoded by pseudo-genes) and even more receptors are
found in rodents (Borowsky et al., 2001; Bunzow et al., 2001;
Lindemann and Hoener, 2005). Interestingly, in the TAAR family
only TAAR1 and TAAR4 possess any demonstrable TA responsive-
ness. TA binding to TAAR1 engages G
a
s
-type G proteins that activate
adenylyl cyclases (Berry, 2004). TAAR1 is the best characterized
TAAR member and is found in some areas of the central nervous
system and in certain peripheral tissues (Revel et al., 2013). This
distribution, which includes components of the limbic system, such
as the amygdala, and areas rich in monoaminergic cell bodies, such
as the dorsal raphe nucleus and the ventral tegmental area (VTA)
(Lindemann et al., 2008), makes TAAR1 a promising target for
pharmaceutical treatment of monoamine-related disorders (Revel
et al., 2012a, 2013). Because TAs affect multiple targets including
TAAR1, TAAR4, and DA transporter (DAT), adrenergic and serotonin
receptors, their use in the identication of specic functions of
TAAR1 are limited. Only the generation of TAAR1-decent and
-overexpressing mice (TAAR1-KO and TAAR1-OE mice) (Lindemann
et al., 2008; Wolinsky et al., 2007; Revel et al., 2012b), the devel-
opment of selective TAAR1 agonists, such as RO5166017 (Revel
et al., 2011), and antagonists, such as ((N-(3-Ethoxy-phenyl)-4-
pyrrolidin-1-yl-3-triuoromethyl-benzamide EPPTB) (Bradaia
et al., 2009), provided an opportunity to evaluate the specic
roles and mechanisms mediated by TAAR1. TAAR1-KO mice appear
to be similar to control animals at basal state, but they show
enhanced hyperlocomotion and exaggerated striatal release of DA,
NE, and 5-HT when challenged with
D
-amphetamine. Recent
studies on TAAR1-KO mice have demonstrated that TAAR1 is able to
negatively modulate monoaminergic neurotransmission
(Lindemann et al., 2008; Wolinsky et al., 2007). For example, the
genetic ablation of TAAR1 induces an increase in the spontaneous
ring rate of DA neurons (Lindemann et al., 2008), and similar ef-
fects are mediated via application of the selective TAAR1 antagonist
EPPTB in control animals (Bradaia et al., 2009), corroborating the
idea that TAAR1 normally exerts an inhibitory effect on DA neurons.
Although the underlying TAAR1 signaling mechanism remained
unclear, Bradaia et al. clearly showed that TAAR1 activates inwardly
rectifying K
þ
channels (Bradaia et al., 2009). They also demon-
strated that both the acute application of EPPTB and the constitu-
tive genetic lack of TAAR1 increase the potency of DA at D2
receptors in DA neurons (Bradaia et al., 2009). Studies in vitro and
in vivo offered further indication of a physical and functional
interaction between TAAR1 and D2 receptors (Espinoza et al., 2011),
whereas others have suggested that TAAR1 may directly alter DAT
function (Miller et al., 2005). To complement the information about
TAAR1 function, a line of transgenic mice that overexpresses TAAR1
in the brain has been recently generated (Revel et al., 2012b). This
model is hyposensitive to amphetamine and it shows constitutive
hyperactivity of monoaminergic nuclei (Revel et al., 2012b). Overall,
the growing body of evidence suggests a modulatory role of TAAR1
on monoaminergic activity (Reese et al., 2014; Cichero et al., 2013),
particularly on presynaptic function. To determine whether dele-
tion of the Taar1 gene or application of TAAR1 ligands perturbs the
functional presynaptic activity of DA neurons at the level of axon
terminals, we investigated extracellular DA dynamics using fast
scan cyclic voltammetry (FSCV) and in vivo microdialysis tech-
niques in the dorsal striatum (DStr) and nucleus accumbens (NAcc)
of wild type (WT) and TAAR1-KO mice. Furthermore, we applied
FSCV to evaluate the evoked DA release and clearance rates in these
brain regions of WT and TAAR1-KO mice in the presence of the
selective TAAR1 agonist (RO5166017) and a TAAR1 antagonist ((N-
(3-Ethoxy-phenyl)-4-pyrrolidin-1-yl-3-triuoromethyl-benza-
mide EPPTB).
We found that DA release evoked by a single stimulus was
higher in NAcc, and the basal extracellular level of DA was signi-
cantly higher in this brain region in TAAR1-KO mice. The TAAR1
agonist RO5166017 decreases DA release in WT mice but not in
TAAR1-KO animals, and application of the TAAR1 antagonist EPPTB
prevented the reduction in the evoked DA release induced by the
TAAR1 agonist in WT animals. We further gained functional evi-
dence suggesting that these presynaptic effects could be mediated
by an interaction between TAAR1 and D2 DA autoreceptors.
2. Materials and methods
2.1. Animals
All experiments were conducted in compliance with the Italian Ministry of
Health (DL 116/92; DL 111/94-B) and European Community (86/609/EEC) directives
regulating animal research. All efforts were made to minimize animal suffering, to
reduce the number of animals used, and to utilize alternatives to in vivo techniques,
if available. Animals were housed under a 12 h light/12 h dark cycle with ad libitum
access to food and water.TAAR1-KO mice of mixed backgrounds (C57BL/6J 129Sv/J
backgrounds) were generated as described previously (Lindemann et al., 2008;
Wolinsky et al., 2007; Espinoza et al., 2011). TAAR1-KO and WT littermates were
obtained from heterozygous matings. Genotyping was performed by PCR. Mice of
both sexes of at least 2.5 months of age were used in FSCV experiments. Three-
month-old males were used for microdialysis studies.
2.2. HPLC measurements of the tissue content of monoamines and their metabolites
Striatal tissue (including both DStr and NAcc) was dissected from WT and
TAAR1-KO mice and homogenized in 40 volumes of 0.1 M HClO
4
. Following
centrifugation and ltration, the samples were analyzed by HPLC as described
below. The protocol for sample preparation for HPLC determination of tissue
monoamines and their metabolites was performed as previously described (Jones
et al., 1998; Gainetdinov et al., 20 03).
2.3. In vivo microdialysis
2.3.1. Surgery
In vivo brain microdialysis was performed in the right dorsal striatum or in the
right NAcc of freely moving mice (Budygin et al., 2004; Jones et al., 1998; Carboni
et al., 2001) using 2 mm (for striatum) or 1 mm (for NAcc) concentric micro-
dialysis probes (membrane length cut off 6000 Da; CMA-11, CMA/Microdialysis,
Solna, Sweden). Stereotaxic coordinates for the position of the probes were chosen
according to the atlas of Franklin and Paxinos (1997) and are relative to the bregma:
AP 0.0; L 2.5; DV 4.4 for the striatum and AP þ1.3; L 0.9; DV 5 for the NAcc. Prior
to xation in the stereotaxic apparatus, the animals were anesthetized with an
oxygen/isourane mixture. The probes were implanted in the brain vertically
through a small drilled aperture in the scull and xed with dental cement. During
implantation into the brain and for 1 h afterward, the dialysis probes were perfused
with articial cerebrospinal uid (aCSF) (NaCl 147 mM, KCl 2.7 mM, CaCl
2
1.2 m M,
MgCl
2
0.85 mM; CMA Microdialysis). 1 h after the operation, the animals were
returned to their home cages.
2.3.2. Sample collecting procedure
Approximately 24 h after surgery, the dialysis probes were connected to a sy-
ringe pump and perfused with the aCSF at 1.0
m
l/min for 60 min (equilibration
period). To reliably determine the basal extracellular DA levels in the DStr vs. the
NAcc of freely moving mice, a quantitative low perfusionrate microdialysis
D. Leo et al. / Neuropharmacology 81 (2014) 283e291284
experiment was conducted (Gainetdinov et al., 2003). The perfusate was collected at
a perfusion rate of 0.1
m
l/min every 90 min over a 6 h period into collection tubes
containing 2
m
l of 1 M perchloric acid.
2.3.3. Analytical procedure
Measurements of DA, 5-HT and metabolites in tissue samples and DA in
microdialysis samples were performed by HPLC with electrochemical detection
(ALEXYS LC-EC system, Antec Leyden BV, Netherlands) with a 0.7 mm glass carbon
electrode (Antec; VT-03). The system was equipped with a reverse-phase column
(3
m
m particles, ALB-215 C18, 1 150 mm, Antec) at a ow rate of 200
m
l/min. The
mobile phase contained 50 mM H
3
PO
4
, 50 mM citric acid, 8 mM KCl, 0.1 mM EDTA,
400 mg/l octanesulfonic acid sodium salt and 10% (vol/vol) methanol, pH 3.9. The
sensitivity of this method permitted the detection of w3 fmol DA. Dialyzate samples
(11
m
l) were injected into HPLC without any additional purication.
2.4. Fast scan cyclic voltammetry (FSCV)
Briey, mice were anesthetized with halothane and decapitated. The brain was
sectioned in cold carboxygenated articial cerebrospinal uid (aCSF) (126 mM NaCl,
2.5 mM KCl, 1.2 mM NaH
2
PO
4
, 25 mM NaHCO
3
, 2.4 mM CaCl
2
,11mM
D
-glucose,
1.2 mM MgCl
2
) on a VT1000S vibrating microtome (Leica Microsystems, Nussloch,
Germany) at a thickness of 300
m
m. Coronal slices containing the dorsal striatum and
nucleus accumbens were allowed to recover for at least 1 h at room temperature in
carboxygenated aCSF. For recordings, slices were superfused with 32
C carboxy-
genated aCSF at a ow rate of 1 ml/min. Experimental recordings started 20 min
after transfer to the slice chamber.
Carbon ber electrodes (7
m
m, Goodfellow, Huntingdon, England) were made as
previously described (Kawagoe et al., 1993; Kuhr and Wightman, 1986). They were
trimmed to obtain a basal current between 140 and 180 nA. The electrodes were
inserted w100
m
m into the dorsal striatal brain slice. The potential of the working
electrode was held at 0.4 V vs. the Ag/AgCl control between scans and was ramped
to þ1.3 V at 300 V/s and back to 0.4 V every 100 ms via an EVA8 amplier (HEKA
Elektronik, Germany). Axonal DA release in the striatum was evoked using a twisted
bipolar stimulating electrode (Plastics One, Roanoke, VA). Stimulations were deliv-
ered every 2 min by a single electrical pulse (1 ms, monophasic single stimuli). The
stimulus was delivered via a stimulus isolator (AM-system, Carlsborg, WA).
Background-subtracted cyclic voltammograms were obtained by subtracting the
current obtained before the stimulation from all recordings. The peak oxidation
current for DA in each voltammogram was converted into a measure of the DA
concentration by post-calibration of the electrode using 1
m
M DA (SigmaeAldrich, St.
Louis, MO, USA). Data were normalized to the rst 5 recordings (10 min) of their
respective control period and graphically plotted against time (means SEM).
2.4.1. FSCV kinetic analysis
TarHeel CV (ESA Biosciences, Inc, Chelmsford, USA) was used for all data analysis.
Computations were based on user dened positions on current traces for baseline
(Pre-Stim cursor), peak (Peak cursor) and return to baseline (Post-Stim cursor) po-
sitions. Tau and half-life values were determined from exponential t curves based
on Peak cursor and Post-Stim cursor positions using a least squares constrained
exponential t algorithm (National Instruments, Milan, Italy). Both tau and half-life
are considered to be reliable measures for detecting changes in DA uptake and
accurately represent changes in uptake accordingly with more traditional measures
of the uptake (V
max
and K
m
), as demonstrated by Yorgason et al. (Yorgason et al.,
2011). Particularly, the rst-order rate constant (k,or1/
s
) provides a sensitive in-
dex of the efciency (V
max
/K
m
) of dopamine clearance mediated via the dopamine
transporter at low dopamine concentrations (Chen et al., 2008; Sabeti et al., 2002;
Bass et al., 2010).
2.5. Drugs
RO5166017 and N-(3-Ethoxy-phenyl)-4-pyrrolidin-1-yl-3-triuoromethyl-ben-
zamide (EPPTB, RO5212773) were generous gifts from F. Hoffmann-La Roche. These
drugs were dissolved in 1% DMSO as described (Revel et al., 2011; Bradaia et al.,
2009). All other drugs were purchased from SigmaeAldrich and were dissolved in
saline immediately before use.
2.6. Statistical analysis
The data are presented as the means SEM. Simple two-group comparisons
were performed using Studentst-test. The minimal signicance level for tests was
set at p<0.05. We elected to present normalized current values since multiple
groups were compared with each slice serving as its own precondition control. Thus
maximal DA concentrations were normalized to a respective baseline period rep-
resented the rst 10 min of stable recording before drug application.
One-way ANOVA with the pos-hoc Tukey test was used for multiple compari-
sons. All statistical analyses were performed using GraphPad Prism5 software
(GraphPad Software, Inc., La Jolla, CA).
3. Results
3.1. Lack of TAAR1 leads to an increased DA release predominantly
in the nucleus accumbens
Previous studies employing conventional microdialysis have
revealed similar basal levels of dialyzate DA in the NAcc and
striatum (Di Cara et al., 2011) of TAAR1-KO and WT mice, revealing
a difference in monoaminergic transmission only following
amphetamine challenge (Lindemann et al., 2008). To evaluate the
mechanisms of TAAR1-dependent modulation of DA and 5-HT
transmission, we rst analyzed the total tissue content of mono-
amines and their metabolites in different brain areas (Fig. 1). HPLC
analysis revealed that monoamines and their metabolite levels in
the prefrontal cortex and hippocampus are generally similar in both
genotypes (Figs. 1B and C, WT ¼10, HET ¼11, KO ¼11; Studentst-
test). However, while DA and its intraneuronal metabolite 3,4-
dihydroxyphenylacetic acid (DOPAC) levels were not altered in
the striatal tissue (including both DStr and NAcc; Fig. 1A) of TAAR1-
KO mice, the levels of predominantly extracellular DA metabolite
homovanillic acid (HVA) were elevated, suggesting an increase in
extracellular DA in mutant mice. Because most of the evidence
indicates a predominant effect of the TAAR1 regulatory effects in
the NAcc in comparison to the DStr (Revel et al., 2012b), we decided
to dissect the contribution of TAAR1 in different striatal subregions,
NAcc and DStr separately, using in vivo microdialysis (Fig. 2). Using
a quantitative low perfusion rate microdialysis approach that, un-
like conventional microdialysis, provides a true measure of extra-
cellular neurotransmitter concentrations (Justice, 1993), we found a
signicant increase in extracellular DA in the NAcc of TAAR1-KO
animals (Fig. 2A; WT, n¼5; TAAR1-KO, n¼5; p¼0.0412, Stu-
dentst-test) while DA levels were not changed in the DStr of
TAAR1-KO mice in comparison to controls (Fig. 2B; WT, n¼9;
TAAR1-KO, n¼7; p¼0.4379, Studentst-test).
Similar results were obtained ex vivo using FSCV performed on
brain slices (Fig. 3). FSCV was used to study the kinetics and the
amount of evoked DA release in the DStr and NAcc brain slices
prepared from TAAR1-KO mice and wild-type littermates. We
recorded DA overow following single pulses (400
m
A, 1 ms,
monophasic). The oxidation peak occurred at approximately þ0.6 V
and the reduction peak at approximately 0.2 V (Fig. 3A), consis-
tent with the voltammetric characteristics of DA. The maximal
amplitude of DA overow evoked by single pulses in the DStr was
stable over time and comparable in both the TAAR1-KO and control
mice (Fig. 3E), while in the TAAR1-KO NAcc slices the DA overow
was signicantly increased (Fig. 3B; WT, n¼18 vs. TAAR1-KO,
n¼19; p¼0.0293; Studentst-test). To determine whether the
difference in evoked the DA overow might be associated with an
unbalanced DA uptake (John and Jones, 2007), we evaluated the
uptake kinetics from an exponential t curve using a least squares-
constrained exponential t algorithm (National Instruments in
Demon Voltammetry software, Wake Forest University Health
Sciences, USA) (Yorgason et al., 2011) and quantied the Tau and
half-life parameters for an estimation of DA uptake rates. We
observed that, under basal conditions, the DA uptake as determined
by Tau measurements was similar between the control and TAAR1-
KO animals (see Methods section for details; Fig. 3C and F) Under
basal conditions DA uptake was similar in WT and TAAR-1 KO mice:
NAcc: Tau WT ¼0.6880 0.127 s, N¼18; Ta u
KO ¼0.8927 0.1264 s, N¼19 ; p¼0.2618 Fig. 3C; DStr: Tau WT
0.5087 0.08085 s, N¼12; Tau KO ¼0.6942 0.1490 s, N¼12,
p¼0.2811; Studentst-test. Fig. 3F) suggesting that DAT function is
unaltered in mice lacking TAAR1. Additionally, the DA half life was
similar in both genotypes (see Methods section for details; Fig. 3D
and G) (NAcc: Half life WT ¼0.4751 0.08814 s, N¼18; p¼0.2624;
D. Leo et al. / Neuropharmacology 81 (2014) 283e291 285
Half life KO ¼0.6161 0.08718 s, N¼19; Fig. 3D; DStr: Half life
WT ¼0.3310 0.04690 s, N¼12; Half life KO ¼0.4088 0.06821 s,
N¼24; p¼0.3380; Student t-test. Fig. 3G).
3.2. Selective TAAR1 agonist RO5166017 reduces DA overow
To further elucidate the role of TAAR1 in DA transmission, we
next evaluated the effect of the selective TAAR1 agonist RO5166017
on electrically evoked DA overow in the NAcc and DStr in WT and
TAAR1-KO mice (Fig. 4). The effects of two concentrations of TAAR1
agonist (1
m
M and 10
m
M) were tested using single electrical pulses.
While no signicant effect was found upon the application of 1
m
M
RO5166017 to brain slices (data not shown), 10
m
M of RO5166017
was able to reduce the evoked DA release selectively in control
animals in both the DStr (Fig. 4A; One-way ANOVA, p<0.0001) and
the NAcc (Fig. 4B; n¼10; One-way ANOVA, p<0.0001) without
signicant effects on the Tau and half-life measures of DA uptake
(data not shown). Diminished DA release following RO5166017
seems to be in accordance with a TAAR1 agonist ability to modulate
DA-related functions (Revel et al., 2011).
3.3. TAAR1 antagonist EPPTB increases evoked DA release and
blocks the TAAR1 agonist effect
Previous studies have already demonstrated that treatment
with EPPTB, under current-clamp conditions, signicantly
increased the ring frequency of VTA dopaminergic neurons over
the basal level (Bradaia et al., 2009). We tested the effect of EPPTB
on evoked DA release in the NAcc and DStr by applying 10
m
M of the
TAAR1 antagonist to brain slices (Fig. 4C and D). EPPTB failed to
signicantly increase DA overow in the DStr of both control and
TAAR1-KO animals (Fig. 4C; n¼10; One-way ANOVA) but induced
an augmented DA levels in the NAcc of control mice (Fig. 4D, n¼6
One-way ANOVA, p¼0.0210). Thus, the EPPTB effect on evoked DA
release seems to be selective for the NAcc of control animals
(Fig. 4D). Moreover, we evaluated the ability of the TAAR1 antag-
onist to block the effects of the agonist (Fig. 4E). We applied 10
m
M
of EPPTB, and immediately after, 10
m
M of the TAAR1 agonist
(RO5166017) was added. We then measured the amplitude of the
evoked DA release in the WT NAcc. We found that pre-application
of 10
m
M EPPTB is able to block the decrease in the evoked DA
release induced by the TAAR1 agonist (Fig. 4E). None of the drugs
applied changed DA uptake because the Tau and half-life values
were comparable among the naïve and treated brain slices (data
not shown).
3.4. Alterations in the D2 class of DA autoreceptor-mediated
regulation of DA release in TAAR1-KO mice
Because there are several lines of evidence suggesting that
TAAR1 can modulate DA transmission via an interaction with the
DA receptor 2 (D2R) (Espinoza et al., 2011; Ledonne et al., 2010;
Revel et al., 2011), the activity of the D2 class of DA release-
regulating autoreceptors was evaluated in the NAcc of control
and KO animals (Fig. 5). DA autoreceptors of the D2 class regulate
the extracellular levels of DA through a negative feedback mecha-
nism where increasing agonist concentrations result in a reduction
in the ring rate, synthesis and release of DA (Maina and Mathews,
KO
WT
NAcc DStr
0
10
20
30
40
*
DA (nM)
0
10
20
30
40
DA (nM)
AB
Fig. 2. Low perfusion rate (LPR) microdialysis revealed that, while the DA levels are not
altered in the striatum of TAAR1-KO mice (B), the levels of DA in NAcc are elevated in
the mutants (A). *p<0.05, Studentst-test.
A
B
C
0
5
10
15
20
DA DOPAC HVA 5-HIAA 5-HT
*
WT
KO
STRIATUM
ng/mg wet tissue
0.0
0.5
1.0
1.5
NE DA HVA 5-HIAA 5-HT
FRONTAL CORTEX
ng/mg wet tissue
0.0
0.5
1.0
1.5
2.0
NE DA 5-HIAA 5-HT
HIPPOCAMPUS
ng/mg wet tissue
Fig. 1. HPLC analysis of total tissue content in total Str (A), the frontal cortex (B) and
the hippocampus (C). A) In the total striatum, the levels of DA and its intraneuronal
metabolite 3,4-dihydroxyphenylacetic acid (DOPAC) are not altered in TAAR1-KO mice,
but the levels of predominantly extracellular DA metabolite homovanillic acid (HVA)
are elevated in the mutants. (*p<0.05; one-way ANOVA, Tukeys test). (Band C)
Neither of the analytes were different in the frontal cortex or the hippocampus be-
tween the control and mutant mice.
D. Leo et al. / Neuropharmacology 81 (2014) 283e291286
2010; Gainetdinov et al., 1996). We evaluated the inhibitory effect of
the D2 class receptor agonist quinpirole and the TAAR1 agonist on
DA release in WT (Fig. 5A) and TAAR1-KO (Fig. 5B) mice using FSCV
in NAcc brain slices. In both W T and KO slices treated with 0.1
m
Mof
quinpirole, we observed a well-known decrease in DA release
(Martel et al., 2011; Fawaz et al., 2009; Maina and Mathews, 2010;
Phillips et al., 2003)(Fig. 5A and B). Furthermore, after a washing
period to restore the basal DA levels, we examined the combined
effect of application of the D2 and TAAR1 agonists on DA release. As
shown in Fig. 5, quinpirole and RO5166017 are able to decrease DA
overow in an additive manner in control animals (WT
quinpirole þRO5166017, vs. WT quinpirole, p¼0.0079, Studentst-
test, Fig. 5A) but not in TAAR1-KO mice (Fig. 5B), suggesting an
altered D2R autoreceptor-mediated regulation of DA release in the
absence of TAAR1.
As an additional approach to conrm the role of DA autor-
eceptors in TAAR1 neuromodulation, a paired-pulse stimulation
test was performed in slices from WT and TAAR1-KO mice (Fig. 5C).
In this voltammetric approach, the autoinhibitory effect of DA
release following the rst stimulation is detected by measuring the
magnitude of the decrease in response to the second stimulation
performed 500 ms later. It is believed that this DA release sup-
pression induced by autoinhibition is directly related to D2R class
autoreceptor activation (Phillips et al., 2002). We applied an elec-
trical paired pulse (2 pulses, 400 mA, 500 ms interval), and we
measured the difference between the DA released following the
rst stimulus (S1) and the second stimulus (S2) as an indication of
D2R autoreceptor activation (Fig. 5C). We found that the ratio of S2
to S1 is signicantly higher in the NAcc slices from TAAR1-KO mice
compared to the control mice, indicating that D2R autoinhibition is
indeed reduced by the lack of TAAR1 (N¼5, p¼0.0029, Studentst-
test). These results further support the role of the D2 class of DA
autoreceptors in TAAR1-mediated modulation of DA release.
4. Discussion
The aim of this study was to understand the mechanism of the
modulatory action of TAAR1 on presynaptic DA transmission. Our
-0.4 V
-0.4 V
1.3 V
V (vs Ag/AgCl)
-0.4 V 1.3 V
Dopamine
15 sec
0.5 μM
A
CB Nucleus Accumbens
Dorsal Striatum
D
FEG
WT KOWT KOWT KO
0.0
0.5
1.0
1.5
DA concentration ( M)
WT KO
WT KO
0.0
0.2
0.4
0.6
0.8
1.0
Tau ( s)
WT KO
0.0
0.2
0.4
0.6
0.8
1.0
Half life (s)
DA concentration (μ
μ
M)
*
0.0
0.5
1.0
1.5
2.0
Tau ( s)
0.0
0.5
1.0
1.5
Half life (s)
0.0
0.2
0.4
0.6
0.8
1.0
μ
Fig. 3. Dopamine (DA) release from mice brain slices. A) The background-subtracted cyclic voltammogram identies the detected analyte as DA. The color plots represent the
voltammetric currents (encoded in color in the zaxis) plotted against the applied potential (yaxis) and time (xaxis). (A; bottom) Representative traces in the NAcc of control and
TAAR1 mice exemplify the higher peak height in the TAAR1-KO NAcc. B) Quantication of evoked DA release in the NAcc of control and TAAR1-KO mice (*p<0.05, Studentst-test) C
and D) Kinetic analyses of DA reuptake. C) Half-life and Tau measures of DA uptake.Tau and half life were determined from an exponential t curve using a least squares constrained
exponential t algorithm (National Instruments in Demon Voltammetry software). The exponential decay constant Tau measures changes in dopamine uptake. TAAR1 mice and
controls have similar uptake kinetics as conrmed by DA half-life measurements (D). E) Quantication of the evoked DA release in the DStr of control and TAAR1-KO mice show
similar DA overow. FEand GF) Tau (F) and half life (G) of DA uptake do not differ between control and TAAR1-KO mice.
D. Leo et al. / Neuropharmacology 81 (2014) 283e291 287
data support the previous reports showing a close interaction be-
tween TAAR1 and the dopaminergic system (Lindemann et al.,
2008; Revel et al., 2011; Bradaia et al., 2009) and specically
highlights an interaction between the D2 class of autoreceptors and
TAAR1 receptors. While several recent studies have been per-
formed on understanding the role of TAAR1 in the modulation of
monoaminergic systems in general, we particularly focused on the
presynaptic mechanisms by analyzing the neurochemical effects of
TAAR1 and DA ligands and the consequencesof the lack of TAAR1 in
mice.
First, we found an increased level of the predominantly extra-
cellular DA metabolite HVA in striatal/accumbal tissue that includes
both the DStr and NAcc of TAAR1-KO animals, indirectly indicating
an increase in DA release in this brain area. Furthermore, in
microdialysis studies, we have observed elevated levels of extra-
cellular DA in the NAcc but not in the DStr of mutant mice. In
agreement with these microdialysis studies, voltammetric in-
vestigations of evoked DA release have also demonstrated an
increased DA release selectively in the NAcc of TAAR1-KO mice. To
our knowledge, this is the rst report to show a signicant increase
in DA release in TAAR1-KO mice. Previous microdialysis studies
have reported markedly enhanced effects of amphetamine and
MDMA on the extracellular levels of monoamines in independently
developed strains of TAAR1-KO mice (Lindemann et al., 2008; Di
Cara et al., 2011; Wolinsky et al., 2007), but not altered basal
levels of DA in the NAcc and DStr. However, all of these measure-
ments were performed by using conventional microdialysis, an
approach known to be limited for assessment of basal absolute
extracellular levels. We therefore employed a low perfusion rate
quantitative microdialysis, providing an opportunity to overcome
the technical limitations of conventional microdialysis in the
assessment of basal neurotransmitter concentrations (Di Chiara
et al., 1996; Chefer et al., 2009; Handbook of Microdialysis, 2007).
Importantly, Lindemann et al. have found an increase in ring rates
Fig. 4. TAAR1-mediated modulation of evoked DA release. Aand B) Effects of
RO5166017 on evoked DA release in the DStr (A) and NAcc (B).10
m
M of TAAR1 agonist
decreases DA overow in both structures but only in control animals (*p<0.05, One-
way ANOVA, Tukeys test). Cand D)10
m
MofN-(3-Ethoxy-phenyl)-4-pyrrolidin-1-yl-
3-triuoromethyl-benzamide (EPPTB) does not inuence the evoked DA release in the
DStr (C) but increases the DA overow in control NAcc slices (D;*p<0.05, One-way
ANOVA, Tukey test). E) Pretreatment with 10
m
M EPPTB blocks the reduction of
evoked DA release mediated by 10
m
M RO5166017.
Fig. 5. 10
m
M of TAAR1 agonist and 0.1
m
M of quinpirole (D2R agonist) have an additive
effect on decreasing DA transmission in control animals (A) but not in TAAR1-KO
animals (B)(*p<0.05, One-way ANOVA with Bonferroni correction) suggesting a
regulatory action of TAAR1 on D2R signaling. C) After using paired stimuli to evoke DA
release in TAAR1-KO and control NAcc slices, TAAR1-KO mice have higher amplitude of
DA release following the second pulse of stimulation (*p<0.05, Studentst-test)
indicating that D2R-mediated autoinhibition is less active in the absence of TAAR1
gene.
D. Leo et al. / Neuropharmacology 81 (2014) 283e291288
in the VTA of TAAR1-KO animals, which is consistent with the
augmented DA release in the NAcc observed in our study. Thus,
TAAR1 seems to be tonically active in physiological conditions,
maintaining a negative control on DA release at least in the NAcc
(Lindemann et al., 2008). The lack of TAAR1 activity then induces a
substantially stronger enhancement in DA overow in the NAcc. In
agreement, TAAR1 expression was reported in the VTA (Lindemann
et al., 2008) that mainly projects to the NAcc, among other areas.
Importantly, we have documented that neither Tau nor the half-life
of released DA are changed in slices from TAAR1-KO animals,
indicating that TAAR1-KO mice exhibit unaltered DA uptake ability
and thereby normal dopamine transporter (DAT) functionality. It is
believed that Tau and the half-life of released DA are reliable
measures for detecting changes in DA uptake because they are
strongly correlated with changes in the K
m
of DAT mediated DA
uptake (Yorgason et al., 2011). Thus, these neurochemical in vivo
studies, as well as previous demonstrations of the functional ac-
tivity of TAAR1 ligands in mice lacking the DAT (Sotnikova et al.,
2004; Revel et al., 2012a), provide little support for the postu-
lated role of TAAR1 in modulating DAT activity that is based mostly
on in vitro cell culture experiments (Miller et al., 2005; Xie et al.,
2008; Miller, 2011).
We have also found that the TAAR1 agonist is able to reduce
evoked DA release in control animals but that it is ineffective in
TAAR1-KO mice. RO5166017 exhibits a high binding afnity for
mouse TAAR1 and a high potency to stimulate cAMP production,
and it is known to inhibit the VTA ring rate (Revel et al., 2011). In
our ex vivo experiments, 10
m
M RO5166017 exerts an effect on DA
release in both the NAcc and the DStr (Fig. 4), suggesting that while
the inuence of TAAR1 over DA transmission is predominant in the
NAcc, it might also occur at the level of the DStr. The concentration
of the TAAR1 agonist seems to be quite high compared to the one
used in in vitro experiments (Revel et al., 2011), but generally,
voltammetric slice studies require application of higher concen-
trations of drugs that differ substantially from those used in the
intact organism or in cultured cells (Borland et al., 2007). As
RO5166017 is able to reduce DA release, TAAR1 antagonist EPPTB
has the opposite effect and not only induces an augmentation of
evoked DA release but also blocks the agonist effect (Bradaia et al.,
2009). Importantly, both the effects of the TAAR1 agonist
(RO5166017) and antagonist (EPPTB) are specic for wild-type
animals and do not affect the kinetics of evoked DA release in
TAAR1-KO mice. Notably, neither of these treatments changed the
kinetics of DA uptake as evidenced by the Tau and DA half-life es-
timations, indicating that DAT-mediated function is not altered by
the action of the drugs on TAAR1. This is in line with the fact that
TAAR1 agonists have a pronounced inhibitory action on DA-
dependent hyperactivity in mice lacking the DAT (Revel et al.,
2011), ruling out the contribution of DAT at least these functional
effects of selective TAAR1 ligands.
Because we have not found evidence of the postulated
involvement of DAT in the effects of TAAR1 in our in vivo studies, we
explored an alternative potential mechanism of action by which
TAAR1 can inuence DA release. Thus, we focused our attention on
the status of the DA D2 class receptor (D2R) autoreceptors, per-
forming FSCV in the presence of the D2R agonist quinpirole.
Importantly, while D2 DA autoreceptors have a predominant role in
presynaptic autoinhibition of ring rates, synthesis and release
(Anzalone et al., 2012), an important contribution of the DA D3
autoreceptors on DA release regulation has also been demonstrated
(Accili et al., 1996; Joseph et al., 2002; Gainetdinov et al., 1996).
While we are not aware of any studies focused on the interaction
between TAAR1 and the DA D3 receptor, there are several lines of
evidence indicating a role of D2 receptors in functions mediated by
TAAR1. Our group has previously demonstrated that TAAR1 is able
to physically and functionally interact with the D2 receptors,
potentially via heterodimerization both in vitro and in vivo
(Espinoza et al., 2011). Moreover, it has been reported that TAAR1-
KO mice have a larger proportion of striatal D2 receptors that are in
a high-afnity state (Wolinsky et al., 2007). Because FSCV provides
an opportunity to follow real-time inhibition of DA release by
autoreceptors (Palij et al., 1990; Kennedy et al., 1992), we used an
application of 0.1
m
M quinpirole to NAcc slices, which has a well-
known effect of decreasing the evoked DA release (Yorgason
et al., 2011). A signicant quinpirole-induced reduction in DA
release was observed in both TAAR1-KO and WT mice, indicating a
preserved D2R autoreceptor-mediation in the absence of TAAR1.
However, the D2R agonist showed an additive effect when com-
bined with the TAAR1 agonist RO5166017 in WT mice, but not in
TAAR1-decient animals. Presumably, there is an increase in the
D2R-mediated autoinhibition of DA neurons under tonic activation
of TAAR1 that supports the role of TAAR1 as a homeostatic regu-
latory mechanism preventing the excess activity of DA neurons. DA
autoreceptors are known to exert their effects by down-regulating
adenylate cyclase via a G protein-coupled mechanism (Missale
et al., 1998), and it can decrease tyrosine hydroxylase activity by
phosphorylation. Importantly, it has been reported that TAAR1-KO
mice have an increased basal phosphorylation state of tyrosine
hydroxylase at Ser19, Ser31, and Ser40 (Di Cara et al., 2011).
Moreover, it has been proposed that the D2R autoreceptor-
mediated modulation of secretion might occur via modulation of
potassium and possibly calcium channels in the adenylate cyclase-
independent pathway (Beaulieu and Gainetdinov, 2011). TAAR1
induces a G protein-dependent inwardly rectifying K
þ
current that
is inhibited by Ba
2þ
and tertiapin, suggesting that TAAR1 reduces
the ring rate of DA neurons by activating Kir3 channels (Bradaia
et al., 2009). G protein-mediated activation of K
þ
channels gener-
ally hyperpolarizes the membrane, thus decreasing the probability
of release.
Finally, to further investigate the relationship between the level
of autoreceptor stimulation and the resulting inhibition of DA
release, we used paired stimuli to evoke DA release in TAAR1-KO
and control NAcc slices. In this paradigm, DA released by the rst
pulse activates D2R autoreceptors and thus results in less amount
of DA release evoked by the following pulse. In this test, TAAR1-KO
animals have a higher amplitude of DA release following the second
pulse of stimulation, thus directly indicating that D2R-mediated
autoinhibition is less active in the absence of TAAR1 (Phillips
et al., 2002). Importantly, while both presynaptic and post-
synaptic D2 receptors are present in the recording site and there is
growing evidence that both these populations could be regulated
by TAAR1 (Borowsky et al., 2001; Espinoza et al., 2011) only the
former are involved in DA autoinhibition. Indeed, DA autoinhibition
is still observed in the striatum of mice lacking only postsynaptic
D2 receptors (Benoit-Marand et al., 2001; Usiello et al., 2000).
These observations, demonstrating ability of TAAR1 to modulate
presynaptic DA autoreceptor function suggest intriguing new pos-
sibility how TAs may inuence monoaminergic transmission in
general. Thus, not only direct activation of D2R autoreceptors by
released DA but also action of endogenous TAAR1 ligands such as
TAs and the extracellular DA metabolite 3-methoxytyramine on
TAAR1 (Bunzow et al., 2001; Sotnikova et al., 2010) may exert ne-
tuning regulatory action on presynaptic dopaminergic trans-
mission. It would be interesting to explore if this presynaptic
autoreceptor-based mechanism of action could be extended to
other TAs and monoaminergic systems.
In conclusion, we report that TAAR1 is able to regulate DA
release predominantly in the NAcc by exerting a negative modu-
lation of DA tone. Thus, TAAR1-KO mice have an increased evoked
DA release compared to control mice. The uptake rates are similar
D. Leo et al. / Neuropharmacology 81 (2014) 283e291 289
between the two groups of mice, excluding an involvement of DAT
in the modulatory action of TAAR1. We also observed a functional
link between TAAR1 and D2R autoreceptors localized on DA ter-
minals, further indicating that TAAR1 and D2R can modulate each
others activity. These observations uncover a mechanism of close
interaction between TAAR1 and the DA system at the level of the
presynaptic neurons and autoreceptor regulation that further
promotes the general strategy of targeting TAAR1 to modulate the
dopaminergic system in a range of neuropsychiatric disorders.
Acknowledgments
This work was supported in part by research awards to RRG from
F. Hoffmann-La Roche Ltd. (Basel, Switzerland) and Fondazione
Compagnia di San Paolo (Torino, Italy). We are grateful to Lundbeck
A/G and Lundbeck USA for generously providing the TAAR1
knockout mice. We thank Dr. M. Morini, D. Cantatore and F. Piccardi
for their excellent technical assistance.
References
Accili, D., et al., 1996. A targeted mutation of the D3 dopamine receptor gene is
associated with hyperactivity in mice. Proc. Natl. Acad. Sci. USA 93, 1945e194 9.
Anzalone, A., et al., 2012. Dual control of dopamine synthesis and release by pre-
synaptic and postsynaptic dopamine D2 receptors. J. Neurosci. 32, 9023e9034.
Axelrod, J., Saavedra, J.M., 1977. Octopamine. Nature 265, 501e504.
Bass, C.E., Grinevich, V.P., Vance, Z.B., Sullivan, R.P., Bonin, K.D., Budygin, E.A., 2010.
Optogenetic control of striatal dopamine release in rats. J. Neurochem. 114,
134 4e1352.
Beaulieu, J.-M., Gainetdinov, R.R., 2011. The physiology, signaling, and pharma-
cology of dopamine receptors. Pharmacol. Rev. 63, 182e217.
Benoit-Marand, M., Borrelli, E., Gonon, F., 2001. Inhibition of dopamine release via
presynaptic D2 receptors: time course and functional characteristics in vivo.
J. Neurosci. 21, 9134e9141.
Berry, M.D., 2004. Mammalian central nervous system trace amines. Pharmacologic
amphetamines, physiologic neuromodulators. J. Neurochem. 90, 257e271.
Borland, L.M., Michael, A.C., 2007. In: Michael, A.C., Borland, L.M. (Eds.), Electro-
chemical Methods for Neuroscience. CRC Press.
Borowsky, B., et al., 2001. Trace amines: identication of a family of mammalian G
protein-coupled receptors. Proc. Natl. Acad. Sci. USA 98, 8966e8971.
Boulton, A.A., 1980. Trace amines and mental disorders. Can. J. Neurol. Sci. 7, 261e
263.
Bradaia, A., et al., 2009. The selective antagonist EPPTB reveals TAAR1-mediated
regulatory mechanisms in dopaminergic neurons of the mesolimbic system.
Proc. Natl. Acad. Sci. USA 106, 20081e20086.
Branchek, T.A., Blackburn, T.P., 2003. Trace amine receptors as targets for novel
therapeutics: legend, myth and fact. Curr. Opin. Pharmacol. 3, 90e97.
Budygin, E.A., et al., 2004. Dissociation of rewarding and dopamine transporter-
mediated properties of amphetamine. Proc. Natl. Acad. Sci. USA 101, 7781e
7786.
Bunzow, J.R., et al., 2001. Amphetamine, 3,4-methylenedioxymethamphetamine,
lysergic acid diethylamide, and metabolites of the catecholamine neurotrans-
mitters are agonists of a rat trace amine receptor. Mol. Pharmacol. 60, 1181e
1188.
Carboni, E., et al., 2001. Cocaine and amphetamine increase extracellular dopamine
in the nucleus accumbens of mice lacking the dopamine transporter gene.
J. Neurosci. 21 (RC141), 1e4.
Chefer, V.I., Thompson, A.C., Zapata, A., Shippenberg, T.S., 2009. Overview of brain
microdialysis. Curr. Protoc. Neurosci. CHAPTER 7, Unit7.1.
Chen, Y.-H., et al., 2008. MPTP-induced decits in striatal synaptic plasticity are
prevented by glial cell line-derived neurotrophic factor expressed via an adeno-
associated viral vector. FASEB J. 22, 261e275.
Cichero, E., Espinoza, S., Gainetdinov, R.R., Brasili, L., Fossa, P., 2013. Insights into the
structure and pharmacology of the human trace amine-associated receptor 1
(hTAAR1): homology modelling and docking studies. Chem. Biol. Drug. Des. 81,
509e516.
Cooper, S.E., Venton, B.J., 2009. Fast-scan cyclic voltammetry for the detection of
tyramine and octopamine. Anal. Bioanal. Chem. 394, 329e336.
Di Cara, B., et al., 2011. Genetic deletion of trace amine 1 receptors reveals their role
in auto-inhibiting the actions of ecstasy (MDMA). J. Neurosci. 31, 16928e16940.
Di Chiara, G., Tanda, G., Carboni, E., 1996. Estimation of in-vivo neurotransmitter
release by brain microdialysis: the issue of validity. Behav. Pharmacol. 7, 640e
657.
Espinoza, S., et al., 2011. Functional interaction between trace amine-associated
receptor 1 and dopamine D2 receptor. Mol. Pharmacol. 80, 416e425.
Fawaz, C.S., Martel, P., Leo, D., Trudeau, L.-E., 2009. Presynaptic action of neuro-
tensin on dopamine release through inhibition of D2 receptor function. BMC
Neurosci. 10, 96.
Franklin, K., Paxinos, G., 1997. The Mouse Brain in Stereotaxic Coordinates. Aca-
demic Press, San Diego. ISBN 0-12-26607-6.
Gainetdinov, R.R., et al., 2003. Dopaminergic supersensitivity in G protein-coupled
receptor kinase 6-decient mice. Neuron 38, 291e303.
Gainetdinov, R.R., Sotnikova, T.D., Grekhova, T.V., Rayevsky, K.S., 1996. In vivo evi-
dence for preferential role of dopamine D3 receptor in the presynaptic regu-
lation of dopamine release but not synthesis. Eur. J. Pharmacol. 308, 261e269.
Grandy, D.K., 2007. Trace amine-associated receptor 1 efamily archetype or icon-
oclast? Pharmacol. Ther. 116, 355e390.
Handbook of Microdialysis: Methods, Applications and Perspectives, 2007. Aca-
demic Press.
John, C.E., Jones, S.R., 2007. Voltammetric characterization of the effect of mono-
amine uptake inhibitors and releasers on dopamine and serotonin uptake in
mouse caudate-putamen and substantia nigra slices. Neuropharmacology 52,
1596e1605.
Jones, S.R., Gainetdinov, R.R., Wightman, R.M., Caron, M.G., 1998. Mechanisms of
amphetamine action revealed in mice lacking the dopamine transporter.
J. Neurosci. 18, 1979e1986.
Joseph, J.D., et al., 2002. Dopamine autoreceptor regulation of release and
uptake in mouse brain slices in the ab sence of D(3) receptors. Neuroscience
112, 39e49.
Justice Jr., J.B., 1993. Quantitative microdialysis of neurotransmitters. J. Neurosci.
Methods 48, 263e276.
Kawagoe, K.T., Zimmerman, J.B., Wightman, R.M., 1993. Principles of voltammetry
and microelectrode surface states. J. Neurosci. Methods 48, 225e240.
Kennedy, R.T., Jones, S.R., Wightman, R.M., 1992. Dynamic observation of dopamine
autoreceptor effects in rat striatal slices. J. Neurochem. 59, 449e455.
Kuhr, W.G., Wightman, R.M., 1986. Real-time measurement of dopamine release in
rat brain. Brain Res. 381, 168e17 1.
Ledonne, A., et al., 2010. Trace amines depress D(2)-autoreceptor-mediated re-
sponses on midbrain dopaminergic cells. Br. J. Pharmacol. 160, 1509e1520.
Lindemann, L., et al., 20 05. Trace amine-associated receptors form structurally and
functionally distinct subfamilies of novel G protein-coupled receptors. Geno-
mics 85, 372e385.
Lindemann, L., et al., 2008. Trace amine-associated receptor 1 modulates dopami-
nergic activity. J. Pharmacol. Exp. Ther. 324, 948e956.
Lindemann, L., Hoener, M.C., 2005. A renaissance in trace amines inspired by a novel
GPCR family. Trends Pharmacol. Sci. 26, 274e281.
Maguire, J.J., et al., 2009. International Union of Pharmacology. LXXII. Recommen-
dations for trace amine receptor nomenclature. Pharmacol. Rev. 61, 1e8.
Maina, F.K., Mathews, T.A., 2010. Functional fast scan cyclic voltammetry assay to
characterize dopamine D2 and D3 autoreceptors in the mouse striatum. ACS
Chem. Neurosci. 1, 450e462.
Martel, P., Leo, D., Fulton, S., Bérard, M., Trudeau, L.-E., 2011. Role of Kv1 potassium
channels in regulating dopamine release and presynaptic D2 receptor function.
PLoS ONE 6, e20402.
Miller, G.M., 2011. The emerging role of trace amine-associated receptor 1 in the
functional regulation of monoamine transporters and dopaminergic activity.
J. Neurochem. 116, 164e176 .
Miller, G.M., et al., 2005. Primate trace amine receptor 1 modulation by the dopa-
mine transporter. J. Pharmacol. Exp. Ther. 313, 983e994.
Missale, C., Nash, S.R., Robinson, S.W., Jaber, M., Caron, M.G., 1998. Dopamine re-
ceptors: from structure to function. Physiol. Rev. 78, 189e225.
Palij, P., et al., 1990. Presynaptic regulation of dopamine release in corpus striatum
monitored in vitro in real time by fast cyclic voltammetry. Brain Res. 509, 172e
174.
Parker, E.M., Cubeddu, L.X., 1988. Comparative effects of amphetamine, phenyl-
ethylamine and related drugs on dopamine efux, dopamine uptake and
mazindol binding. J. Pharmacol. Exp. Ther. 245, 199e210.
Phillips, P.E.M., et al., 2003. Presynaptic dopaminergic function is largely unaltered
in mesolimbic and mesostriatal terminals of adult rats that were prenatally
exposed to cocaine. Brain Res. 961, 63e72.
Phillips, P.E.M., Hancock, P.J., Stamford, J.A., 2002. Time window of autoreceptor-
mediated inhibition of limbic and striatal dopamine release. Synapse 44, 15e
22.
Reese, E.A., et al., 2014. Exploring the determinants of trace amine-associated re-
ceptor 1s functional selectivity for the stereoisomers of amphetamine and
methamphetamine. J. Med. Chem. 57, 378e390.
Revel, F.G., et al., 2011. TAAR1 activation modulates monoaminergic neurotrans-
mission, preventing hyperdopaminergic and hypoglutamatergic activity. Proc.
Natl. Acad. Sci. USA 108, 8485e8490.
Revel, F.G., et al., 2012a. Trace amine-associated receptor 1 partial agonism re-
veals novel paradigm for neuropsychiatric therapeutics. Biol. Psychiatry 72,
934e942.
Revel, F.G., et al., 2012b. Brain-specic overexpression of trace amine-associated
receptor 1 alters monoaminergic neurotransmission and decreases sensitivity
to amphetamine. Neuropsychopharmacology 37, 2580e2592.
Revel, F.G., et al., 2013. A new perspective for schizophrenia: TAAR1 agonists reveal
antipsychotic- and antidepressant-like activity, improve cognition and control
body weight. Mol. Psychiatry 18, 543e556.
Sabeti, J., Adams, C.E., Burmeister, J., Gerhardt, G.A., Zahniser, .R., 2002. Kinetic
analysis of striatal clearance of exogenous dopamine recorded by chro-
noamperometry in freely-moving rats. J. Neurosci. Methods 121, 41e52.
Sandler, M., et al., 1980. Trace amine de cit in depressive illness: the phenylalanine
connexion. Acta Psychiatr. Scand. Suppl. 280, 29e39.
D. Leo et al. / Neuropharmacology 81 (2014) 283e291290
Sotnikova, T.D., et al., 2004. Dopamine transporter-dependent and -inde-
pendent actions of trace amine beta-phenylethylamine. J. Ne urochem. 91,
362e373.
Sotnikova, T.D., et al., 2010. The dopamine metabolite 3-methoxytyramine is a
neuromodulator. PLoS ONE 5, e13452.
Sotnikova, T.D., Caron, M.G., Gainetdinov, R.R., 2009. Trace amine-associated re-
ceptors as emerging therapeutic targets. Mol. Pharmacol. 76, 229e235.
Usiello, A., et al., 2000. Distinct functions of the two isoforms of dopamine D2 re-
ceptors. Nature 408, 199e203.
Wolinsky, T.D., et al., 20 07. The Trace Amine 1 receptor knockout mouse: an animal
model with relevance to schizophrenia. Genes Brain Behav. 6, 628e639.
Xie, Z., Westmoreland, S.V., Miller, G.M., 2008. Modulation of monoamine trans-
porters by common biogenic amines via trace amine-associated receptor 1 and
monoamine autoreceptors in human embryonic kidney 293 cells and brain
synaptosomes. J. Pharmacol. Exp. Ther. 325, 629e640.
Yorgason, J.T., España, R.A., Jones, S.R., 2011. Demon voltammetry and analysis
software: analysis of cocaine-induced alterations in dopamine signaling using
multiple kinetic measures. J. Neurosci. Methods 202, 158e164.
D. Leo et al. / Neuropharmacology 81 (2014) 283e291 291
... They stimulate the production of cAMP via Gs-protein coupling and also recruit a second signalling pathway, the β-arrestin2 cascade; second messenger systems that also mediate dopamine D2 receptor activity (Espinoza et al., 2013). Early observations indicated that TAAR1 influenced dopaminergic neurotransmission (Lindemann et al., 2008); it was subsequently reported to have effects through the modulation of presynaptic D2 receptors (Leo et al., 2014). Notable in the current context is that this modulation appears to be through TAAR1 agonist-induced attenuation of the function of the dopamine D2 receptor: the two receptors act in opposing ways on second messenger systems as TAAR1 has excitatory effects via stimulation of cAMP formation, while D2 receptor activity inhibits this process. ...
Article
Full-text available
A major effort of the pharmaceutical industry has been to identify and market drug treatments that are effective in ameliorating the symptoms of psychotic illness but without the limitations of the current treatments acting at dopamine D2 receptors. These limitations include the induction of a range of adverse effects, the inadequate treatment response of a substantial proportion of people with schizophrenia, and the generally poor response to negative and cognitive features of the disease. Recently introduced drug treatments have gone some way to avoiding the first of these, with a reduced propensity for weight gain, cardiovascular risk and extrapyramidal motor effects. Despite claims of some small improvements in negative symptoms, these drugs have not demonstrated substantial increases in efficacy. Of the drugs currently in development as antipsychotic agents, several are misleadingly described as having novel ‘non-dopaminergic’ mechanisms that may offer improvements in addressing the limitations of adverse effects and efficacy. It will be argued, using the trace amine-associated receptor 1 agonist as an example, that several of these new drugs still act primarily through modulation of dopaminergic neurotransmission and, in not addressing the primary pathology of schizophrenia, are therefore unlikely to have the much-needed improvements in efficacy required to address the unmet need associated with resistance to current treatments.
... Establishing animal models such as TAAR1 knock-out mice and TAAR1 overexpressing mice, 55,60 followed by selective TAAR1 agonists and antagonists development, 61-63 enabled researchers to discover the connection between TAAR1 activity and neurotransmission in dopaminergic, serotonergic, and glutamatergic pathways. Several studies showed that TAAR1 stabilizes dopamine and serotonin neurotransmission by modulating pre-synaptic and post-synaptic D 2 receptors and 5-HT 1A receptors, 62,64,65 respectively. Research conducted using TAAR1 knockout mice has illustrated that they have an increased proportion of high-affinity state D 2 receptors in the striatum, which translates into their greater amphetamine sensitivity compared to wild-type mice. ...
Article
Despite extensive research efforts aimed at discovering novel antipsychotic compounds, a satisfactory pharmacological strategy for schizophrenia treatment remains elusive. All the currently available drugs act by modulating dopaminergic neurotransmission, leading to insufficient management of the negative and cognitive symptoms of the disorder. Due to these challenges, several attempts have been made to design agents with innovative, non‐dopaminergic mechanisms of action. Consequently, a number of promising compounds are currently progressing through phases 2 and 3 of clinical trials. This review aims to examine the rationale behind the most promising of these strategies while simultaneously providing a comprehensive survey of study results. We describe the versatility behind the cholinergic neurotransmission modulation through the activation of M 1 and M 4 receptors, exemplified by the prospective drug candidate KarXT. Our discussion extends to the innovative approach of activating TAAR1 receptors via ulotaront, along with the promising outcomes of iclepertin, a GlyT‐1 inhibitor with the potential to become the first treatment option for cognitive impairment associated with schizophrenia. Finally, we evaluate the 5‐HT 2A antagonist paradigm, assessing two recently developed serotonergic agents, pimavanserin and roluperidone. We present the latest advancements in developing novel solutions to the complex challenges posed by schizophrenia, offering an additional perspective on the diverse investigated drug candidates.
... Due to these reasons, the link between TAAR1 and PD pathology is still not clear in the present study. TAAR1 knockout animals might give some clues, but most of the previous studies were conducted in TAAR1 conventional knockout mice [65,67,68]. A cell-type-specific or region-specific TAAR1 knockout mice should be used in future studies. ...
Article
Full-text available
Parkinson’s disease (PD) is characterized not only by motor symptoms but also by non-motor dysfunctions, such as olfactory impairment; the cause is not fully understood. Our study suggests that neuronal loss and inflammation in brain regions along the olfactory pathway, such as the olfactory bulb (OB) and the piriform cortex (PC), may contribute to olfactory dysfunction in PD mice, which might be related to the downregulation of the trace amine-associated receptor 1 (TAAR1) in these areas. In the striatum, although only a decrease in mRNA level, but not in protein level, of TAAR1 was detected, bioinformatic analyses substantiated its correlation with PD. Moreover, we discovered that neuronal death and inflammation in the OB and the PC in PD mice might be regulated by TAAR through the Bcl-2/caspase3 pathway. This manifested as a decrease of anti-apoptotic protein Bcl-2 and an increase of the pro-apoptotic protein cleaved caspase3, or through regulating astrocytes activity, manifested as the increase of TAAR1 in astrocytes, which might lead to the decreased clearance of glutamate and consequent neurotoxicity. In summary, we have identified a possible mechanism to elucidate the olfactory dysfunction in PD, positing neuronal damage and inflammation due to apoptosis and astrocyte activity along the olfactory pathway in conjunction with the downregulation of TAAR1.
... DA release is also stimulated by TAAR1 antagonists and inhibited by TAAR1 agonists in wild-type mice. However, TAAR1 insufficiency does not affect the kinetics of DA uptake and DAT function [128,130]. ...
Article
Full-text available
Currently, metabolic syndrome treatment includes predominantly pharmacological symptom relief and complex lifestyle changes. Trace amines and their receptor systems modulate signaling pathways of dopamine, norepinephrine, and serotonin, which are involved in the pathogenesis of this disorder. Trace amine-associated receptor 1 (TAAR1) is expressed in endocrine organs, and it was revealed that TAAR1 may regulate insulin secretion in pancreatic islet β-cells. For instance, accumulating data demonstrate the positive effect of TAAR1 agonists on the dynamics of metabolic syndrome progression and MetS-associated disease development. The role of other TAARs (TAAR2, TAAR5, TAAR6, TAAR8, and TAAR9) in the islet’s function is much less studied. In this review, we summarize the evidence of TAARs’ contribution to the metabolic syndrome pathogenesis and regulation of insulin secretion in pancreatic islets. Additionally, by the analysis of public transcriptomic data, we demonstrate that TAAR1 and other TAAR receptors are expressed in the pancreatic islets. We also explore associations between the expression of TAARs mRNA and other genes in studied samples and demonstrate the deregulation of TAARs’ functional associations in patients with metabolic diseases compared to healthy donors.
... D2 dopamine D2 receptor, 5-HT1A and 5-HT2A serotonin 1A and 2A receptor subtypes, ADHD attention deficit disorder with hyperactivity. *Antipsychotic (purple) and high-dose antipsychotic (dark purple); **antidepressant (green) and high-dose antidepressant (dark green) electrically evoked dopamine release are reduced by TAAR1 full agonists, whereas generally opposite effects are seen with the TAAR1 antagonists such as N-(3-Ethoxy-phenyl)-4-pyrrolidin-1-yl-3-trifluoromethyl-benzamide (EPPTB) as well as in TAAR1-KO mice [48][49][50][51][52]. Interestingly, the inhibitory effects on dopaminergic neurotransmission appear to be most pronounced under hyperdopaminergic conditions. ...
Article
Full-text available
Ulotaront is a trace amine-associated receptor 1 (TAAR1) agonist in Phase 3 clinical development for the treatment of schizophrenia. Ulotaront was discovered through a unique, target-agnostic approach optimized to identify drug candidates lacking D2 and 5-HT2A receptor antagonism, while demonstrating an antipsychotic-like phenotypic profile in vivo. The mechanism of action (MOA) of ulotaront is thought to be mediated by agonism at TAAR1 and serotonin 5-HT1A receptors. Ulotaront has completed two Phase 2 trials (4-week acute study and 26-week open-label extension) which led to Breakthrough Therapy Designation from the US Food and Drug Administration for the treatment of schizophrenia. In the double-blind, placebo-controlled, acute study, ulotaront was associated with significant (p < 0.001) improvement in Positive and Negative Syndrome Scale (PANSS) total score (effect size [ES]: 0.45), with improvements vs. placebo also observed across secondary endpoints. Post-hoc analyses of the acute trial revealed additional evidence to support the effect of ulotaront on negative symptoms. In the 4-week study, ulotaront was well-tolerated, with an incidence of adverse events (AEs) numerically lower compared to placebo (45.8% vs. 50.4%; with a number needed to harm [NNH] for individual ulotaront AEs all > 40). The open-label extension demonstrated further improvement across schizophrenia symptoms and confirmed the tolerability of ulotaront, with a 6-month completion rate of 67%. Based on current data, ulotaront shows potential to be a first-in-class TAAR1 agonist for the treatment of schizophrenia with a safety and efficacy profile distinct from current antipsychotics.
... Specifically, TAAR1 activation can stimulate the Gβγ-dependent inwardly rectifying potassium channel Kir3 to induce potassium efflux. Given potassium efflux through Kir3, it is proposed that TAAR1 agonists may reduce the positive symptoms of schizophrenia by decreasing dopaminergic neuronal firing in the ventral tegmental area ( Figure 1) [30,31]. ...
Article
Introduction: Schizophrenia is a mental illness that can disrupt emotions, perceptions, cognition, and reduce quality of life. The classical approach to treat schizophrenia uses typical and atypical antipsychotics; however, limitations include low efficacy in mitigating negative symptoms and cognitive dysfunctions, and a range of adverse effects. Evidence has accumulated on trace amine-associated receptor 1 (TAAR1) as a novel therapeutic target for treating schizophrenia. This systematic review investigates the available evidence on a TAAR1 agonist, ulotaront, as a treatment for schizophrenia. Methods: A systematic search was conducted on PubMed/MEDLINE, and Ovid databases for English-published articles from inception to December 18, 2022. Literature focusing on the association between ulotaront and schizophrenia were evaluated based on an inclusion/exclusion criterion. Selected studies were assessed for risk of bias, using Cochrane Collaboration tool, and summarized in a table to generate discussion topics. Results: Three clinical, two comparative, and five preclinical studies examining ulotaront's pharmacology, tolerability and safety, and/or efficacy were identified. Results indicate that ulotaront has a differing adverse effects profile from other antipsychotics, may mitigate metabolic-related adverse effects commonly associated with antipsychotics, and may be effective for treating positive and negative symptoms. Conclusions: Findings from available literature present ulotaront as a potential and promising alternative treatment method for schizophrenia. Despite this, our results were limited due to lack of clinical trials on ulotaront's long-term efficacy and mechanisms of action. Future research should focus on these limitations to elucidate ulotaront's efficacy and safety for the treatment of schizophrenia and other mental disorders with similar pathophysiology.
Article
Schizophrenia (SZ) is a serious, destructive neurodevelopmental disorder. Antipsychotic medications are the primary therapy approach for this illness, but it's important to pay attention to the adverse effects as well. Clinical studies for SZ are currently in phase ΙΙΙ for SEP‐363856 (SEP‐856)‐a new antipsychotic that doesn't work on dopamine D 2 receptors. However, the underlying action mechanism of SEP‐856 remains unknown. This study aimed to evaluate the impact and underlying mechanisms of SEP‐856 on SZ‐like behavior in a perinatal MK‐801 treatment combined with social isolation from the weaning to adulthood model (MK‐SI). First, we created an animal model that resembles SZ that combines the perinatal MK‐801 with social isolation from weaning to adulthood. Then, different classical behavioral tests were used to evaluate the antipsychotic properties of SEP‐856. The levels of proinflammatory cytokines (tumor necrosis factor‐α, interleukin‐6, and interleukin‐1β), apoptosis‐related genes (Bax and Bcl‐2), and synaptic plasticity‐related genes (brain‐derived neurotrophic factor [BDNF] and PSD‐95) in the hippocampus were analyzed by quantitative real‐time PCR. Hematoxylin and eosin staining were used to observe the morphology of neurons in the hippocampal DG subregions. Western blot was performed to detect the protein expression levels of BDNF, PSD‐95, Bax, Bcl‐2, PI3K, p‐PI3K, AKT, p‐AKT, GSK‐3β, p‐GSK‐3β in the hippocampus. MK‐SI neurodevelopmental disease model studies have shown that compared with sham group, MK‐SI group exhibit higher levels of autonomic activity, stereotyped behaviors, withdrawal from social interactions, dysregulated sensorimotor gating, and impaired recognition and spatial memory. These findings imply that the MK‐SI model can mimic symptoms similar to those of SZ. Compared with the MK‐SI model, high doses of SEP‐856 all significantly reduced increased activity, improved social interaction, reduced stereotyping behavior, reversed sensorimotor gating dysregulation, and improved recognition memory and spatial memory impairment in MK‐SI mice. In addition, SEP‐856 can reduce the release of proinflammatory factors in the MK‐SI model, promote the expression of BDNF and PSD‐95 in the hippocampus, correct the Bax/Bcl‐2 imbalance, turn on the PI3K/AKT/GSK‐3β signaling pathway, and ultimately help the MK‐SI mice's behavioral abnormalities. SEP‐856 may play an antipsychotic role in MK‐SI “dual‐hit” model‐induced SZ‐like behavior mice by promoting synaptic plasticity recovery, decreasing death of hippocampal neurons, lowering the production of pro‐inflammatory substances in the hippocampal region, and subsequently initiating the PI3K/AKT/GSK‐3β signaling cascade.
Article
Trace amine-associated receptor 1 (TAAR1) is an intracellular expressed G-protein-coupled receptor that is widely expressed in major dopaminergic areas and plays a crucial role in modulation of central dopaminergic neurotransmission and function. Pharmacological studies have clarified the roles of dopamine D1 receptor (D1R) in the medial prefrontal cortex (mPFC) in cognitive function and social behaviors, and chronic stress can inhibit D1R expression due to its susceptibility. Recently, we identified TAAR1 in the mPFC as a potential target for treating chronic stress-induced cognitive and social dysfunction, but whether D1R is involved in mediating the effects of TAAR1 agonist remains unclear. Combined genomics and transcriptomic studies revealed downregulation of D1R in the mPFC of TAAR1-/- mice. Molecular dynamics simulation showed that hydrogen bond, salt bridge, and Pi-Pi stacking interactions were formed between TAAR1 and D1R indicating a stable TAAR1-D1R complex structure. Using pharmacological interventions, we found that D1R antagonist disrupted therapeutic effect of TAAR1 partial agonist RO5263397 on stress-related cognitive and social dysfunction. Knockout TAAR1 in D1-type dopamine receptor-expressing neurons reproduced adverse effects of chronic stress, and TAAR1 conditional knockout in the mPFC led to similar deficits, along with downregulation of D1R expression, all of these effects were ameliorated by viral overexpression of D1R in the mPFC, suggesting the functional interaction between TAAR1 and D1R. Collectively, our data elucidate the possible molecular mechanism that D1R in the mPFC mediates the effects of TAAR1 activation on chronic stress-induced cognitive and social deficits.
Article
Full-text available
Behavioral and biochemical studies suggest that dopamine (DA) plays a role in the reinforcing and addictive properties of drugs of abuse. Recently, this hypothesis has been challenged on the basis of the observation that, in mice genetically lacking the plasma membrane dopamine transporter [DAT-knock out (DAT-KO)], cocaine maintained its reinforcing properties of being self-administered and inducing place preference, despite the failure to increase extracellular dopamine in the dorsal striatum. Here we report that, in DAT-KO mice, cocaine and amphetamine increase dialysate dopamine in the medial part of the nucleus accumbens. Moreover, reboxetine, a specific blocker of the noradrenaline transporter, increased DA in the nucleus accumbens of DAT-KO but not of wild-type mice; in contrast, GBR 12909, a specific blocker of the dopamine transporter, increased dialysate dopamine in the nucleus accumbens of wild-type but not of DAT-KO mice. These observations provide an explanation for the persistence of cocaine reinforcement in DAT-KO mice and support the hypothesis of a primary role of nucleus accumbens dopamine in drug reinforcement.
Article
Full-text available
Trace amines (TAs) such as β-phenylethylamine, p-tyramine, or tryptamine are biogenic amines found in the brain at low concentrations that have been implicated in various neuropsychiatric disorders like schizophrenia, depression, or attention deficit hyperactivity disorder. TAs are ligands for the recently identified trace amine-associated receptor 1 (TAAR1), an important modulator of monoamine neurotransmission. Here, we sought to investigate the consequences of TAAR1 hypersignaling by generating a transgenic mouse line overexpressing Taar1 specifically in neurons. Taar1 transgenic mice did not show overt behavioral abnormalities under baseline conditions, despite augmented extracellular levels of dopamine and noradrenaline in the accumbens nucleus (Acb) and of serotonin in the medial prefrontal cortex. In vitro, this was correlated with an elevated spontaneous firing rate of monoaminergic neurons in the ventral tegmental area, dorsal raphe nucleus, and locus coeruleus as the result of ectopic TAAR1 expression. Furthermore, Taar1 transgenic mice were hyposensitive to the psychostimulant effects of amphetamine, as it produced only a weak locomotor activation and failed to alter catecholamine release in the Acb. Attenuating TAAR1 activity with the selective partial agonist RO5073012 restored the stimulating effects of amphetamine on locomotion. Overall, these data show that Taar1 brain overexpression causes hyposensitivity to amphetamine and alterations of monoaminergic neurotransmission. These observations confirm the modulatory role of TAAR1 on monoamine activity and suggest that in vivo the receptor is either constitutively active and/or tonically activated by ambient levels of endogenous agonist(s).
Article
Full-text available
Dysfunctions of dopaminergic homeostasis leading to either low or high dopamine (DA) levels are causally linked to Parkinson's disease, schizophrenia, and addiction. Major sites of DA synthesis are the mesencephalic neurons originating in the substantia nigra and ventral tegmental area; these structures send major projections to the dorsal striatum (DSt) and nucleus accumbens (NAcc), respectively. DA finely tunes its own synthesis and release by activating DA D2 receptors (D2R). To date, this critical D2R-dependent function was thought to be solely due to activation of D2Rs on dopaminergic neurons (D2 autoreceptors); instead, using site-specific D2R knock-out mice, we uncover that D2 heteroreceptors located on non-DAergic medium spiny neurons participate in the control of DA levels. This D2 heteroreceptor-mediated mechanism is more efficient in the DSt than in NAcc, indicating that D2R signaling differentially regulates mesolimbic- versus nigrostriatal-mediated functions. This study reveals previously unappreciated control of DA signaling, shedding new light on region-specific regulation of DA-mediated effects.
Article
Full-text available
Schizophrenia is a chronic, severe and highly complex mental illness. Current treatments manage the positive symptoms, yet have minimal effects on the negative and cognitive symptoms, two prominent features of the disease with critical impact on the long-term morbidity. In addition, antipsychotic treatments trigger serious side effects that precipitate treatment discontinuation. Here, we show that activation of the trace amine-associated receptor 1 (TAAR1), a modulator of monoaminergic neurotransmission, represents a novel therapeutic option. In rodents, activation of TAAR1 by two novel and pharmacologically distinct compounds, the full agonist RO5256390 and the partial agonist RO5263397, blocks psychostimulant-induced hyperactivity and produces a brain activation pattern reminiscent of the antipsychotic drug olanzapine, suggesting antipsychotic-like properties. TAAR1 agonists do not induce catalepsy or weight gain; RO5263397 even reduced haloperidol-induced catalepsy and prevented olanzapine from increasing body weight and fat accumulation. Finally, TAAR1 activation promotes vigilance in rats and shows pro-cognitive and antidepressant-like properties in rodent and primate models. These data suggest that TAAR1 agonists may provide a novel and differentiated treatment of schizophrenia as compared with current medication standards: TAAR1 agonists may improve not only the positive symptoms but also the negative symptoms and cognitive deficits, without causing adverse effects such as motor impairments or weight gain.Molecular Psychiatry advance online publication, 29 May 2012; doi:10.1038/mp.2012.57.
Book
Since the first implant of a carbon microelectrode in a rat 35 years ago, there have been substantial advances in the sensitivity, selectivity and temporal resolution of electrochemical techniques. Today, these methods provide neurochemical information that is not accessible by other means. The growing recognition of the versatility of electrochemical techniques indicates a need for a greater understanding of the scientific foundation and use of these powerful tools. Electrochemical Methods for Neuroscience provides an updated summary of the current, albeit evolving, state of the art and lays the scientific foundation for incorporating electrochemical techniques into on-going or newly emerging research programs in the neuroscience disciplines. With contributions from pioneers in the field, the text outlines the applications and benefits of a wide range of electrochemical techniques. It explores the methodology behind the acquisition of neurochemical and neurobiological data through continuous amperometry, fast scan cyclic voltammetry, high-speed chronoamperometry, ion-selective microelectrodes, enzyme based microelectrodes, and in vivo voltammetry with telemetry. The text also introduces emerging concepts in the field such as the correlation of electrochemical recordings with information obtained from patch clamp, electrophysiological, and behavioral techniques. By presenting up-to-date information on the growing collection of electrochemical methods, microsensors, and research techniques, Electrochemical Methods for Neuroscience assists seasoned researchers and newcomers to the field in making sound decisions about adopting the most appropriate of these tools for their future research objectives.
Article
Trace amines such as p-tyramine and beta-phenylethylamine are found endogenously as well as in the diet. Concomitant ingestion of these foodstuffs with monoamine oxidase inhibitors may result in the hypertensive crisis known as the "beer, wine, and cheese effect" attributed to their sympathomimetic action. Trace amines have been shown to act on one of a novel group of mammalian seven transmembrane spanning G protein-coupled receptors belonging to the rhodopsin superfamily, cloned in 2001. This receptor encoded by the human TAAR1 gene is also present in rat and mouse genomes (Taar1) and has been shown to be activated by endogenous trace amine ligands, including p-tyramine and beta-phenylethylamine. A number of drugs, most notably amphetamine and its derivatives, act as agonists at this receptor. This review proposes an official nomenclature designating TAAR1 as the trace amine 1 receptor following the convention of naming receptors after the endogenous agonist, abbreviated to TA(1) where necessary. It goes on to discuss briefly the significance of the receptor, agents acting upon it, its distribution, and currently hypothesized physiological and pathophysiological roles. In humans, a further five genes are thought to encode functional receptors (TAAR2, TAAR5, TAAR6, TAAR8, and TAAR9). TAAR3 seems to be a pseudogene in some individuals but not others. TAAR4 is a pseudogene in humans, but occurs with TAAR3 as a functional gene in rodents. Nine further genes are present in rats and mice. The endogenous ligands are not firmly established but some may respond to odorants consistent with their expression in olfactory epithelium
Article
Amphetamines are widely abused drugs that interfere with dopamine transport and storage. Recently however, another mechanism of action was identified: stereoselective activation of the Gαs protein-coupled trace amine-associated receptor 1. To identify structural determinants of this stereoselectivity we functionally evaluated six mutant receptors in vitro then used homology modeling and dynamic simulation to predict drug affinities. Converting Asp102 to Ala rendered mouse and rat TAAR1 insensitive to β-phenylethylamine, AMPH and METH. Mutating Met268 in rTAAR1 to Thr shifted the concentration-response profiles for AMPH and METH isomers rightward an order of magnitude whereas replacing Thr268 with Met in mTAAR1 resulted in profiles leftward shifted 10-30 fold. Replacing Asn287 with Tyr in rTAAR1 produced a mouse-like receptor while the reciprocal mTAAR1 mutant was rTAAR1-like. These results confirm TAAR1 is an AMPH/METH receptor in vitro and establish residues 102 and 268 as major contributors to AMPH/METH binding with 287 determining species stereoselectivity.
Article
Trace Amine-Associated Receptor 1 (TAAR1) is a G protein-coupled receptor that belongs to the family of TAAR receptors and responds to a class of compounds called trace amines, such as β-phenylethylamine (β-PEA) and 3-iodothyronamine (T(1) AM). The receptor is known to have a very rich pharmacology and could be also activated by other classes of compounds, including adrenergic and serotonergic ligands. It is expected, that targeting TAAR1 could provide a novel pharmacological approach to correct monoaminergic dysfunctions found in several brain disorders, such as schizophrenia, depression, attention deficit hyperactivity disorder and Parkinson's disease. Only recently, the first selective TAAR1 agonist RO5166017 has been identified. To explore the molecular mechanisms of protein-agonist interaction and speed up the identification of new chemical entities acting on this biomolecular target, we derived a homology model for the hTAAR1. The putative protein binding site has been explored by comparing the hTAAR1 model with the β2-adrenoreceptor binding site, available by X-ray crystallization studies, and with the homology modeled 5HT(1A) receptor. The obtained results, in tandem with docking studies performed with RO5166017, β-PEA and T(1) AM, provided an opportunity to reasonably identify the hTAAR1 key residues involved in ligand recognition and thus define important starting points to design new agonists. © 2012 John Wiley & Sons A/S.
Article
Background: Trace amines, compounds structurally related to classical biogenic amines, represent endogenous ligands of the trace amine-associated receptor 1 (TAAR1). Because trace amines also influence the activity of other targets, selective ligands are needed for the elucidation of TAAR1 function. Here we report on the identification and characterization of the first selective and potent TAAR1 partial agonist. Methods: The TAAR1 partial agonist RO5203648 was evaluated for its binding affinity and functional activity at rodent and primate TAAR1 receptors stably expressed in HEK293 cells, for its physicochemical and pharmacokinetic properties, for its effects on the firing frequency of monoaminergic neurons ex vivo, and for its properties in vivo with genetic and pharmacological models of central nervous system disorders. Results: RO5203648 showed high affinity and potency at TAAR1, high selectivity versus other targets, and favorable pharmacokinetic properties. In mouse brain slices, RO5203648 increased the firing frequency of dopaminergic and serotonergic neurons in the ventral tegmental area and the dorsal raphe nucleus, respectively. In various behavioral paradigms in rodents and monkeys, RO5203648 demonstrated clear antipsychotic- and antidepressant-like activities as well as potential anxiolytic-like properties. Furthermore, it attenuated drug-taking behavior and was highly effective in promoting attention, cognitive performance, and wakefulness. Conclusions: With the first potent and selective TAAR1 partial agonist, RO5203648, we show that TAAR1 is implicated in a broad range of relevant physiological, behavioral, and cognitive neuropsychiatric dimensions. Collectively, these data uncover important neuromodulatory roles for TAAR1 and suggest that agonists at this receptor might have therapeutic potential in one or more neuropsychiatric domains.