ArticlePDF Available

Abstract

Introduction. The trace amines, endogenous amines closely related to the biogenic amine neurotransmitters, have been known to exert physiological and neurological effects for decades. The recent identification of a trace amine-sensitive G protein-coupled receptor, trace amine-associated receptor 1 (TAAR1), and subsequent development of TAAR1-selective small-molecule ligands, has renewed research into the therapeutic possibilities of trace amine signaling. Areas covered. Recent efforts in elucidating the neuropharmacology of TAAR1, particularly in neuropsychiatric and neurodegenerative disease, addiction, and regulation of arousal state, will be discussed. Focused application of TAAR1 mutants, synthetic TAAR1 ligands and endogenous biomolecules such as 3-iodothyronamine (T1AM) has yielded a basic functional portrait for TAAR1, despite a complex biochemistry and pharmacology. The close functional relationship between TAAR1 and dopaminergic signaling is likely to underlie many of its CNS effects. However, TAAR1’s influences on serotonin and glutamate neurotransmission will also be highlighted. Expert opinion. TAAR1 holds great promise as a therapeutic target for mental illness, addiction, and sleep disorders. A combination of preclinical and translationally-driven studies has solidified TAAR1 as a key node in the regulation dopaminergic signaling. Continued focus on the mechanisms underlying TAAR1’s regulation of serotonin and glutamate signaling, as well as dopamine, will yield further disease-relevant insights.
Full Terms & Conditions of access and use can be found at
http://www.tandfonline.com/action/journalInformation?journalCode=iett20
Expert Opinion on Therapeutic Targets
ISSN: 1472-8222 (Print) 1744-7631 (Online) Journal homepage: http://www.tandfonline.com/loi/iett20
Trace amine-associated receptor 1: a multimodal
therapeutic target for neuropsychiatric diseases
Michael D. Schwartz, Juan J. Canales, Riccardo Zucchi, Stefano Espinoza, Ilya
Sukhanov & Raul R. Gainetdinov
To cite this article: Michael D. Schwartz, Juan J. Canales, Riccardo Zucchi, Stefano Espinoza,
Ilya Sukhanov & Raul R. Gainetdinov (2018) Trace amine-associated receptor 1: a multimodal
therapeutic target for neuropsychiatric diseases, Expert Opinion on Therapeutic Targets, 22:6,
513-526, DOI: 10.1080/14728222.2018.1480723
To link to this article: https://doi.org/10.1080/14728222.2018.1480723
Accepted author version posted online: 25
May 2018.
Published online: 05 Jun 2018.
Submit your article to this journal
Article views: 8
View related articles
View Crossmark data
REVIEW
Trace amine-associated receptor 1: a multimodal therapeutic target for
neuropsychiatric diseases
Michael D. Schwartz
a
, Juan J. Canales
b
, Riccardo Zucchi
c
, Stefano Espinoza
d
, Ilya Sukhanov
e
and Raul R. Gainetdinov
f,g
a
Center for Neuroscience, SRI International, Menlo Park, CA, USA;
b
Division of Psychology, School of Medicine, College of Health and Medicine,
University of Tasmania, Hobart, Australia;
c
Department of Pathology, University of Pisa, Pisa, Italy;
d
Fondazione Istituto Italiano di Tecnologia,
Neuroscience and Brain Technologies Dept., Genoa, Italy;
e
Institute of Pharmacology, Pavlov Medical University, St. Petersburg, Russia;
f
Institute of
Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia;
g
Center for Translational Biomedicine, Skolkovo Institute of
Science and Technology, Moscow, Russia
ABSTRACT
Introduction: The trace amines, endogenous amines closely related to the biogenic amine neurotrans-
mitters, have been known to exert physiological and neurological effects for decades. The recent
identification of a trace amine-sensitive G protein-coupled receptor, trace amine-associated receptor
1 (TAAR1), and subsequent development of TAAR1-selective small-molecule ligands, has renewed
research into the therapeutic possibilities of trace amine signaling.
Areas covered: Recent efforts in elucidating the neuropharmacology of TAAR1, particularly in neurop-
sychiatric and neurodegenerative disease, addiction, and regulation of arousal state, will be discussed.
Focused application of TAAR1 mutants, synthetic TAAR1 ligands, and endogenous biomolecules such as
3-iodothyronamine (T1AM) has yielded a basic functional portrait for TAAR1, despite a complex
biochemistry and pharmacology. The close functional relationship between TAAR1 and dopaminergic
signaling is likely to underlie many of its CNS effects. However, TAAR1s influences on serotonin and
glutamate neurotransmission will also be highlighted.
Expert opinion: TAAR1 holds great promise as a therapeutic target for mental illness, addiction, and
sleep disorders. A combination of preclinical and translationally driven studies has solidified TAAR1 as a
key node in the regulation of dopaminergic signaling. Continued focus on the mechanisms underlying
TAAR1s regulation of serotonin and glutamate signaling, as well as dopamine, will yield further disease-
relevant insights.
ARTICLE HISTORY
Received 9 February 2018
Accepted 21 May 2018
KEYWORDS
Dopamine; serotonin;
addiction; schizophrenia;
depression; sleep;
psychostimulants;
neuropharmacology
1. Introduction
1.1. Trace amines and trace amine-associated receptor 1
(TAAR1)
The trace amines, endogenous amines closely related to the bio-
genic amine neurotransmitters (e.g. dopamine (DA), serotonin (5-
hydroxytryptamine; 5-HT) and norepinephrine (NE)), have been
known to exert physiological and neurological effects since the
early twentieth century [1]. However, the lack of an identifiable
endogenous receptor for these molecules, coupled with their
markedly low in vivo concentrations, led in part to the idea that
trace amines were false neurotransmitters[2]. In 2001, this con-
ventional wisdom was challenged with the identification of a
vertebrate G-protein coupled receptor (GPCR) that preferentially
responded to trace amines [3,4]. This receptor, trace amine-asso-
ciated receptor 1 (TAAR1), is part of a large and evolutionarily
diverse family of TAARs with six functional members in humans
[5]. Several TAARs act as olfactory receptors [6]. In the mammalian
brain, the finding that TAAR1 powerfully modulates monoaminer-
gic neurotransmission [79] has rejuvenated research efforts into
the function and therapeutic implications of TAAR1 and its ligands.
1.2. TAAR1 expression and function
In the brain, TAAR1 expression is enriched throughout the
limbic and aminergic systems, encompassing the dopaminer-
gic ventral tegmental area (VTA)/substantia nigra and seroto-
nergic dorsal raphe nucleus (DRN) [1,10,11], and is therefore
ideally positioned to regulate the activity of these neurotrans-
mitter systems. Indeed, transgenic mice lacking TAAR1 exhibit
markedly elevated discharge rates of DA and 5-HT neurons
[12], suggesting that TAAR1 activation down-regulates mono-
aminergic neurotransmission. The strategic neuroanatomical
location of TAAR1 and its remarkable ability to regulate ami-
nergic neurotransmission suggest that this receptor could
serve as a target to develop more effective, new-generation
pharmacotherapies for neuropsychiatric diseases, addiction,
and sleep disorders.
This review will highlight recent efforts in elucidating the
neurological and neurophysiological effects and potential
therapeutic utility of TAAR1 activation via recently developed
TAAR1-specific small molecules, as well as endogenous bio-
molecules such as 3-iodothyronamine. While beyond the
scope of this review, there is also significant peripheral
CONTACT Raul R. Gainetdinov gainetdinov.raul@gmail.com Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya
embarkment 7-9, 199034 St. Petersburg, Russia
EXPERT OPINION ON THERAPEUTIC TARGETS
2018, VOL. 22, NO. 6, 513526
https://doi.org/10.1080/14728222.2018.1480723
© 2018 Informa UK Limited, trading as Taylor & Francis Group
TAAR1 expression in pancreas, stomach, and leukocytes, sug-
gesting potential for TAAR1-based drugs in diabetes, obesity,
and possibly immune disorders [2].
1.3. Synthetic TAAR1 agonists and antagonists
Hoffmann-La Roche investigators performed a large-scale effort
to derivatize adrenergic ligands, which were screened for TAAR1
activation by cAMP assays in heterologous cells expressing
TAAR1, and for specificity via radioligand binding experiments
involving over a hundred different proteins. This effort yielded
several full (e.g. RO5166017 and RO5256390) and partial (e.g.
RO5203648 and RO5263397) TAAR1 agonists [11,13,14] that to
date have been successfully used in experimental models of
neurological diseases such as drug addiction, schizophrenia,
and Parkinsons disease. In general, the RO compoundsare
over 100-fold selective for TAAR1 versus other aminergic recep-
tors, but the Kis for some other receptors namely α
2
adrener-
gic, 5-HT
2
5-HTergic, µ and κopioid, and I
1
imidazoline receptors
are in the nanomolar range, and so additional effects on
different targets cannot be excluded in all cases.
Pharmacological research has also aimed at developing
selective TAAR1 antagonists. Screening of about 700,000
Roche compounds led to the identification of N-(3-Ethoxy-
phenyl)-4-pyrrolidin-1-yl-3-trifluoromethyl-benzamide (EPPTB)
[10,15]. This benzamide derivative had high selectivity and
affinity for mouse TAAR1 (Ki = 0.9 nM), although the affinity
for human TAAR1 was in the micromolar range. In particular,
EPPTB was critical in revealing the constitutive background
activity of the TAAR1 system because it caused a significant
increase in the firing rate of mouse VTA DA neurons [10].
2. 3-iodothyronamine: an endogenous TAAR1 ligand
2.1. Biochemistry and pharmacology of T1AM
3-iodothyronamine (T1AM) is an endogenous compound
whose chemical structure is related to thyroid hormones
[16]. The differences consist in the absence of the carboxyl
group and of all iodine atoms except one. It is thought to be
synthesized from 3,5,3ʹ-triiodothyronine (T3) through the
sequential action of deiodinases and amino acid decarboxy-
lases (possibly ornithine decarboxylase) [17], but the precise
biosynthetic pathway and the physiological site(s) of produc-
tion are still unclear. T1AM has been identified in rodent and
human blood, and in most rodent organs including the brain,
where its average endogenous level is on the order of a few
pmoles per g [16,1823].
In 2004, T1AM was reported to be a powerful activator of
TAAR1 [16]. Its EC
50
for a biochemical response (cAMP produc-
tion) in human neoplastic cell lines expressing rat or mouse
TAAR1 averaged 14 nM and 112 nM, respectively, and so it
was lower than observed for endogenous trace amines,
namely tyramine and β-phenylethylamine. With human
TAAR1, the affinity for T1AM was in the micromolar range,
but it was still higher than observed for tyramine and β-
phenylethylamine [24]. So, T1AM qualifies as a bone fide phy-
siological TAAR1 agonist.
2.2. Neurophysiological effects of T1AM
T1AM modulates several integrative functions, particularly
feeding behavior, sleep, and cognition (reviewed by [25]). In
fed animals, i.c.v. T1AM administration increased food intake at
dosages as low as 1.2 nmol/Kg, and a similar effect was
observed after arcuate nucleus injection [26]. However, in fast-
ing animals, the response was biphasic: dosages in the low
nanomolar range were anorexic, while higher dosages
(51 nmol/Kg) had the opposite effect [27]. T1AM injection
(3 µg) in the preoptic region enhanced locomotor activity and
increased wakefulness while decreasing non-rapid eye move-
ment (NREM) sleep time [28]. i.c.v. T1AM injection (1.32 4 µg/
Kg) elicited prolearning and antiamnestic effects in the passive
avoidance paradigm as well as increased curiosity in the novel
object recognition task [21].
2.3. Mechanisms of action
2.3.1. Neuromodulatory actions of T1AM
The basic cellular processes targeted by T1AM remain to be
determined but are proposed to be neuromodulatory in nature.
Manni et al. [21] reported that active dosages of T1AM increased
average brain T1AM concentration by about 30-fold, consistent
with a physiological role of endogenous T1AM. T1AM applied
locally in rat locus coeruleus modulated the activity of adrenergic
neurons with EC
50
= 2.7 µM [29]. T1AMs pro-learning and anti-
amnestic effects may depend on histaminergic activity because
they were dampened or abolished by histamine receptor
antagonists and in transgenic mice lacking histidine decarbox-
ylase, the key enzyme regulating histamine biosynthesis [21,22].
Modulation of adrenergic and histaminergic activity could also
underlie wake promotion because both neurotransmitters are
robustly wake promoting [3032].
2.3.2. T1AM and TAAR1
Preliminary evidence for a TAAR1-mediated effect has been
recently reported by electrophysiological recordings performed
in rat entorhinal cortex. In this model, T1AM rescued long-term
Article Highlights
TAAR1 regulates DA, 5-HT, and glutamate neurotransmission by
decreasing basal firing rates and negatively modulating receptor
sensitivity.
Selective full and partial TAAR1 agonists exhibit potent antipsychotic,
antidepressant, anti-impulsive and procognitive effects.
TAAR1 agonism reduces stimulant-induced reward, stimulant self-
administration, and relapse to drug seeking.
T1AM, an endogenous TAAR1 agonist derived from thyroid hormone,
modulates food intake, increases wakefulness and improves cognitive
performance.
TAAR1 partial, but not full agonists, promote wakefulness, while both
full and partial agonists suppress cataplexy.
Based on preclinical studies, TAAR1 agonism represents a novel
strategy for treating neuropsychiatric diseases involving dysregulated
monoaminergic signaling such as schizophrenia, addiction, depres-
sion, ADHD, Parkinsons disease, and OCD.
This box summarizes key points contained in the article.
514 M. D. SCHWARTZ ET AL.
potentiation in the presence of toxic concentrations of beta
amyloid, and the effect was abolished by the TAAR1 antagonist
EPPTB [33]. This exciting observation suggests additional ther-
apeutic utility for TAAR1 agonists because of the putative role
of beta amyloid in Alzheimers disease and other forms of
cognitive impairment.
However, it is presently unclear whether all the actions
produced by T1AM are mediated by TAAR1. Other potential
targets include 5HT1b receptors [Braunig 2018 [34] Front Ph],
other TAARs (particularly TAAR5), α
2A
adrenergic receptors,
transient receptor potential channels (particularly TRPM8),
and monoamine transporters [25,34]. The presence of multiple
physiological targets is not unusual for a chemical messenger,
but it casts doubts on a TAAR1-specific mechanism for T1AM.
Conversely, some effects observed after T1AM administration
could actually be mediated by 3-iodothyroacetic acid, the
product of T1AM oxidative deamination. For example, hista-
mine and 3-iodothyroacetic acid are involved in the acceler-
ated response to the hot plate test, suggesting reduced pain
threshold [20,22]. Further investigation with TAAR1 antago-
nists and/or KO mice is necessary to resolve this crucial issue.
2.4. Synthetic T1AM analogs
To address these questions, synthetic T1AM analogs have
been developed by modifying the thyronamine scaffold. The
first two series of analogs consisted of phenyltyramine deriva-
tives [35,36], while more recently halogen-free biaryl-methane
thyronamine analogs (the so-called SG compounds) have
been synthesized [37,38]. Some of these compounds were
equipotent or even more potent than T1AM, as measured by
cAMP induction. They also reproduced the in vivo effects of
T1AM on glucose homeostasis and cognitive function in
mouse. However, their selectivity has not yet been specifically
evaluated, so the same questions raised for T1AM regarding
interaction with different targets apply here. Systematic eva-
luation of T1AM analogs is expected to clarify several features
of T1AM/TAAR1 interactions and structure-activity relation-
ships [39] and provides a valuable background for further
research in the TAAR1 pharmacology.
3. TAAR1 in neuropsychiatric disorders
3.1. Trace amines, DA, and neuropsychiatric disease
Trace amine dysregulation has long been associated with several
psychiatric and neurological diseases. For example, elevated PEA
content has been documented in schizophrenia [4042],
whereas decreased PEA levels were associated with depression
[4345]. However, the lack of an identifiable endogenous recep-
tor and dearth of suitable investigative tools limited advance-
ment on this front for some time [2]. The identification of TAAR1
[3,4] and subsequent demonstration that TAAR1 modulates DA
and 5-HT neurotransmission [79] has renewed interest in this
association. Brain DA is critically involved in the etiology and
pathogenesis of neuropsychiatric disorders including schizo-
phrenia, Attention Deficit and Hyperactivity Disorder (ADHD),
and Parkinsons disease; DA dysregulation is proposed to con-
tribute to Obsessive-Compulsive and Related Disorders (OCD),
bipolar disorder, major depression, and dyskinesias [46]. Most of
these conditions have been previously linked to dysregulated
endogenous trace amines [1,47]. With the development of
TAAR1-specific mutant mice [4850] and selective pharmacolo-
gical compounds [10,11,1315], these advances have vastly
enhanced understanding of TAAR1s role in neuropsychiatric
disorders and their potential therapeutic applications.
3.2. TAAR1 and schizophrenia
3.2.1. Dopaminergic dysregulation and schizophrenia
The DA theory of schizophrenia asserts that increased dopa-
minergic tone or D
2
receptor sensitivity resulting in dysregu-
lated DA signaling underlies the development of
schizophrenia, particularly its positive symptoms (e.g. halluci-
nations, delusions, and disordered thoughts and speech). This
theory has a strict predictive validity because all known clini-
cally effective antipsychotics are D
2
receptor antagonists [51].
Hyperactivity induced by dopaminergic psychostimulants is
considered a behavioral manifestation of increased dopami-
nergic activity in the mesolimbic pathway [52], and poten-
tiated psychostimulant-induced activity is used as an animal
correlate of positive symptoms [53]. Accordingly, the ability of
a drug to antagonize this hyperactivity has been used for
decades as a preclinical screening tool to identify novel anti-
psychotics [54,55].
3.2.2. TAAR1 and dopaminergic tone
Taar1 KO mice do not differ in size, weight, and temperature
from wild type (WT) littermates and perform normally in beha-
vioral tests including motor coordination, visual acuity, grip
test, nociception, and locomotor activity in the open field
[49,50,56]. However, these mice exhibit increased locomotor
responses to dopaminergic psychostimulants such as amphe-
tamine, methamphetamine, or MDMA compared to WTs
[48,50,57,58] as well as high doses of the selective D
2
/D
3
agonist
quinpirole [59]. Taar1 KO mice were also more sensitive to
locomotor sensitization induced by repeated amphetamine
and methamphetamine administration [57,60]. TAAR1 agonists
prevented cocaine- and amphetamine-induced hyperlocomo-
tion in WT mice [11,14,61] and Wistar rats [13] and enhanced
olanzapine (atypical antipsychotic)-induced inhibition of loco-
motor activity following cocaine [14]. Conversely, ampheta-
mine-induced hyperactivity was strongly attenuated in
TAAR1-overexpressing (OE) mice and rescuedby the selective
partial agonist RO5073012 [61]. Together, these findings indi-
cate TAAR1 as a viable locus for treatment of positive psychotic
symptoms, likely mediated by modulation of dopaminergic
signaling. Consistent with this hypothesis, TAAR1 agonists
block hyperlocomotion in hyperdopaminergic DA transporter
knockout (DAT) KO mice and rats [11,13,62].
3.2.3. TAAR1 and NMDA hypofunction
N-methyl-D-aspartate (NMDA) receptor antagonists also
increase motor activation [63,64], and this hyperactivity is simi-
larly reversed by antipsychotics [65]. As with dopaminergic
psychostimulants, TAAR1 agonists reversed hyperlocomotion
induced by the NMDA antagonists L-687,414 and phencyclidine
[11,14]. The partial TAAR1 agonist RO5203648 also blocked
EXPERT OPINION ON THERAPEUTIC TARGETS 515
hyperlocomotion in NMDA receptor-deficient mice [13].
However, L-687,414-induced hyperactivity was not further
potentiated in Taar1 KO mice [11], in contrast to the exagger-
ated response to dopaminergic stimulants in these animals.
Future studies should examine the respective contributions of
dopaminergic versus glutamatergic pathways to the expression
of hyperactivity vis a vis TAAR1.
3.2.4. Caveats and questions
Some variability is reported in the direct locomotor effects of
TAAR1 agonists themselves. Some studies reported that
TAAR1 agonists decreased locomotor activity in intact animals
[13,14], while others did not find any effects of these drugs
when administered alone [61,6668]. Intriguingly, TAAR1 dele-
tion greatly attenuated climbing and other stereotypy beha-
viors induced by high doses of the mixed D
1
/D
2
agonist
apomorphine [69], likely due to a direct agonistic action of
apomorphine at TAAR1 [4]. Because apomorphine-induced
stereotypies are also used to screen for antipsychotics [70
72], these findings imply a need for care when interpreting
apomorphine-induced behaviors in rodents.
Whereas all clinically used antipsychotics augment haloper-
idol-induced catalepsy, partial TAAR1 agonists actually reduce
this phenomenon [13,14] and do not themselves induce cata-
lepsy [14]. Intriguingly, the catalepsy induced by haloperidol
was significantly reduced in Taar1 KO mice [8].
3.2.5. Cognitive symptoms of schizophrenia
Memory, attention, and other cognitive deficits form another
important aspect of the symptomatology of schizophrenia
[73]. Taar1 KO mice showed impairment in prepulse inhibition
of the acoustic startle response [50], indicating a sensorimotor
gating deficit. This abnormality is commonly used to demon-
strate schizophrenia-like phenotypes in animals because
human schizophrenic patients exhibit an analogous deficit
[74]. By contrast, spatial working memory in the forced alter-
nation test was intact in Taar1 KO mice [50]. The TAAR1
agonists RO5256390, RO5203648, and RO5263397 increased
accuracy in the object retrieval task in cynomolgus macaques
[13,14], and RO5256390 (1.0 and 3.0 mg/kg) fully reversed
executive function deficits induced by repeated PCP treatment
(5.0 mg/kg) in the attention set shift task in rats [14]. Further
assessment of procognitive effects of TAAR1 agonists is there-
fore warranted, particularly in key cognitive domains such as
attention and learning.
In general, the loss of TAAR1 induces elevated DA neuro-
transmission in the mesolimbic pathway in mice. Specific
TAAR1 agonists ameliorate pharmacologically and genetically
induced hyperlocomotion without the undesirable motor side
effects characteristic of D
2
receptor-blocking antipsychotics.
Moreover, TAAR1 agonists may be effective in treating cogni-
tive deficits associated with schizophrenia.
3.3. TAAR1 and Parkinsons disease
Loss of nigrostriatal dopaminergic neurotransmission is the
key point in pathogenesis of Parkinsons disease. L-DOPA,
the chemical precursor of DA, is the first and the most widely
used treatment of Parkinsons disease. Taar1-KO mice in which
dopaminergic neurons were unilaterally lesioned with the
neurotoxin 6-OHDA had increased sensitivity to L-DOPA-
induced rotational behavior and dyskinesia compared to WT
littermates [75]. Thus, it would be important to determine
whether partial TAAR1 agonists ameliorate L-DOPA-associated
side-effects such as dyskinesia. Intriguingly, Sotnikova and
colleagues observed antiparkinsonianeffects of ampheta-
mines and MDMA in DA-depleted DAT KO mice [76], but the
same effect persisted in double knockout mice lacking both
DAT and TAAR1, indicating that this potential antiparkinso-
nianaction of amphetamines is TAAR1-independent [58].
3.4. TAAR1 and ADHD
Evidence increasingly suggests that DAT dysfunction is
involved in the pathogenesis of ADHD [7779]; indeed, DAT
KO mice are a known genetic animal model of ADHD [80]. In a
novel environment, these mice exhibit profound hyperactivity
[81,82]. TAAR1 agonists blocked hyperlocomotion in these
mice [11], mimicking the calming effect of amphetamine and
methylphenidate [83], the drugs clinically used to treat ADHD
patients. Similar effects of the partial TAAR1 agonist
RO5203648 were observed in DAT KO rats [62]. Conversely,
double DAT/Taar1 KO mice demonstrated further increased
hyperactivity over DAT KO mice [13], providing additional
support for TAAR1s role in dopaminergic control.
ADHD patients are also characterized by increased
impulsivity. A recent study has shown that lack of TAAR1
led to perseverative and impulsive behaviors that corre-
lated with deficient prefrontal cortical glutamatergic trans-
mission [84]. Furthermore, RO5166017 and RO5203648
decreased premature responding in a fixed interval condi-
tioningscheduleinWTmice[84]. RO5256390 and
RO5263397 also increased the number of reinforcers
earned in a differential reinforcement of low response
rates test in cynomolgus macaques [13]. RO5263397
reduced hyperimpulsivity (5CSRTT) in methamphetamine-
treated rats but did not affect the number of premature
responses in 5CSRTT and choice of large reward in the
delay discounting task in vehicle-treated rats [85].
Similarly, RO5203648 failed to change impulsive behavior
in DAT-null and WT rats in the Intolerance-to-Delay Task
[86]. Thus, some studies to date have supported a role for
TAAR1 in impulsive and compulsive behavior, while others
have not; future studies should try to tease apart the
contributions of dopaminergic versus glutamatergic sys-
tems to these behaviors, as TAAR1sinfluencecouldbe
more weightedtoward one or the other.
3.5. TAAR1 and OCD
Although selective 5-HT reuptake inhibitors (SSRI) form the
first line of drugs to treat OCD [87], a role for DA function in
OCD pathogenesis has been actively studied in recent years
(for review, see [88]). Antipsychotics form a second line of OCD
treatment because about 50% OCD patients are resistant to
SSRI therapy. RO5263397 reduced obsessive drinking in sche-
dule-induced polydipsia, a popular preclinical model of com-
pulsive behavior [89]. Additionally, the full TAAR1 agonist
516 M. D. SCHWARTZ ET AL.
RO5256390 blocked compulsive eating in rats [90]. Thus, stu-
dies on TAAR1s involvement in animal models of compulsivity
and OCD, while promising, require further exploration.
3.6. TAAR1 and affective disorders
RO5263397 and RO5203648, but not RO5256390, demon-
strated antidepressant action in the forced swim test in rats
[13,14,90], a test that has high predictive validity for identifica-
tion of new antidepressants [91]. Similarly, RO5203648 showed
anxiolytic effects in the stress-induced hyperthermia test in
mice [13,14,90]. These effects could be mediated via TAAR1-
dependent enhancement of serotonergic signaling because
the antidepressant effect was only seen with partial agonists,
which increase 5-HT firing [11,13,14]. However, several lines of
evidence support dysregulated DA receptor and/or transpor-
ter homeostasis in affective disorders [92]. Consistent with this
hypothesis, RO5263397, RO5203648, and RO5256390 were
effective in another test reflective of antidepressant action in
Cynomolgus monkeys, the differential reinforcement of low-
rate behavior paradigm [13,14,90]. Further studies aimed at
the neurochemical basis for the antidepressive effects of
TAAR1 agonism are therefore warranted.
4. TAAR1 and addiction therapeutics
4.1. Therapeutic challenges in psychostimulant
addiction
Drug addiction is a multifaceted neuropsychiatric disorder
with widespread medical and societal implications.
Improving the treatment, support, and rehabilitation of those
affected by drug addiction continues to be an important
research agenda. Although behavioral and cognitive therapy,
combined with psychosocial support and community inter-
ventions, constitute irreplaceable initiatives to aid in recovery,
considerable agreement exists among psychiatrists and health
professionals specializing in addiction that novel pharmacolo-
gical approaches are needed to treat the disorder more effec-
tively [93,94]. Chiefly, new medications are required to better
manage withdrawal symptoms and craving in the early days
and weeks after drug discontinuation. Such medications could
facilitate compliance and engagement in behavioral therapy,
multiplying the beneficial effects of non-pharmacological
approaches. In this context, addiction to stimulant drugs,
such as cocaine and methamphetamine, is particularly proble-
matic due to the lack of effective treatment options.
4.2. The biogenic amines and addiction
Research into the neurobiological mechanisms that contribute
to drug addiction suggests that the classical biogenic amines,
including DA, norepinephrine and 5-HT, and their correspond-
ing receptor targets, play a critical role [95]. The DA substitu-
tion approach in stimulant addiction involves the use of a
competing dopaminergic agonist to potentially suppress with-
drawal and drug craving in abstinent individuals [96].
Although this is still an avenue under investigation, com-
pounds that act directly at the DA transporter (e.g. slow-acting
transporter blockers), or at DA receptors, are themselves more
likely to have abuse potential and long-term side effects. This
liability justifies the search for new receptor targets to indir-
ectly modulate DA transmission through the ups and downs
of the addiction cycle. Because of its unique association with
ascending dopaminergic projections and key associated limbic
circuits, TAAR1 has emerged as one of the most promising
targets for the treatment of neuropsychiatric disorders, espe-
cially addiction.
4.3. TAAR1 and stabilization of dysregulated DA
signaling
The development of synthetic TAAR1 ligands has proven cri-
tical in elucidating its physiological and behavioral functions.
While both full and partial TAAR1 agonists decrease stimulant-
induced DA overflow in the nucleus accumbens (NAcb)
[7,97,98], their effects on DA neuron firing rate can be differ-
ent. In patch clamp preparations, the full agonist RO5256390
attenuated neuronal firing in the VTA [11], whereas the partial
agonist RO5263397 augmented the firing frequency as did the
antagonist EPPTB [10]. This suggests that TAAR1 is constitu-
tively active and/or tonically activated by endogenous ligands
at the level of midbrain such that partial agonism results in
antagonistic-like effects. Consequently, the use of a partial
agonist may be more advantageous in situations where neu-
rochemical imbalance (e.g. induced by drug exposure) leads to
insufficient or excessive TAAR1 stimulation, providing a means
to stabilizeTAAR1 activity. In addition, through induction of
pacemaker activation, partial agonism may contribute to nor-
malizingDA neuron cell discharge, which is known to be
dysregulated following chronic cocaine exposure [99,100]. In
agreement with these physiological observations, and sup-
porting the notion that TAAR1 activation may indeed dampen
DA transmission under certain conditions, Taar1 KO mice
exhibited increased sensitivity to amphetamine and increased
striatal DA release [49], whereas brain-specific TAAR1 over-
expression reduced the psychomotor stimulant effects of
amphetamine [61].
The development of TAAR1-selective agonists has since
allowed the accumulation of compelling evidence in support
of TAAR1 as a candidate for the design of addiction medica-
tions. Motor sensitization, a process that evolves following
repeated psychomotor stimulant treatment and that involves
plasticity changes in the mesolimbic DA system, was attenu-
ated by the partial TAAR1 agonists RO5203648 [101] and
RO5263397 [102,103]. Self-administration models are the
gold standard in addiction research, allowing the study of a
variety of behavioral processes. TAAR1 activation with the
partial agonist, RO5203648, dose-dependently decreased
cocaine self-administration [13], with similar reductions
being observed in methamphetamine self-administration
[101,102]. Importantly, RO5203648 was able to block stimu-
lant self-administration without concomitant decreases in
response rates for food self-administration, thus ruling out
motor or motivational deficits. Subsequent work demon-
strated that both RO5203648 and the full TAAR1 agonist,
RO5256390, flattened the dose-response curve for cocaine
EXPERT OPINION ON THERAPEUTIC TARGETS 517
self-administration, indicating that TAAR1 activation effec-
tively decreased the reinforcing effects of cocaine [104].
4.4. TAAR1 regulates reward mechanisms
In addition to perturbing motor behavior and promoting rein-
forcement learning, stimulants are known to increase brain
reward and recruit motivational mechanisms to instigate
their procurement. TAAR1 activation regulates reward and
motivational processes induced by stimulant drugs. Using an
intracranial self-stimulation paradigm, Pei et al. (2015) showed
that both RO5263397 and RO5256390 lowered cocaine self-
stimulation thresholds, thus suggesting reduced cocaine
reward. Moreover, in a progressive ratio schedule of reinforce-
ment, RO5203648 dose-dependently shifted both cocaines
and methamphetamines response rate curve rightward and
delayed the time to reach break point while elevating the
break point for food self-administration [97,98]. These data
clearly indicate that TAAR1 activation reduces stimulant
reward and the motivation to seek and self-administer stimu-
lant drugs.
4.5. Preventing drug relapse
There is a myriad of catalysts that can trigger drug relapse, one of
the most insidious problems associated with drug addiction.
Models of relapse have been employed in the laboratory to
investigate the potential of TAAR1 agonists to regulate relapse
to drug-seeking behavior. Data have been similarly convincing in
that both RO5203648 and RO5263397 dose-dependently pre-
vented context-induced cocaine relapse in a model of forced
abstinence [97] and cue- and cocaine prime-induced reinstate-
ment of cocaine and methamphetamine seeking after extinction
training [97,98,103]. Recent local microinfusion studies suggest
that TAAR1 acts in the VTA, prelimbic cortex and especially NAc
to attenuate drug-seeking and reinstatement-related behaviors
[105,106]. These observations support the notion that TAAR1
agonists may be useful in relapse prevention and management
of rehabilitation processes in addiction.
4.6. TAAR1 and the molecular mechanisms of addiction
The molecular mechanisms and signaling pathways through
which TAAR1 exerts such remarkable effects on stimulant-
induced behaviors are still poorly understood. TAAR1 distribu-
tion is predominantly intracellular [4,107], stimulating both
accumulation of cAMP, via Gα
s
-adenylyl cyclase activation
that promotes PKA and PKC phosphorylation [3,4,108], and a
G protein-independent, β-arrestin2-dependent pathway invol-
ving a DA D
2
receptor-regulated protein kinase B (AKT)/glyco-
gen synthase kinase (GSK-3) [109]. To uncover the
mechanisms through which TAAR1 prevents cocaine effects
on DA transmission, Asif-Malik et al. (2017) recently conducted
in vitro fast-scan cyclic voltammetry experiments, elegantly
demonstrating a new pathway to control cocaines neuro-
chemical actions that involves TAAR1 (Figure 1). Upon TAAR1
stimulation, such pathway recruits D
2
autoreceptors function-
ally linked to TAAR1 and downstream molecular targets con-
verging on GSK-3, but not on PKA or PKC [7]. It is worth noting
that GSK-3 has been previously implicated in cocaine sensiti-
zation [110] and cocaine reward memory [111]. These results
open new avenues to further explore such complex molecular
interactions, with a view to optimize TAAR1-based drug devel-
opment in the area of addiction treatment.
4.7. Summary
Although changes in DA transmission are undoubtedly impor-
tant, it is now recognized that the spiral of cycles of absti-
nence and relapse that characterizes stimulant addiction is
associated with widespread metabolic changes in the brain
and alterations in the way that different brain regions connect,
communicate, and function. These connectivity problems,
especially loss of prefrontal-to-striatal functional connectivity,
Figure 1. Intracellular mechanisms though which TAAR1 controls cocaine-induced neurochemical effects on dopamine (DA) transmission. Cocaine blocks the DAT and as
a result increases DA transmission at the synapse (1). The DA D
2
receptor and TAAR1 can form heterodimeric complex that signals through alternative intracellular
messenger systems. This complex potentiates DA D
2
receptor-mediated pre-synaptic DA autoinhibition. TAAR1 is sensitive to shifts in DA concentrations and
promotes DA homeostasis and therefore acts as an intracellular sentinel (2). The DA D
2
receptor agonist, sumanirole, causes a similar inhibition of cocaine-induced
changes in DA uptake (3) whereas the DA D
2
receptor antagonist, L-741,626, enhances cocaine-induced effects on DA clearance (4). DA D
2
receptor activation
activates the GSK-3 β-arrestin2-dependent pathway (5) but activation of the TAAR1/DA D
2
receptor complex inhibits GSK-3 through the same pathway (6). Inhibition
of GSK-3 with the inhibitor, SB631736, activates AKT, which is bound to the D
2
/DAT complex, thereby enhancing DA reuptake (7). Adapted from [7].
518 M. D. SCHWARTZ ET AL.
have been linked to impaired top-downcontrol and impul-
sivity trait, which predict drug escalation and increased relapse
to drug abuse. Recent data showed that Taar1 KO mice exhib-
ited impulsive behavior and dysregulated function in the pre-
frontal cortex, whereas pharmacological activation of TAAR1
with selective agonists reduced premature impulsive
responses [84]. In agreement with these findings, RO5263397
attenuated methamphetamine-induced impulsive behavior
[85]. This evidence suggests that TAAR1 also exerts control
over addiction-related circuits and behaviors that extend
beyond the DA system and its associated functions.
In conclusion, the evidence reviewed in this section sug-
gests that TAAR1 is uniquely placed to exert a decisive influ-
ence over key neurochemical processes and behaviors
associated with drug effects and addiction. Indeed, both neu-
rochemical and behavioral observations demonstrate the abil-
ity of TAAR1 to regulate not only the effects of cocaine and
methamphetamine on DA transmission but also a wide range
of behavioral, motivational, and cognitive processes that are
affected by chronic drug exposure. As noted, the effects of
TAAR1 activation on drug self-administration, drug reward,
and relapse are particularly striking. Taken together, these
findings support the candidacy of TAAR1 as one of the most
promising therapeutic targets in addiction.
5. TAAR1 and wakefulness
5.1. TAAR1, the monoamines, and sleep-wake regulation
Sleep disturbances exert significant costs in terms of personal
health consequences and economic productivity [112,113].
Sleep and circadian dysregulation are common comorbidities
in neuropsychiatric and neurodegenerative disorders [114
116] as well as addiction [117,118]. This link is not surprising
because the monoaminergic and glutamatergic systems
whose dysregulation underlies these diseases also play funda-
mental roles in regulating sleep and wakefulness (for detailed
reviews see [3032,119]). Investigations of TAAR1s involve-
ment in arousal state control suggest an important role in
regulating basal sleep and wakefulness as well as potential
therapeutic value for the sleep disorder narcolepsy.
5.2. TAAR1 mutants and sleep
5.2.1. Basal sleep-wake regulation in TAAR1 mutants
To determine the role of endogenous TAAR1 in regulating
sleep and wakefulness, Taar1 KO and OE mice were instru-
mented for EEG/EMG recording and compared to a common
pool of WT littermates under standard 12h light:dark (LD)
cycles [120]. Circadian organization of locomotor activity,
core body temperature, sleep and waking was normal in
both mutant strains, with wakefulness concentrated in the
dark phase. Total wake time was increased in OE mice relative
to WTs over 24h and was associated with an increased number
of wake bouts; by contrast, KO mice exhibited decreased
wakefulness and increased NREM sleep at the lights-on transi-
tion compared to OEs and WTs. Compensatory recovery sleep
following a 6h sleep deprivation was normal in both mutants,
indicating that homeostatic sleep regulation was unaffected
by TAAR1 mutation. Thus, constitutive TAAR1 overexpression
and deletion elicit a mild but significant increase and decrease
in basal wakefulness, respectively. These opposing effects are
somewhat surprising because both knockout and overexpres-
sion are associated with elevated VTA DA and DRN 5-HT firing
rates in vitro [11,61], and both DA [121123] and 5-HT
[124,125] activities are positively correlated with wakefulness.
However, TAAR1 overexpression also elevates firing in LC
noradrenergic neurons and VTA GABAergic neurons [61],
both of which are associated with wake promotion [126,127],
which may explain the enhanced wakefulness seen in OE, but
not KO mice. Conditional deletion/expression of TAAR1 in vivo
would be of considerable help in identifying how TAAR1
influences basal arousal states.
By contrast, TAAR1 deletion was associated with impulsivity
and increased nocturnal nose-poke activity in a goal-directed
task [84], suggesting a greater perturbation of rest-activity
cycles than seen in the sleep EEG studies. Such a phenotype
could arise from an interaction between the normal diurnal
cycling of extracellular DA, which peaks at lights-off [128], and
dysregulated DA signaling in Taar1 KO mice [9]. This hypothesis
could explain both the temporal bias of the phenotype and its
emergence in a reward-associated context (compared to a
more neutral homecage environment) but has yet to be tested.
5.2.2. EEG spectral abnormalities
TAAR1 mutation elicited marked alterations in EEG spectral
composition; specifically, theta (48 Hz) and gamma (> 30Hz)
band activity was elevated in KO compared to OE mice in both
sleep and wakefulness, with WT mice intermediate between
them. Such a phenotype could arise from serotonergic dysre-
gulation in KO mice [11], although 5-HT suppresses gamma
and theta activity [129,130], rather than enhancing it as seen
in Taar1 KOs. Alternatively, dysregulated arousal states and
EEG spectra could result from abnormal glutamatergic regula-
tion [84]. Enhancing glutamatergic transmission via group II
metabotropic glutamate receptors [131], particularly mGluR2
[132], as well as group I mGluR5 receptors [133135] promotes
waking and high-frequency EEG activity (i.e. gamma power),
while inhibition tends to potentiate NREM sleep and EEG slow
wave activity (i.e. delta power, 0.54Hz).
5.2.3. TAAR1 deletion and stimulant efficacy
TAAR1 deletion is associated with exaggerated neurochemical
and locomotor responses to amphetamines and their deriva-
tives [49,50]. By contrast, wake-promoting responses to the
psychostimulants modafinil and caffeine were largely normal
in TAAR1 KO mice [136], while locomotor upregulation
induced by both drugs was attenuated in KO mice, as was
drug-induced upregulation of EEG gamma activity. These
results suggest that TAAR1 may have a more central influence
on locomotor and spectral components of psychostimulant
actions than on wake promotion itself, even in the case of
dopaminergic psychostimulants (modafinil acts primarily by
inhibiting DAT [121]).
EXPERT OPINION ON THERAPEUTIC TARGETS 519
5.3. TAAR1 agonism and wake promotion
5.3.1. TAAR1 partial agonism
The partial agonists RO5203648 and RO5263397 increased
total time awake while suppressing non-REM and REM sleep
for up to 3h in WT rats [13,14] and mice [120]. Importantly,
both RO5203648 and RO5263397 promoted wakefulness with-
out increasing locomotor activity, in contrast to the hyperac-
tivity frequently produced by psychostimulants. In mice,
RO5263397 decreased mid- to high-range frequencies in the
waking and NREM EEG spectra, representing the alpha, beta,
and gamma bands [120]; in rats RO5263397 decreased NREM
delta power. This pharmaco-EEG profile was entirely depen-
dent on TAAR1 expression, as RO5263397 was entirely ineffec-
tual when given to Taar1 KO mice, while wake promotion and
REM suppression was strongly potentiated in OE mice [120].
5.3.2. TAAR1 full agonism
The full agonist RO5256390 failed to increase wakefulness in
WT rats and mice when administered in the mid-dark and mid-
light phase, respectively [14,137]. In rats, RO5256390 was
totally ineffective at all doses tested, whereas in WT mice
RO5256390 suppressed REM sleep [137]. This unexpected
result was hypothesized to reflect species differences in the
intrinsic activity of RO5256390, increasing the likelihood of
some partial agonist-like effects in mice compared to rats.
Timing of drug administration may also have played a part;
RO5256390 was tested in rats during the dark phase, when
REM sleep is normally reduced compared to the light phase
[138,139]. On the other hand, RO5256390 elicited a similar EEG
spectral profile as RO5263397 in mice (i.e. decreased power in
the theta, alpha and beta bands; M. Schwartz, unpublished
observations). As with RO5263397, all observed effects on
sleep and waking following RO5256390 were abolished in
Taar1 KO mice, indicating a TAAR1-mediated effect [137].
5.3.3. Prospective mechanisms underlying TAAR1-
mediated wake promotion
The wake-promoting effects of RO5203648 and RO5263397
could result from enhanced monoaminergic signaling follow-
ing partial TAAR1 agonism [14], especially since the full ago-
nist RO5256390 which suppresses monoaminergic signaling
failed to promote wakefulness [14,137]. Similarly, the pro-
found REM-suppressing effect of TAAR1 partial agonism could
be mediated via enhanced DA signaling [122,140,141] or via
interactions with 5-HT1a and 5-HT1b receptors [11,142,143],
both of which regulate REM sleep [144146]. Thus, the
enhancement of wakefulness would appear to depend heavily
on the antagonist-likeactions of the partial agonists. On the
other hand, the similarity in EEG power spectral profiles
induced by the full and partial agonists suggests a common
mechanism relying on the agonist-induced activation of
TAAR1. While still speculative, this striking combination of
TAAR1 activation and inhibition is rarely seen within the
same assay. To help isolate the contributions of these possible
mechanisms, a specific TAAR1 antagonist suitable for in vivo
studies would be a welcome addition to the existing pharma-
cological toolkit.
5.4. TAAR1 agonism as a narcolepsy therapeutic
Narcolepsy is a sleep disorder characterized by hypersomno-
lence, sleep disruption, sleep paralysis, and cataplexy, a sud-
den loss of skeletal muscle tone during wakefulness.
Narcolepsy arises from dysregulation of the wake-promoting
and wakestabilizing hypocretin/orexin (Hcrt) neurons located
in the lateral hypothalamus [147149]. Current pharmacologi-
cal treatments for narcolepsy, including stimulants such as
amphetamines and modafinil and the GABA agonist gamma-
hydroxy butyrate (GHB), either offer limited therapeutic value
(e.g. Modafinil treats the somnolence but does not improve
cataplexy) or carry significant side effects (e.g. tolerance/abuse
risk, sedation); thus novel therapeutics are needed [150]. To
test efficacy of TAAR1 as a therapeutic target for narcolepsy,
RO5263397 and RO5256390 were given to two different
mouse narcolepsy models, the orexin-ataxin3 mouse [151]in
which Hcrt neurons degenerate shortly after birth, and the
orexin/tTA- diphtheria toxin A fragment (DTA) mouse, in which
Hcrt degeneration is conditionally regulated via doxycycline
access [152]. Both RO5263397 and RO5256390 reduced the
number of cataplexy episodes and the time spent in cataplexy
[137], comparing favorably with the norepinephrine reuptake
inhibitor desipramine, a known anticataplectic [153].
Anticataplectic effects could be mediated via serotonergic
modulation [154,155] and/or D2 signaling [59,156]. At the
highest dose, RO5256390 increased wakefulness in DTA but
not ataxin mice, with no further effects on NREM or REM sleep.
RO5263397 suppressed REM sleep in ataxin but not DTA mice,
without altering wake or NREM sleep. As in WT mice and rats,
neither drug elicited hyperlocomotion.
5.5. Summary
In contrast to the similarity of full and partial agonism on
neurobehavioral assays and studies of addiction, the sleep
studies to date highlight the complexity of TAAR1 signaling.
For example, the full and partial agonists exhibit divergent
actions on wakefulness, but similar impact on EEG spectral
profiles and anticataplectic efficacy. These divergent effects
likely reflect the multimodal nature of TAAR1s actions in vivo
and highlight the complexity of manipulating endogenous
TAAR1 signaling, particularly with partial agonists that exhibit
both agonist- and antagonist-like properties. Nevertheless, the
studies to date suggest a potentially key role for TAAR1 in
regulating arousal state and cortical activation. TAAR1 agon-
ism may also be useful in treating sleep disorders, as demon-
strated by the narcolepsy studies.
6. Conclusions
TAAR1 is a promising locus for treatment of neurological,
neuropsychiatric, and behavioral conditions that have histori-
cally proven difficult to address, including schizophrenia,
addiction, and sleep disorders (Table 1). In fact, two pharma-
cological companies have already initiated late-stage clinical
trials of TAAR1-based drugs in schizophrenia patients [2].
While the influence of TAAR1 on dopaminergic systems is
likely critical to its efficacy, serotonergic and glutamatergic
520 M. D. SCHWARTZ ET AL.
signaling likely also play prominent roles. Indeed, such multi-
modal actions could underlie the utility of TAAR1 agonists in
ameliorating side effects (motor dysregulation, weight gain),
abuse potential (especially for dopaminergic drugs), and lim-
ited therapeutic profile (e.g. positive vs. negative symptoms of
schizophrenia; sleep disturbance vs. cataplexy in narcolepsy).
Similarly, the T1AM studies illustrate the importance of study-
ing endogenous trace amines, which may reveal new thera-
peutic applications [33] as well as roles for other TAARs [157].
7. Expert opinion
TAAR1 is a confirmed regulator of monoaminergic and glu-
tamatergic neurotransmitter systems intimately involved with
psychosis, motivation, affect, impulse control, and cognition.
Research on TAAR1s mechanisms of action to date has
revealed a critical role in regulating dopaminergic tone via
multiple intracellular pathways. In particular, TAAR1 power-
fully regulates mesocorticolimbic dopamine circuits underly-
ing motivation and reward, and TAAR1-based interventions
are effective in curbing addictive responses. Pharmacological
targeting of TAAR1 shows great promise in a variety of
neuropsychiatric diseases, including but not limited to schi-
zophrenia, addiction, depression, ADHD, Parkinson disease,
and OCD. Indeed, TAAR1-based small molecules have already
entered clinical trials for treatment of depression and schizo-
phrenia. The addiction field in particular appears promising
for development of TAAR1 as a therapeutic target, on the
basis of TAAR1s involvement in regulating behaviors asso-
ciated with abuse of psychostimulants, ethanol [158], and
nicotine [105,161].
While TAAR1 also regulates serotonergic and glutamatergic
signaling, the downstream impact of these actions is less clear
at present and therefore represents a key knowledge gap.
Major questions include improved understanding of the intra-
cellular signaling pathways underlying TAAR1s actions in 5HT
and glutamate neurons; whether TAAR1 similarly regulates
homeostasisof 5HT and glutamate signaling as it appears
to do for DA circuits, and whether such processes generalize
across multiple neurotransmitter systems, as well as within
them. Thus, in the coming years, TAAR1 is expected to play
a central role in elucidating the neurochemical bases of affect,
cognition, and their disturbances in mental illness. Further
novel therapeutic applications are not only conceivable, but
likely.
A second major growth area for future exploration lies in
integrative physiological functions like metabolism and sleep.
A full TAAR1 agonist was shown to decrease weight gain, food
intake, and glucose dysregulation [159], while T1AM exhibits
both thermoregulatory and food intake-related effects. TAAR1
as well as other TAARs are expressed in the periphery; while
not a focus of this article, the roles of peripheral TAARs and
trace amine signaling in regulating physiological functions like
metabolism should be evaluated alongside the CNS-related
effects.
Similarly, the actions of TAAR1 partial agonism on sleep
and wakefulness, EEG spectra and cataplectic symptoms likely
involve a combination of dopaminergic, serotonergic, and
glutamatergic effects. This presents an opportunity to explore
TAAR1s actions on a more mechanistic level, particularly in
aspects of neural function known to be dysregulated in neu-
ropsychiatric illness, such as EEG gamma band activity in
schizophrenia. Sleep disturbance is a common comorbidity
for mental illness [160]. By directly acting on monoamine
and glutamate transmission, TAAR1 may therefore provide
an entry point for understanding the neural basis for these
commonalities. Novel TAAR1-directed therapeutics thus have
the potential to ameliorate both primary neurosychiatric
symptoms along with debilitating comorbidities such as
sleep disruption.
Our understanding of TAAR1s actions in CNS has benefited
immensely from the recent development of transgenic ani-
mals and especially from new, highly selective small-molecule
ligands such as the Roche full and partial agonists. However, a
lack of selective brain-permeable TAAR1 antagonists with
good in vivo pharmacokinetic properties has hampered devel-
opment of preclinical animal models. The partial and full
TAAR1 agonists developed by Roche show a combination of
similar effects (e.g. antipsychotic actions) and divergent
actions (e.g. wake promotion); while the divergent effects are
typically hypothesized to be mediated by the antagonist-like
profile of the partial agonists, a bona fide TAAR1 antagonist
suitable for in vivo study would be most helpful in parsing out
these mechanisms. Future development of such compounds
will certainly broaden the repertoire of pathological conditions
that could be managed by targeting TAAR1.
A significant gap remains between what is known of the
cellular and molecular actions of TAAR1 and the behavioral
outputs resulting from those actions. Future efforts should
therefore work toward isolating the individual contributions
of dopaminergic, serotonergic, and glutamatergic circuits to
the behavioral/organismal effects seen to date as described
Table 1. Potential therapeutic applications for TAAR1, based on preclinical/
proof-of-concept studies.
Clinical
indication Assay Species Source
Schizophrenia Stimulant-induced Hyperactivity Mouse, rat 13, 14
Object retrieval task NHP 13, 14
Attentional set shift Rat 14
Prepulse inhibition Mouse 50
Depression Forced swim test Rat 13, 14, 90
Anxiety Stress-induced hyperthermia Mouse 13, 14, 90
ADHD Spontaneous hyperactivity Mouse, rat 11, 13, 62
Impulsivity Mouse 84
Reinforcement of low-rate
behavior
NHP 13
OCD Schedule-induced polydipsia Rat 89
Compulsive eating Rat 90
Addiction Locomotor sensitization Rat 101103
Self-administration (cocaine) Rat 13, 97, 104
Self-administration
(methamphetamine)
Rat 98, 101102
Reinstatement/relapse (cocaine,
methamphetamine)
Rat 97, 98, 103,
106
Nicotine addiction Rat 105
Ethanol sensitivity, consumption Mouse 160
Metabolism Food intake Mouse 2526, 158
Weight gain Rat, mouse 13, 158
Glucose regulation mouse 27, 158
Narcolepsy Wake promotion Mouse, rat 13, 14, 28,
120
Cataplexy reduction mouse 137
EXPERT OPINION ON THERAPEUTIC TARGETS 521
above. These will require novel tools and applications, espe-
cially genetic approaches to isolate, identify, and interrogate
TAAR1-expressing neurons and development of selective
human and animal TAAR1 antibodies. Such work will clarify
how TAAR1 regulates neuronal toneand thereby modulates
how the brain experiences, and responds to, environmental
stimuli in intact and pathological conditions. Finally, clinical
studies evaluating the efficacy of TAAR1 agonists in psychiatric
patients are in progress; the results of these studies will
powerfully shape the direction of future research in this field.
Funding
M. D. Schwartz is supported by NIH #NS098813, I. Sukhanov is supported
by the Russian Science Foundation Grant #17-75-20177 and R. R.
Gainetdinov is supported by the Russian Science Foundation Grant #14-
50-00069.
Declaration of Interest
The authors have no relevant affiliations or financial involvement with any
organization or entity with a financial interest in or financial conflict with
the subject matter or materials discussed in the manuscript. This includes
employment, consultancies, honoraria, stock ownership or options, expert
testimony, grants or patents received or pending, or royalties. Peer
reviewers on this manuscript have no relevant financial or other relation-
ships to disclose
ORCID
Michael D. Schwartz http://orcid.org/0000-0002-3472-8900
Raul R. Gainetdinov http://orcid.org/0000-0003-2951-6038
References
Papers of special note have been highlighted as either of interest ()orof
considerable interest (••) to readers.
1. Grandy DK. Trace amine-associated receptor 1family archetype
or iconoclast? Pharmacology & Therapeutics. 2007;116(3):355390.
2. Berry MD, Gainetdinov RR, Hoener MC, et al. Pharmacology of
human trace amine-associated receptors: therapeutic opportunities
and challenges. Pharmacol Ther. 2017 Jul 16;180:161180.
A comprehensive review of trace amines and TAAR1
pharmacology.
3. Borowsky B, Adham N, Jones KA, et al. Trace amines: identification
of a family of mammalian G protein-coupled receptors. Proc Natl
Acad Sci U S A. 2001 Jul 31;98(16):89668971. PubMed PMID:
11459929; PubMed Central PMCID: PMCPMC55357.
•• One of two papers that independently identified TAAR1.
4. Bunzow JR, Sonders MS, Arttamangkul S, et al. Amphetamine, 3,4-
methylenedioxymethamphetamine, lysergic acid diethylamide, and
metabolites of the catecholamine neurotransmitters are agonists of
a rat trace amine receptor. Mol Pharmacol. 2001 Dec;60(6):1181
1188. PubMed PMID: 11723224.
•• The second of two papers that independently identified TAAR1.
5. Lindemann L, Hoener MC. A renaissance in trace amines inspired
by a novel GPCR family. Trends Pharmacol Sci. 2005 May;26(5):274
281. PubMed PMID: 15860375
6. Liberles SD, Buck LB. A second class of chemosensory receptors in
the olfactory epithelium. Nature. 2006 Aug 10;442(7103):645650.
PubMed PMID: 16878137.
Identification of TAAR family members as olfactory receptors.
7. Asif-Malik A, Hoener MC, Canales JJ. Interaction between the trace
amine-associated receptor 1 and the dopamine D2 receptor con-
trols cocaines neurochemical actions. Sci Rep. 2017 Oct 24;7
(1):13901. 10.1038/s41598-017-14472-z. PubMed PMID: 29066851;
PubMed Central PMCID: PMCPMC5655641.
•• Reveals TAAR1-dopaminergic interactions underlying addic-
tive properties of cocaine.
8. Espinoza S, Salahpour A, Masri B, et al. Functional interaction
between trace amine-associated receptor 1 and dopamine D2
receptor. Mol Pharmacol. 2011 Sep;80(3):416425. PubMed PMID:
21670104; PubMed Central PMCID: PMCPMC3164335.
9. Leo D, Mus L, Espinoza S, et al. Taar1-mediated modulation of
presynaptic dopaminergic neurotransmission: role of D2 dopamine
autoreceptors. Neuropharmacology. 2014 Jun;81:283291. PubMed
PMID: 24565640.
10. Bradaia A, Trube G, Stalder H, et al. The selective antagonist EPPTB
reveals TAAR1-mediated regulatory mechanisms in dopaminergic
neurons of the mesolimbic system. Proc Natl Acad Sci U S A. 2009
Nov 24;106(47):2008120086. PubMed PMID: 19892733; PubMed
Central PMCID: PMCPMC2785295.
•• Reveals inhibitory actions of TAAR1 on monoaminergic
neurotransmission.
11. Revel FG, Moreau JL, Gainetdinov RR, et al. TAAR1 activation mod-
ulates monoaminergic neurotransmission, preventing hyperdopa-
minergic and hypoglutamatergic activity. Proc Natl Acad Sci U S A.
2011 May 17;108(20):84858490. PubMed PMID: 21525407;
PubMed Central PMCID: PMCPMC3101002.
First report about in vivo effects of TAAR1 agonists.
12. Revel F, Bradaia A, Trube G, et al. Modulation of dopaminergic activity
in the mesolimbic system by trace amine-associated receptor 1
(TAAR1) modification. Eur Neuropsychopharmacol. 2009;19:S273.
13. Revel FG, Moreau JL, Gainetdinov RR, et al. Trace amine-associated
receptor 1 partial agonism reveals novel paradigm for neuropsy-
chiatric therapeutics. Biol Psychiatry. 2012 Dec 1;72(11):934942.
PubMed PMID: 22705041.
This study demonstrates antipsychotic effects of TAAR1
agonists.
14. Revel FG, Moreau JL, Pouzet B, et al. A new perspective for schizo-
phrenia: TAAR1 agonists reveal antipsychotic- and antidepressant-
like activity, improve cognition and control body weight. Mol
Psychiatry. 2013 May;18(5):543556. PubMed PMID: 22641180.
•• This study demonstrated preclinical efficacy of TAAR1 agonists
in neuropsychiatric disease models, addiction, and modulating
sleep-wake state.
15. Stalder H, Hoener MC, Norcross RD. Selective antagonists of mouse
trace amine-associated receptor 1 (mTAAR1): discovery of EPPTB
(RO5212773). Bioorg Med Chem Lett. 2011 Feb 15;21(4):12271231.
PubMed PMID: 21237643.
16. Scanlan TS, Suchland KL, Hart ME, et al. 3-Iodothyronamine is an
endogenous and rapid-acting derivative of thyroid hormone. Nat
Med. 2004 Jun;10(6):638642. PubMed PMID: 15146179.
•• Identifies T1AM as an endogenous TAAR1 agonist.
17. Hoefig CS, Wuensch T, Rijntjes E, et al. Biosynthesis of 3-iodothyr-
onamine from t4 in murine intestinal tissue. Endocrinology. 2015
Nov;156(11):43564364. PubMed PMID: 26348473.
18. Galli E, Marchini M, Saba A, et al. Detection of 3-iodothyronamine
in human patients: a preliminary study. J Clin Endocrinol Metab.
2012 Jan;97(1):E6974. PubMed PMID: 22031514.
19. Hoefig CS, Kohrle J, Brabant G, et al. Evidence for extrathyroidal
formation of 3-iodothyronamine in humans as provided by a novel
monoclonal antibody-based chemiluminescent serum immunoas-
say. J Clin Endocrinol Metab. 2011 Jun;96(6):18641872. PubMed
PMID: 21490071.
20. Laurino A, De Siena G, Saba A, et al. In the brain of mice, 3-
iodothyronamine (T1AM) is converted into 3-iodothyroacetic acid
(TA1) and it is included within the signaling network connecting
thyroid hormone metabolites with histamine. Eur J Pharmacol.
2015 Aug 15;761:130134. PubMed PMID: 25941083.
21. Manni ME, De Siena G, Saba A, et al. Pharmacological effects of 3-
iodothyronamine (T1AM) in mice include facilitation of memory
acquisition and retention and reduction of pain threshold. Br J
Pharmacol. 2013 Jan;168(2):354362. PubMed PMID: 22889145;
PubMed Central PMCID: PMCPMC3572562.
522 M. D. SCHWARTZ ET AL.
First report of T1AMs procognitive effects.
22. Musilli C, De Siena G, Manni ME, et al. Histamine mediates beha-
vioural and metabolic effects of 3-iodothyroacetic acid, an endo-
genous end product of thyroid hormone metabolism. Br J
Pharmacol. 2014 Jul;171(14):34763484. PubMed PMID: 24641572;
PubMed Central PMCID: PMCPMC4105934.
23. Saba A, Chiellini G, Frascarelli S, et al. Tissue distribution and
cardiac metabolism of 3-iodothyronamine. Endocrinology. 2010
Oct;151(10):50635073. PubMed PMID: 20739399.
24. Coster M, Biebermann H, Schoneberg T, et al. Evolutionary conserva-
tion of 3-iodothyronamine as an agonist at the trace amine-asso-
ciated receptor 1. Eur Thyroid J. 2015 Sep;4(Suppl 1):920. PubMed
PMID: 26601069; PubMed Central PMCID: PMCPMC4640299.
25. Zucchi R, Accorroni A, Chiellini G. Update on 3-iodothyronamine
and its neurological and metabolic actions. Front Physiol.
2014;5:402. PubMed PMID: 25360120; PubMed Central PMCID:
PMCPMC4199266. .
Review of T1AM and its effects on CNS and peripheral physiology.
26. Dhillo WS, Bewick GA, White NE, et al. The thyroid hormone deri-
vative 3-iodothyronamine increases food intake in rodents.
Diabetes Obes Metab. 2009 Mar;11(3):251260. PubMed PMID:
18671794.
27. Manni ME, De Siena G, Saba A, et al. 3-Iodothyronamine: a mod-
ulator of the hypothalamus-pancreas-thyroid axes in mice. Br J
Pharmacol. 2012 May;166(2):650658. PubMed PMID: 22225569;
PubMed Central PMCID: PMCPMC3417495.
28. James TD, Moffett SX, Scanlan TS, et al. Effects of acute microinjec-
tions of the thyroid hormone derivative 3-iodothyronamine to the
preoptic region of adult male rats on sleep, thermoregulation and
motor activity. Horm Behav. 2013 Jun;64(1):8188. PubMed PMID:
23702093; PubMed Central PMCID: PMCPMC4091812.
29. Gompf HS, Greenberg JH, Aston-Jones G, et al. 3-Monoiodo
thyronamine: the rationale for its action as an endogenous adrener-
gic-blocking neuromodulator. Brain Res. 2010 Sep 10;1351:130140.
PubMed PMID: 20615397; PubMed Central PMCID: PMCPMC2926234.
30. Brown RE, Basheer R, McKenna JT, et al. Control of sleep and
wakefulness. Physiol Rev. 2012 Jul;92(3):10871187. PubMed
PMID: 22811426; PubMed Central PMCID: PMCPMC3621793.
31. Jones BE. From waking to sleeping: neuronal and chemical sub-
strates. Trends Pharmacol Sci. 2005 Nov;26(11):578586. PubMed
PMID: 16183137.
32. Saper CB, Fuller PM, Pedersen NP, et al. Sleep state switching.
Neuron. 2010 Dec 22;68(6):10231042.
33. Accorroni A, Criscuolo C, Sabatini M, et al. 3-iodothyronamine and
trace amine-associated receptor 1 are involved in the expression of
long-term potentiation in mouse enthorhinal cortex. [abstract]. Eur
Thyroid J. 2016;5(1):2122.
34. Bräunig J, Dinter J, Höfig CS, Paisdzior S, Szczepek M, Scheerer P,
Rosowski M, Mittag J, Kleinau G, Biebermann H. The Trace Amine-
Associated Receptor 1 Agonist 3-Iodothyronamine Induces Biased
Signaling at the Serotonin 1b Receptor. Front Pharmacol. 2018 Mar
12;9222. DOI: 10.3389/fphar.2018.00222. eCollection 2018. PMID:
29593543
35. Hoefig CS, Zucchi R, Kohrle J. Thyronamines and derivatives: physiolo-
gical relevance, pharmacological actions, and future research directions.
Thyroid. 2016 Dec;26(12):16561673. PubMed PMID: 27650974.
Comprehensive review of thyronamine biology.
36. Hart ME, Suchland KL, Miyakawa M, et al. Trace amine-associated
receptor agonists: synthesis and evaluation of thyronamines and
related analogues. J Med Chem. 2006 Feb 9;49(3):11011112.
PubMed PMID: 16451074.
37. Tan ES, Miyakawa M, Bunzow JR, et al. Exploring the structure-
activity relationship of the ethylamine portion of 3-iodothyrona-
mine for rat and mouse trace amine-associated receptor 1. J Med
Chem. 2007 Jun 14;50(12):27872798. PubMed PMID: 17497842.
38. Chiellini G, Nesi G, Digiacomo M, et al. Design, synthesis, and
evaluation of thyronamine analogues as novel potent Mouse
Trace Amine Associated Receptor 1 (mTAAR1) agonists. J Med
Chem. 2015 Jun 25;58(12):50965107. PubMed PMID: 26010728.
39. Chiellini G, Nesi G, Sestito S, et al. Hit-to-lead optimization of
Mouse Trace Amine Associated Receptor 1 (mTAAR1) agonists
with a diphenylmethane-scaffold: design, synthesis, and biological
study. J Med Chem. 2016 Nov 10;59(21):98259836. PubMed PMID:
27731647.
40. Chiellini G, Bellusci L, Sabatini M, et al. Thyronamines and analo-
gues - the route from rediscovery to translational research on
thyronergic amines. Mol Cell Endocrinol. 2017 Dec 15;458:149
155. PubMed PMID: 28069535.
41. Boulton AA. Some aspects of basic psychopharmacology: the
trace amines [Research Support, Non-U.S. Govt]. Prog
Neuropsychopharmacol Biol Psychiatry. 1982;6(46): 563570.
PubMed PMID: 6298892; eng.
42. Davis BA. Biogenic amines and their metabolites in body fluids of
normal, psychiatric and neurological subjects [Research Support,
Non-U.S. Govt Review]. J Chromatogr. 1989 Apr 19;466:89218.
PubMed PMID: 2663901; eng.
43. Potkin SG, Karoum F, Chuang LW, et al. Phenylethylamine in para-
noid chronic schizophrenia. Science. 1979 Oct 26;206(4417):470
471. PubMed PMID: 504988; eng.
44. Davis BA, Boulton AA. The trace amines and their acidic metabo-
lites in depressionan overview [Research Support, Non-U.S. Govt
Review]. Prog Neuropsychopharmacol Biol Psychiatry. 1994 Jan;18
(1):1745. PubMed PMID: 8115671; eng.
45. Sandler M, Ruthven CR, Goodwin BL, et al. Decreased cerebrospinal
fluid concentration of free phenylacetic acid in depressive illness
[Comparative Study]. Clin Chim Acta. 1979 Apr 2;93(1):169171.
PubMed PMID: 436296; eng.
46. Szabo A, Billett E, Turner J. Phenylethylamine, a possible link to the
antidepressant effects of exercise? [Clinical Trial]. Br J Sports Med.
2001 Oct;35(5):342343. PubMed PMID: 11579070; PubMed Central
PMCID: PMC1724404. eng.
47. Beaulieu JM, Gainetdinov RR. The physiology, signaling, and phar-
macology of dopamine receptors. Pharmacol Rev. 2011 Mar;63
(1):182217. PubMed PMID: 21303898.
Comprehensive review of DA receptor signaling, including
relation to mental illness.
48. Berry MD. Mammalian central nervous system trace amines.
Pharmacologic amphetamines, physiologic neuromodulators. J
Neurochem. 2004 Jul;90(2):257271. PubMed PMID: 15228583.
49. Di Cara B, Maggio R, Aloisi G, et al. Genetic deletion of trace amine
1 receptors reveals their role in auto-inhibiting the actions of
ecstasy (MDMA). J Neurosci. 2011 Nov 23;31(47):1692816940.
PubMed PMID: 22114263.
50. Lindemann L, Meyer CA, Jeanneau K, et al. Trace amine-associated
receptor 1 modulates dopaminergic activity. J Pharmacol Exp Ther.
2008 Mar;324(3):948956. PubMed PMID: 18083911.
Shows that TAAR1 deletion leads to dysregulated DA signaling
and psychostimulant responses.
51. Wolinsky TD, Swanson CJ, Smith KE, et al. The Trace Amine 1
receptor knockout mouse: an animal model with relevance to
schizophrenia. Genes Brain Behav. 2007 Oct;6(7):628639.
PubMed PMID: 17212650.
•• Shows that TAAR1 deletion elicits schizophrenia-relevant
phenotypes.
52. Seeman P. Dopamine D2 receptors as treatment targets in schizo-
phrenia. Clin Schizophr Relat Psychoses. 2010 Apr;4(1):5673.
PubMed PMID: 20643630.
53. Zetterstrom T, Sharp T, Marsden CA, et al. In vivo measurement of
dopamine and its metabolites by intracerebral dialysis: changes
after d-amphetamine. J Neurochem. 1983 Dec;41(6):17691773.
PubMed PMID: 6196446.
54. Zhuang X, Oosting RS, Jones SR, et al. Hyperactivity and impaired
response habituation in hyperdopaminergic mice. Proc Natl Acad
Sci U S A. 2001 Feb 13;98(4):19821987. PubMed PMID: 11172062;
PubMed Central PMCID: PMCPMC29368.
55. Groves PM, Rebec GV. Biochemistry and behavior: some central
actions of amphetamine and antipsychotic drugs. Annu Rev
Psychol. 1976;27:91127. PubMed PMID: 773267.
EXPERT OPINION ON THERAPEUTIC TARGETS 523
56. Lipska BK, Weinberger DR. To model a psychiatric disorder in ani-
mals: schizophrenia as a reality test. Neuropsychopharmacology.
2000 Sep;23(3):223239. PubMed PMID: 10942847.
57. Panas HN, Lynch LJ, Vallender EJ, et al. Normal thermoregulatory
responses to 3-iodothyronamine, trace amines and amphetamine-
like psychostimulants in trace amine associated receptor 1 knock-
out mice. J Neurosci Res. 2010 Jul;88(9):19621969. PubMed PMID:
20155805; PubMed Central PMCID: PMCPMC3587846.
58. Achat-Mendes C, Lynch LJ, Sullivan KA, et al. Augmentation of
methamphetamine-induced behaviors in transgenic mice lacking
the trace amine-associated receptor 1. Pharmacol Biochem Behav.
2012 Apr;101(2):201207. PubMed PMID: 22079347; PubMed
Central PMCID: PMCPMC3288391.
59. Sotnikova TD, Zorina OI, Ghisi V, et al. Trace amine associated
receptor 1 and movement control. Parkinsonism Relat Disord.
2008;14(Suppl 2):S99102. PubMed PMID: 18585080.
60. Espinoza S, Ghisi V, Emanuele M, et al. Postsynaptic D2 dopamine
receptor supersensitivity in the striatum of mice lacking TAAR1.
Neuropharmacology. 2015 Jun;93:308313. PubMed PMID: 25721394.
61. Sukhanov I, Caffino L, Efimova EV, et al. Increased context-depen-
dent conditioning to amphetamine in mice lacking TAAR1.
Pharmacol Res. 2016 Jan;103:206214. PubMed PMID: 26640076.
62. Revel FG, Meyer CA, Bradaia A, et al. Brain-specific overexpression
of trace amine-associated receptor 1 alters monoaminergic neuro-
transmission and decreases sensitivity to amphetamine.
Neuropsychopharmacology. 2012 Nov;37(12):25802592. PubMed
PMID: 22763617; PubMed Central PMCID: PMCPMC3473323.
63. Leo D, Sukhanov I, Zoratto F, et al. Pronounced hyperactivity,
cognitive dysfunctions and Bdnf dysregulation in dopamine trans-
porter knockout rats. J Neurosci. 2018 Jan 18:171931. PubMed
PMID: 29348190. DOI: 10.1523/JNEUROSCI.1931-17.2018
64. Danysz W, Essmann U, Bresink I, et al. Glutamate antagonists have
different effects on spontaneous locomotor activity in rats.
Pharmacol Biochem Behav. 1994 May;48(1):111118. PubMed
PMID: 8029281.
65. Ford LM, Norman AB, Sanberg PR. The topography of MK-801-
induced locomotor patterns in rats. Physiol Behav. 1989 Oct;46
(4):755758. PubMed PMID: 2557649.
66. Gleason SD, Shannon HE. Blockade of phencyclidine-induced
hyperlocomotion by olanzapine, clozapine and serotonin receptor
subtype selective antagonists in mice. Psychopharmacology (Berl).
1997 Jan;129(1):7984. PubMed PMID: 9122367.
67. Harkness JH, Shi X, Janowsky A, et al. Trace amine-associated
receptor 1 regulation of methamphetamine intake and related
traits. Neuropsychopharmacology. 2015 Aug;40(9):21752184.
PubMed PMID: 25740289; PubMed Central PMCID:
PMCPMC4613607.
68. Jing L, Li JX. Trace amine-associated receptor 1: A promising target
for the treatment of psychostimulant addiction. Eur J Pharmacol.
2015 Aug 15;761:345352.
69. Thorn DA, Zhang C, Zhang Y, et al. The trace amine associated
receptor 1 agonist RO5263397 attenuates the induction of cocaine
behavioral sensitization in rats. Neurosci Lett. 2014 Apr 30;566:67
71. PubMed PMID: 24561093; PubMed Central PMCID:
PMCPMC3991844.
70. Sukhanov I, Espinoza S, Yakovlev DS, et al. TAAR1-dependent
effects of apomorphine in mice. Int J Neuropsychopharmacol.
2014 Oct;17(10):16831693. PubMed PMID: 24925023.
71. Costall B, Naylor RJ, Nohria V. Climbing behaviour induced by
apomorphine in mice: a potential model for the detection of
neuroleptic activity. Eur J Pharmacol. 1978 Jul 1;50(1):3950.
PubMed PMID: 28233.
72. Protais P, Costentin J, Schwartz JC. Climbing behavior induced by
apomorphine in mice: a simple test for the study of dopamine
receptors in striatum. Psychopharmacology (Berl). 1976 Oct 20;50
(1):16. PubMed PMID: 827755.
73. Puech AJ, Simon P, Boissier JR. Antagonism by sulpiride of three
apomorphine-induced effects in rodents. Eur J Pharmacol. 1976
Apr;36(2):439441. PubMed PMID: 945169.
74. Sharma T, Antonova L. Cognitive function in schizophrenia. Deficits,
functional consequences, and future treatment. Psychiatr Clin
North Am. 2003 Mar;26(1):2540. PubMed PMID: 12683258.
75. Geyer MA, Krebs-Thomson K, Braff DL, et al. Pharmacological stu-
dies of prepulse inhibition models of sensorimotor gating deficits
in schizophrenia: a decade in review. Psychopharmacology (Berl).
2001 Jul;156(23):117154. PubMed PMID: 11549216.
76. Alvarsson A, Zhang X, Stan TL, et al. Modulation by trace amine-
associated receptor 1 of experimental parkinsonism, L-DOPA
responsivity, and glutamatergic neurotransmission. J Neurosci.
2015 Oct 14;35(41):1405714069. PubMed PMID: 26468205.
77. Sotnikova TD, Beaulieu JM, Barak LS, et al. Dopamine-independent
locomotor actions of amphetamines in a novel acute mouse model
of Parkinson disease. PLoS Biol. 2005 Aug;3(8):e271. PubMed PMID:
16050778; PubMed Central PMCID: PMCPMC1181539.
78. Cheon KA, Ryu YH, Kim YK, et al. Dopamine transporter density in
the basal ganglia assessed with [123I]IPT SPET in children with
attention deficit hyperactivity disorder. Eur J Nucl Med Mol
Imaging. 2003 Feb;30(2):306311. PubMed PMID: 12552351.
79. DiMaio S, Grizenko N, Joober R. Dopamine genes and attention-
deficit hyperactivity disorder: a review. J Psychiatry Neurosci. 2003
Jan;28(1):2738. PubMed PMID: 12587848.
80. Dougherty DD, Bonab AA, Spencer TJ, et al. Dopamine transporter
density in patients with attention deficit hyperactivity disorder.
Lancet. 1999 Dec 18- 25;354(9196):21322133. PubMed PMID:
10609822.
81. Leo D, Gainetdinov RR. Transgenic mouse models for ADHD. Cell
Tissue Res. 2013 Oct;354(1):259271. PubMed PMID: 23681253;
PubMed Central PMCID: PMCPMC3785710.
82. Gainetdinov RR, Mohn AR, Bohn LM, et al. Glutamatergic modula-
tion of hyperactivity in mice lacking the dopamine transporter.
Proc Natl Acad Sci U S A. 2001 Sep 25;98(20):1104711054.
PubMed PMID: 11572967.
83. Giros B, Jaber M, Jones SR, et al. Hyperlocomotion and indifference
to cocaine and amphetamine in mice lacking the dopamine trans-
porter. Nature. 1996 Feb 15;379(6566):606612. PubMed PMID:
8628395.
84. Gainetdinov RR, Wetsel WC, Jones SR, et al. Role of serotonin in the
paradoxical calming effect of psychostimulants on hyperactivity.
Science. 1999 Jan 15;283(5400):397401. PubMed PMID: 9888856.
85. Espinoza S, Lignani G, Caffino L, et al. TAAR1 modulates cortical
glutamate NMDA receptor function. Neuropsychopharmacology.
2015 Aug;40(9):22172227. PubMed PMID: 25749299; PubMed
Central PMCID: PMCPMC4613611.
Study showing TAAR1s role in controlling response of NMDA
receptors.
86. Xue Z, Siemian JN, Johnson BN, et al. Methamphetamine-induced
impulsivity during chronic methamphetamine treatment in rats:
effects of the TAAR 1 agonist RO5263397. Neuropharmacology.
2018 Feb;129:3646. PubMed PMID: 29128305.
87. Cinque S, Zoratto F, Poleggi A, et al. Behavioral phenotyping of
dopamine transporter knockout rats: compulsive traits, motor
stereotypies, and anhedonia. Front Psychiatry. 2018;9:43. PubMed
PMID: 29520239; PubMed Central PMCID: PMCPMC5826953.
88. Kellner M. Drug treatment of obsessive-compulsive disorder.
Dialogues Clin Neurosci. 2010;12(2): 187197. PubMed PMID:
20623923; PubMed Central PMCID: PMCPMC3181958.
89. Koo MS, Kim EJ, Roh D, et al. Role of dopamine in the pathophy-
siology and treatment of obsessive-compulsive disorder. Expert
Rev Neurother. 2010 Feb;10(2):275290. PubMed PMID: 20136383.
90. Dolgorukova A, Dorotenko A, Mus L, et al. Activation of trace
amine-associated receptor 1 reduces schedule-induced polydipsia
in rats. Eur Neuropsychopharmacol. 2017 Oct;27:S673S673.
PubMed PMID: WOS:000413847701074; English.
100. Ferragud A, Howell AD, Moore CF, et al. The trace amine-asso-
ciated receptor 1 agonist RO5256390 blocks compulsive, binge-
like eating in rats. Neuropsychopharmacology. 2017 Jun;42
(7):14581470. PubMed PMID: 27711047; PubMed Central PMCID:
PMCPMC5436108.
524 M. D. SCHWARTZ ET AL.
101. Can A, Dao DT, Arad M, et al. The mouse forced swim test. J Vis Exp.
2012 Jan 29;59:e3638. PubMed PMID: 22314943; PubMed Central
PMCID: PMCPMC3353513. DOI:10.3791/3638
102. Ashok AH, Marques TR, Jauhar S, et al. The dopamine hypothesis of
bipolar affective disorder: the state of the art and implications for
treatment. Mol Psychiatry. 2017 May;22(5):666679. PubMed PMID:
28289283; PubMed Central PMCID: PMCPMC5401767.
103. Kampman KM. Whats new in the treatment of cocaine addiction? Curr
Psychiatry Rep. 2010 Oct;12(5):441447. PubMed PMID: 20697850.
104. Penberthy JK, Ait-Daoud N, Vaughan M, et al. Review of treatment
for cocaine dependence. Curr Drug Abuse Rev. 2010 3;Mar(1):49
62. PubMed PMID: 20088819.
105. Lin Z, Canales JJ, Bjorgvinsson T, et al. Monoamine transporters:
vulnerable and vital doorkeepers. Prog Mol Biol Transl Sci.
2011;98:146. PubMed PMID: 21199769.
106. Velazquez-Sanchez C, Canales JJ. Atypical dopamine transporter
inhibitors: candidates for the treatment of psychostimulant addic-
tion. In: Canales JJ, ed. Emerging targets for drug addiction treat-
ment. New York: Nova Publishers; 2012 Jul 1. p. 103150. S0278-
5846(13)00018-3 [pii]. PubMed PMID: 23385166; eng. DOI:10.1016/j.
pnpbp.2013.01.016
107. Pei Y, Lee J, Leo D, et al. Activation of the trace amine-associated
receptor 1 prevents relapse to cocaine seeking. Neuropsychophar
macology. 2014;39(10):22992308.
108. Pei Y, Asif-Malik A, Hoener M, et al. A partial trace amine-associated
receptor 1 agonist exhibits properties consistent with a metham-
phetamine substitution treatment. Addict Biol. 2017 Sep;22
(5):12461256. PubMed PMID: 27193165.
109. Marinelli M, White FJ. Enhanced vulnerability to cocaine self-admin-
istration is associated with elevated impulse activity of midbrain
dopamine neurons. J Neurosci. 2000 Dec 1;20(23):88768885.
PubMed PMID: 11102497.
110. McCutcheon JE, White FJ, Marinelli M. Individual differences in
dopamine cell neuroadaptations following cocaine self-administra-
tion. Biol Psychiatry. 2009 Oct 15;66(8):801803. PubMed PMID:
19539267.
111. Cotter R, Pei Y, Mus L, et al. The trace amine-associated receptor 1
modulates methamphetamines neurochemical and behavioral
effects. Frontiers in Neuroscience. 2015;9:39.
112. Jing L, Zhang Y, Li J-X. Effects of the trace amine associated
receptor 1 agonist RO5263397 on abuse-related behavioral indices
of methamphetamine in rats. Int J Neuropsychopharmacol. 2014;18
(4):pyu060.
113. Thorn DA, Jing L, Qiu Y, et al. Effects of the trace amine-associated
receptor 1 agonist RO5263397 on abuse-related effects of cocaine
in rats. Neuropsychopharmacology. 2014;39(10:2309-2316.
114. Pei Y, Mortas P, Hoener MC, et al. Selective activation of the trace
amine-associated receptor 1 decreases cocaines reinforcing effi-
cacy and prevents cocaine-induced changes in brain reward
thresholds. Prog Neuropsychopharmacol Biol Psychiatry.
2015;63:7075. PubMed PMID: 26048337.
115. Liu JF, Seaman R Jr., Siemian JN, et al. Role of trace amine-asso-
ciated receptor 1 in nicotinesbehavioral and neurochemical
effects. Neuropsychopharmacology. 2018 Feb 5. 10.1038/s41386-
018-0017-9. PubMed PMID: 29472642.
116. Liu JF, Siemian JN, Seaman R Jr., et al. Role of TAAR1 within the
subregions of the mesocorticolimbic dopaminergic system in
cocaine-seeking behavior. J Neurosci. 2017 Jan 25;37(4):882892.
PubMed PMID: 28123023.
117. Xie Z, Miller GM. Trace amine-associated receptor 1 is a modulator
of the dopamine transporter. J Pharmacol Exp Ther. 2007 Apr;321
(1):128136.
118. Miller GM, Verrico CD, Jassen A, et al. Primate trace amine receptor
1 modulation by the dopamine transporter. J Pharmacol Exp Ther.
2005;313(3):983994.
119. Harmeier A, Obermueller S, Meyer CA, et al. Trace amine-associated
receptor 1 activation silences GSK3βsignaling of TAAR1 and D2R
heteromers. Eur Neuropsychopharmacol. 2015;25:20492061.
120. Xu CM, Wang J, Wu P, et al. Glycogen synthase kinase 3beta in the
nucleus accumbens core mediates cocaine-induced behavioral
sensitization. J Neurochem. 2009 Dec;111(6):13571368. PubMed
PMID: 19799712.
121. Shi X, Miller JS, Harper LJ, et al. Reactivation of cocaine reward
memory engages the Akt/GSK3/mTOR signaling pathway and can
be disrupted by GSK3 inhibition. Psychopharmacology (Berl). 2014
Aug;231(16):31093118. PubMed PMID: 24595501.
122. Daley M, Morin CM, LeBlanc M, et al. The economic burden of
insomnia: direct and indirect costs for individuals with insomnia
syndrome, insomnia symptoms, and good sleepers. Sleep. 2009
Jan;32(1):5564. PubMed PMID: 19189779.
123. Hillman DR, Murphy AS, Pezzullo L. The economic cost of sleep
disorders. Sleep. 2006 Mar;29(3):299305. PubMed PMID: 16553015.
124. Wulff K, Dijk DJ, Middleton B, et al. Sleep and circadian rhythm
disruption in schizophrenia. Br J Psychiatry. 2012 Apr;200(4):308
316. PubMed PMID: 22194182; PubMed Central PMCID:
PMCPMC3317037.
125. Happe S, Baier PC, Helmschmied K, et al. Association of daytime
sleepiness with nigrostriatal dopaminergic degeneration in early
Parkinsons disease. J Neurol. 2007 Aug;254(8):10371043. PubMed
PMID: 17351722.
126. Rye DB. Parkinsons disease and RLS: the dopaminergic bridge.
Sleep Med. 2004 May;5(3):317328. PubMed PMID: 15165542.
127. Brower KJ. Alcohols effects on sleep in alcoholics. Alcohol Res
Health. 2001;25(2): 110125. PubMed PMID: 11584550.
128. Brower KJ, Perron BE. Sleep disturbance as a universal risk factor for
relapse in addictions to psychoactive substances. Med Hypotheses.
2010;74(5):928933. PubMed PMID: 19910125.
129. Scammell TE, Arrigoni E, Lipton JO. Neural Circuitry of Wakefulness
and Sleep. Neuron. 2017 Feb 22;93(4):747765. PubMed PMID:
28231463.
130. Schwartz MD, Black SW, Fisher SP, et al. Trace amine-associated
receptor 1 regulates wakefulness and EEG spectral composition.
Neuropsychopharmacology. 2017 May;42(6):13051314. PubMed
PMID: 27658486; PubMed Central PMCID: PMCPMC5437878.
Shows that endogenous TAAR1 signaling regulates arousal
state.
131. Wisor JP, Nishino S, Sora I, et al. Dopaminergic role in stimulant-
induced wakefulness. J Neurosci. 2001 Mar 01;21(5):17871794.
PubMed PMID: 11222668.
132. Eban-Rothschild A, Rothschild G, Giardino WJ, et al. VTA dopami-
nergic neurons regulate ethologically relevant sleep-wake beha-
viors. Nat Neurosci. 2016 Oct;19(10):13561366. PubMed PMID:
27595385; PubMed Central PMCID: PMCPMC5519826.
133. Cho JR, Treweek JB, Robinson JE, et al. Dorsal raphe dopamine
neurons modulate arousal and promote wakefulness by salient
stimuli. Neuron. 2017 Jun 21;94(6):12051219 e8. PubMed PMID:
28602690.
134. McGinty DJ, Harper RM. Dorsal raphe neurons: depression of firing
during sleep in cats. Brain Res. 1976 Jan 23;101(3):569575.
PubMed PMID: 1244990.
135. Morairty SR, Hedley L, Flores J, et al. Selective 5HT2A and 5HT6
receptor antagonists promote sleep in rats. Sleep. 2008 Jan;31
(1):3444. PubMed PMID: 18220076; PubMed Central PMCID:
PMCPMC2225549.
136. Aston-Jones G, Bloom FE. Activity of norepinephrine-containing
locus coeruleus neurons in behaving rats anticipates fluctuations
in the sleep-waking cycle. J Neurosci. 1981 Aug;1(8):876886.
PubMed PMID: 7346592.
137. Brown RE, McKenna JT. Turning a negative into a positive: ascend-
ing GABAergic control of cortical activation and arousal. Front
Neurol. 2015;6:135. PubMed PMID: 26124745; PubMed Central
PMCID: PMCPMC4463930.
138. Ferris MJ, Espana RA, Locke JL, et al. Dopamine transporters govern
diurnal variation in extracellular dopamine tone. Proc Natl Acad Sci
USA.2014 Jul 01;111(26):E27519. PubMed PMID: 24979798;
PubMed Central PMCID: PMCPMC4084435.
139. Cape EG, Jones BE. Differential modulation of high-frequency
gamma-electroencephalogram activity and sleep-wake state by
noradrenaline and serotonin microinjections into the region of
cholinergic basalis neurons. J Neurosci. 1990;18(7):26532666.
EXPERT OPINION ON THERAPEUTIC TARGETS 525
140. Vertes RP, Kocsis B. Brainstem-diencephalo-septohippocampal sys-
tems controlling the theta rhythm of the hippocampus.
Neuroscience. 1997;81(4):893926. PubMed PMID: 9330355.
141. Feinberg I, Schoepp DD, Hsieh KC, et al. The metabotropic gluta-
mate (mGLU)2/3 receptor antagonist LY341495 [2S-2-amino-2-
(1S,2S-2-carboxycyclopropyl-1-yl)-3-(xanth-9-yl)propanoic acid] sti-
mulates waking and fast electroencephalogram power and blocks
the effects of the mGLU2/3 receptor agonist ly379268 [(-)-2-oxa-4-
aminobicyclo[3.1.0]hexane-4,6-dicarboxylate] in rats. J Pharmacol
Exp Ther. 2005312(2):826833. PubMed PMID: 15383637.
142. Ahnaou A, De Boer P, Lavreysen H, et al. Translational neurophy-
siological markers for activity of the metabotropic glutamate
receptor (mGluR2) modulator JNJ-40411813: sleep EEG correlates
in rodents and healthy men. Neuropharmacology. 2016
Apr;103:290305. PubMed PMID: 26686390.
143. Gilmour G, Broad LM, Wafford KA, et al. In vitro characterisation of
the novel positive allosteric modulators of the mGlu(5) receptor,
LSN2463359 and LSN2814617, and their effects on sleep architec-
ture and operant responding in the rat. Neuropharmacology. 2013
Jan;64:224239. PubMed PMID: 22884720.
144. Ahnaou A, Langlois X, Steckler T, et al. Negative versus positive
allosteric modulation of metabotropic glutamate receptors
(mGluR5): indices for potential pro-cognitive drug properties
based on EEG network oscillations and sleep-wake organization
in rats. Psychopharmacology (Berl). 2015 Mar;232(6):11071122.
PubMed PMID: 25323624.
145. Lindemann L, Porter RH, Scharf SH, et al. Pharmacology of basim-
glurant (RO4917523, RG7090), a unique metabotropic glutamate
receptor 5 negative allosteric modulator in clinical development for
depression. J Pharmacol Exp Ther. 2015 Apr;353(1):213233.
PubMed PMID: 25665805.
146. Schwartz MD, Palmerston JB, Lee DL, et al. Deletion of trace amine-
associated receptor 1 attenuates behavioral responses to caffeine.
Front Pharmacol. 2018;9. DOI:10.3389/fphar.2018.00035
147. Black SW, Schwartz MD, Chen TM, et al. Trace amine-associated
receptor 1 agonists as narcolepsy therapeutics. Biol Psychiatry.
2017 Nov 1;82(9):623633. PubMed PMID: 27919403; PubMed
Central PMCID: PMCPMC5395352.
Demonstrates efficacy of TAAR1 agonism in preclinical models
of narcolepsy.
148. Borbely AA. A two process model of sleep regulation. Hum
Neurobiol. 1982;1(3): 195204. PubMed PMID: 7185792.
149. Mistlberger RE. Circadian regulation of sleep in mammals: role of
the suprachiasmatic nucleus. Brain Res Brain Res Rev. 2005 Nov;49
(3):429454. PubMed PMID: 16269313.
150. Lena I, Parrot S, Deschaux O, et al. Variations in extracellular levels
of dopamine, noradrenaline, glutamate, and aspartate across the
sleepwake cycle in the medial prefrontal cortex and nucleus
accumbens of freely moving rats. J Neurosci Res. 2005 Sep 15;81
(6):891899. PubMed PMID: 16041801.
151. Dahan L, Astier B, Vautrelle N, et al. Prominent burst firing of
dopaminergic neurons in the ventral tegmental area during para-
doxical sleep. Neuropsychopharmacology. 2007 Jun;32(6):1232
1241. PubMed PMID: 17151599.
152. Xie Z, Miller GM. Beta-phenylethylamine alters monoamine trans-
porter function via trace amine-associated receptor 1: implication
for modulatory roles of trace amines in brain. J Pharmacol Exp Ther.
2008 May;325(2):617628. PubMed PMID: 18182557.
153. Xie Z, Westmoreland SV, Miller GM. Modulation of monoamine trans-
porters by common biogenic amines via trace amine-associated
receptor 1 and monoamine autoreceptors in human embryonic kid-
ney 293 cells and brain synaptosomes. J Pharmacol Exp Ther. 2008
May;325(2):629640. PubMed PMID: 18310473.
154. Boutrel B, Franc B, Hen R, et al. Key role of 5-HT1B receptors in the
regulation of paradoxical sleep as evidenced in 5-HT1B knock-out
mice. J Neurosci. 1999;19(8):32043212.
155. Boutrel B, Monaca C, Hen R, et al. Involvement of 5-HT1A recep-
tors in homeostatic and stress-induced adaptive regulations of
paradoxical sleep: studies in 5-HT1A knock-out mice. J Neurosci.
2002 Jun 1;22(11):46864692. doi: 20026427. PubMed PMID:
12040075.
156. Monti JM, Monti D, Jantos H, et al. Effects of selective activation of
the 5-HT1B receptor with CP-94,253 on sleep and wakefulness in
the rat. Neuropharmacology. 1995 Dec;34(12):16471651. PubMed
PMID: 8788962.
157. Chemelli RM, Willie JT, Sinton CM, et al. Narcolepsy in orexin
knockout mice: molecular genetics of sleep regulation. Cell. 1999
Aug 20;98(4):437451. PubMed PMID: 10481909.
158. Thannickal TC, Moore RY, Nienhuis R, et al. Reduced number of
hypocretin neurons in human narcolepsy. Neuron. 2000 Sep;27
(3):469474. PubMed PMID: 11055430.
159. Nishino S, Ripley B, Overeem S, et al. Hypocretin (orexin) deficiency
in human narcolepsy. Lancet. 2000 Jan 1;355(9197):3940. .
160. Black SW, Yamanaka A, Kilduff TS. Challenges in the development
of therapeutics for narcolepsy. Prog Neurobiol. 2017 May;152:89
113. PubMed PMID: 26721620.
161. Trace Amine-Associated Receptor 1 Modulates the Locomotor and
Sensitization Effects of Nicotine. Sukhanov I, Dorofeikova M,
Dolgorukova A, Dorotenko A, Gainetdinov RR. Front Pharmacol.
2018 Apr 6;9:329. doi: 10.3389/fphar.2018.00329. eCollection
2018. PMID: 29681856
162. Hara J, Beuckmann CT, Nambu T, et al. Genetic ablation of
orexin neurons in mice results in narcolepsy, hypophagia, and
obesity. Neuron. 2001 May;30(2):345354. PubMed PMID:
11394998.
163. Tabuchi S, Tsunematsu T, Black SW, et al. Conditional ablation of
orexin/hypocretin neurons: a new mouse model for the study of
narcolepsy and orexin system function. J Neurosci. 2014 May 07;34
(19):64956509. PubMed PMID: 24806676; PubMed Central PMCID:
PMCPMC4012309.
164. Burgess CR, Peever JH. A noradrenergic mechanism functions to
couple motor behavior with arousal state. Curr Biol. 2013;23
(18):17191725. PubMed PMID: 23993842.
165. Hasegawa E, Maejima T, Yoshida T, et al. Serotonin neurons in the
dorsal raphe mediate the anticataplectic action of orexin neurons
by reducing amygdala activity. Proc Natl Acad Sci U S A. 2017 Apr
25;114(17):E3526E3535. PubMed PMID: 28396432; PubMed
Central PMCID: PMCPMC5410844.
166. Hasegawa E, Yanagisawa M, Sakurai T, et al. Orexin neurons sup-
press narcolepsy via 2 distinct efferent pathways. J Clin Invest. 2014
Feb;124(2):604616. PubMed PMID: 24382351; PubMed Central
PMCID: PMCPMC3904620.
167. Burgess CR, Tse G, Gillis L, et al. Dopaminergic regulation of sleep
and cataplexy in a murine model of narcolepsy. Sleep. 2010 Oct;33
(10):12951304. PubMed PMID: 21061851; PubMed Central PMCID:
PMCPMC2941415.
168. Dinter J, Muhlhaus J, Wienchol CL, et al. Inverse agonistic action of
3-iodothyronamine at the human trace amine-associated receptor
5. PLoS One. 2015;10(2):e0117774. PubMed PMID: 25706283;
PubMed Central PMCID: PMCPMC4382497.
169. Lynch LJ, Sullivan KA, Vallender EJ, et al. Trace amine associated
receptor 1 modulates behavioral effects of ethanol. Subst Abuse.
2013;7:117126. PubMed PMID: 23861588; PubMed Central PMCID:
PMCPMC3682756.
170. Raab S, Wang H, Uhles S, et al. Incretin-like effects of small mole-
cule trace amine-associated receptor 1 agonists. Mol Metab. 2016
Jan;5(1):4756. PubMed PMID: 26844206; PubMed Central PMCID:
PMCPMC4703809.
171. Wulff K, Gatti S, Wettstein JG, et al. Sleep and circadian rhythm
disruption in psychiatric and neurodegenerative disease. Nat Rev
Neurosci. 2018 Apr;6(9):329. DOI: 10.3389/fphar.2018.00329.
eCollection 2018. PMID: 29681856
526 M. D. SCHWARTZ ET AL.
... Since trace amines have been shown to alter the release and/or response to dopamine, norepinephrine, acetylcholine, and GABA [210], they are thought to be potential neuromodulators. When TAAR1 and D2R interact, a reduction in β-arrestin 2 recruitment is detected, silencing the GSK3 cascade via Akt [211,212]. Preclinical research suggests that TAAR1 agonists may improve not only the behaviors proxy of positive symptoms but the one mimicking negative and cognitive symptoms too, without causing motor disorders or weight gain [213]. In order to be considered a multimodal therapeutic target for neuropsychiatric diseases, the role of TAAR1 as a critical node in the regulation of dopaminergic signaling has been established through a combination of experimental preclinical and translational studies [212] (Figure 3). ...
... Preclinical research suggests that TAAR1 agonists may improve not only the behaviors proxy of positive symptoms but the one mimicking negative and cognitive symptoms too, without causing motor disorders or weight gain [213]. In order to be considered a multimodal therapeutic target for neuropsychiatric diseases, the role of TAAR1 as a critical node in the regulation of dopaminergic signaling has been established through a combination of experimental preclinical and translational studies [212] (Figure 3). More attention will be paid to dopamine, glutamate, and serotonin regulation by TAAR1 as a non-canonical mechanism of antipsychotic efficacy [212]. ...
... In order to be considered a multimodal therapeutic target for neuropsychiatric diseases, the role of TAAR1 as a critical node in the regulation of dopaminergic signaling has been established through a combination of experimental preclinical and translational studies [212] (Figure 3). More attention will be paid to dopamine, glutamate, and serotonin regulation by TAAR1 as a non-canonical mechanism of antipsychotic efficacy [212]. SEP-363856, a novel therapy approved by the Food and Drug Administration for the treatment of schizophrenia, is a TAAR1 agonist in phase III clinical development. ...
Article
Full-text available
Schizophrenia is a severe psychiatric illness affecting almost 25 million people worldwide and is conceptualized as a disorder of synaptic plasticity and brain connectivity. Antipsychotics are the primary pharmacological treatment after more than sixty years after their introduction in therapy. Two findings hold true for all presently available antipsychotics. First, all antipsychotics occupy the dopamine D2 receptor (D2R) as an antagonist or partial agonist, even if with different affinity; second, D2R occupancy is the necessary and probably the sufficient mechanism for antipsychotic effect despite the complexity of antipsychotics’ receptor profile. D2R occupancy is followed by coincident or divergent intracellular mechanisms, implying the contribution of cAMP regulation, β-arrestin recruitment, and phospholipase A activation, to quote some of the mechanisms considered canonical. However, in recent years, novel mechanisms related to dopamine function beyond or together with D2R occupancy have emerged. Among these potentially non-canonical mechanisms, the role of Na2+ channels at the dopamine at the presynaptic site, dopamine transporter (DAT) involvement as the main regulator of dopamine concentration at synaptic clefts, and the putative role of antipsychotics as chaperones for intracellular D2R sequestration, should be included. These mechanisms expand the fundamental role of dopamine in schizophrenia therapy and may have relevance to considering putatively new strategies for treatment-resistant schizophrenia (TRS), an extremely severe condition epidemiologically relevant and affecting almost 30% of schizophrenia patients. Here, we performed a critical evaluation of the role of antipsychotics in synaptic plasticity, focusing on their canonical and non-canonical mechanisms of action relevant to the treatment of schizophrenia and their subsequent implication for the pathophysiology and potential therapy of TRS.
... Collectively, all these systems play important roles in mood, cognition, emotion, reward, learning, attention, and motor activity [23], and their dysregulation is associated with a variety of neurological and neurodegenerative disorders, several of which are among the leading causes of death and disability worldwide [24]. Recent studies revealed that pharmacologically targeting TAAR1 within the CNS has resulted in successful clinical trials for the treatment of schizophrenia, depression, addiction, and NDDs [25][26][27][28]. More recently, TAAR1 expression and functionality in immune system regulation and immune cell activation has become a topic of emerging interest [29]; nevertheless, so far, few studies have examined the role of TAAR1 in CNS-resident neuroimmune cells [30,31]. ...
... initially assessed the effects of β-amyloid peptide[25][26][27][28][29][30][31][32][33][34][35] [50] on HMC cells' viability. The cells were treated with two different concentrations (1 and 10 µM) o Aβ25-35 for 24 or 48 h. ...
Article
Full-text available
Microglial dysfunction is one of the hallmarks and leading causes of common neurodegenerative diseases (NDDs), including Alzheimer’s disease (AD) and Parkinson’s disease (PD). All these pathologies are characterized by aberrant aggregation of disease-causing proteins in the brain, which can directly activate microglia, trigger microglia-mediated neuroinflammation, and increase oxidative stress. Inhibition of glial activation may represent a therapeutic target to alleviate neurodegeneration. Recently, 3-iodothyronamine (T1AM), an endogenous derivative of thyroid hormone (TH) able to interact directly with a specific GPCR known as trace amine-associated receptor 1 (TAAR1), gained interest for its ability to promote neuroprotection in several models. Nevertheless, T1AM’s effects on microglial disfunction remain still elusive. In the present work we investigated whether T1AM could inhibit the inflammatory response of human HMC3 microglial cells to LPS/TNFα or β-amyloid peptide 25–35 (Aβ25–35) stimuli. The results of ELISA and qPCR assays revealed that T1AM was able to reduce microglia-mediated inflammatory response by inhibiting the release of proinflammatory factors, including IL-6, TNFα, NF-kB, MCP1, and MIP1, while promoting the release of anti-inflammatory mediators, such as IL-10. Notably, T1AM anti-inflammatory action in HMC3 cells turned out to be a TAAR1-mediated response, further increasing the relevance of the T1AM/TAAR1 system in the management of NDDs.
... Starting with RO5256390, new partial or full TAAR1 agonists with high affinity and selectivity for TAAR1 have been created and studied [78]. Initially, TAAR1 agonists were considered for the treatment of schizophrenia [79,80]. Researchers have already shown their effectiveness against the positive, negative (lack of motivation, anhedonia), and cognitive symptoms of schizophrenia, which are often overlooked by typical antipsychotics [64]. ...
Article
Full-text available
Depression is a common mental illness of great concern. Current therapy for depression is only suitable for 80% of patients and is often associated with unwanted side effects. In this regard, the search for and development of new antidepressant agents remains an urgent task. In this review, we discuss the current available evidence indicating that G protein-coupled trace amine-associated receptors (TAARs) might represent new targets for depression treatment. The most frequently studied receptor TAAR1 has already been investigated in the treatment of schizophrenia, demonstrating antidepressant and anxiolytic properties. In fact, the TAAR1 agonist Ulotaront is currently undergoing phase 2/3 clinical trials testing its safety and efficacy in the treatment of major depressive disorder and generalized anxiety disorder. Other members of the TAAR family (TAAR2, TAAR5, TAAR6, TAAR8, and TAAR9) are not only involved in the innate olfaction of volatile amines, but are also expressed in the limbic brain areas. Furthermore, animal studies have shown that TAAR2 and TAAR5 regulate emotional behaviors and thus may hold promise as potential antidepressant targets. Of particular interest is their connection with the dopamine and serotonin systems of the brain and their involvement in the regulation of adult neurogenesis, known to be affected by the antidepressant drugs currently in use. Further non-clinical and clinical studies are necessary to validate TAAR1 (and potentially other TAARs) as novel therapeutic targets for the treatment of depression.
... For example, TA may also indirectly cause increased susceptibility to oxidative stress in tissue through their modulation of 5-HT. The absence of TAAR1 in model animals is associated with increased DA and 5-HT signaling [131,132]. TAAR5-KO mice also showed a deregulated 5-HT and DA system [5,115]. Since it was suggested that TAAR1 could act as a rheostat of 5-HT signaling [1,131], the relation between TAARs and 5-HT signaling components is expected. ...
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.
... Thus, these data suggest that the development of TAAR1-based drugs could provide a novel therapeutic approach for the treatment of neuropsychiatric disorders related to aberrant frontostriatal circuitry. Based on the preclinical studies, it might be expected that TAAR1 could be a potential drug target for several neuropsychiatric disorders, including schizophrenia, depression, bipolar disorder, generalized anxiety disorder, addiction, ADHD, Alzheimer's disorder, etc. [29,[37][38][39][40][41][42][43][44][45][46][47][48][49][50][51][52]. ...
Article
Full-text available
All antipsychotics currently used in clinic block D2 dopamine receptors. Trace amine-associated receptor 1 is emerging as a new therapeutic target for schizophrenia and several other neuropsychiatric disorders. SEP-363856 (International Nonproprietary Name: Ulotaront) is an investigational antipsychotic drug with a novel mechanism of action that does not involve antagonism of dopamine D2 receptors. Ulotaront is an agonist of trace amine-associated receptor 1 and serotonin 5-HT1A receptors, but can modulate dopamine neurotransmission indirectly. In 2019, the United States Food and Drug Administration granted Breakthrough Therapy Designation for ulotaront for the treatment of schizophrenia. Phase 2 clinical studies indicated that ulotaront can reduce both positive and negative symptoms of schizophrenia without causing the extrapyramidal or metabolic side effects that are inherent to most currently used antipsychotics. At present, it is in phase 3 clinical development for the treatment of schizophrenia and is expected to be introduced into clinical practice in 2023–2024. Clinical studies evaluating the potential efficacy of ulotaront in Parkinson’s disease psychosis, generalized anxiety disorder, and major depressive disorder have also been started. The aim of this scoping review is to summarize all currently available preclinical and clinical evidence on the utility of ulotaront in the treatment of schizophrenia. Here, we show the main characteristics and distinctive features of this drug. Perspectives and limitations on the potential use of ulotaront in the pharmacotherapy of several other neuropsychiatric disorders are also discussed.
... Additionally, the TAAR-1 receptor seems to respond to dopamine and glutamine far more than tryptamine, norepinephrine, and serotonin (Krogmann et al., 2019). These characteristics have led to the targeting of TAAR-1 agonists for the treatment of schizophrenia (Revel et al., 2013;Schwartz et al., 2018). ...
Article
Full-text available
Pharmacological treatment for schizophrenia has been a long-standing concern. As a severe neuropsychological illness, schizophrenia is always a challenging disorder to unravel its pathophysiology. Since it exhibits both positive and negative symptoms, such as hallucination and delusion, as well as social isolation and cognitive impairment, following the symptomatic changes is a crucial task for clinicians. Although various pharmacological treatments are available in the form of antipsychotics, however, their actual consequences need to be examined with the observable changes in symptoms as well as the unobservable changes in brain functioning. This study is a first of its kind to critically investigate both clinical and neuroimaging studies to find out the changes being observed in schizophrenia patients after clinical intervention with various antipsychotics. We observed several symptomatic changes being reported in clinical studies incorporating clinical trials of various first-generation and second-generation antipsychotic drugs. Alongside, we encapsulated several neuroimaging studies showing functional and structural changes in the brain of schizophrenia patients triggered by a variety of drugs. The basal ganglia, frontal lobe, temporal lobe, cuneus, and middle occipital gyrus are some of the notable brain regions that were observed to show subtle functional and structural changes. This critical review paper may pave the way for future research into the study of the pathological and morphological changes in the brains of schizophrenia patients as they progress through the course of medicinal therapy.
Article
Full-text available
The domain of psychiatric drug development is currently witnessing a notable transformation, with a paramount emphasis on targeting nonmonoamine receptors and exploring inventive mechanisms of action. This paper presents an overview of the ongoing advancements in antipsychotic and antidepressant drug development. Historically, antipsychotics predominantly targeted dopamine receptors, but there is now an escalating interest in drugs that act on alternative receptors, exemplified by the TAAR1 receptor. One noteworthy candidate is Ulotaront (SEP‐363856), an agent acting as a TAAR1 agonist with 5‐HT1A agonist activity, demonstrating promising outcomes in the treatment of schizophrenia, devoid of extrapyramidal symptoms or metabolic side‐effects. Similarly, MIN‐101 (Roluperidone) and KarXT are currently in development, with its focus on addressing the symptoms in schizophrenia. In the domain of antidepressants, novel therapeutic approaches have surfaced, such as Auvelity, a Food and Drug Administration (FDA)‐approved NMDA receptor antagonist synergistically combined with Bupropion to enhance its effects. Another notable candidate is Zuranolone, operating as a GABA A receptor‐positive allosteric modulator, showcasing efficacy in treating major depressive disorder (MDD) and postpartum depression. Additionally, TAK‐653 (NBI‐1065845) and MJI821 (Onfasprodil) have emerged as potential antidepressants targeting AMPA receptors and NMDA receptor 2B (NR2B) negative allosteric modulation, respectively. This paper underscores the transformative potential of these novel drug candidates in psychiatric treatment and their ability to address cases that were previously treatment‐resistant. By focusing on nonmonoamine receptors and introducing innovative mechanisms, these drugs offer a promising prospect of improved outcomes for individuals suffering from schizophrenia and MDD. Thus, sustained attention and dedication to the development of such drugs are essential to augmenting the therapeutic options available for psychiatric patients.
Article
Trace amine-associated receptor 1 (TAAR1) senses a spectrum of endogenous amine-containing metabolites (EAMs) to mediate diverse psychological functions and is useful for schizophrenia treatment without the side effects of catalepsy. Here, we systematically profiled the signaling properties of TAAR1 activation and present nine structures of TAAR1-Gs/Gq in complex with EAMs, clinical drugs, and synthetic compounds. These structures not only revealed the primary amine recognition pocket (PARP) harboring the conserved acidic D3.32 for conserved amine recognition and “twin” toggle switch for receptor activation but also elucidated that targeting specific residues in the second binding pocket (SBP) allowed modulation of signaling preference. In addition to traditional drug-induced Gs signaling, Gq activation by EAM or synthetic compounds is beneficial to schizophrenia treatment. Our results provided a structural and signaling framework for molecular recognition by TAAR1, which afforded structural templates and signal clues for TAAR1-targeted candidate compounds design.
Article
Introdução: Este artigo discute o novo tratamento para esquizofrenia baseado em TAAR1, a esquizofrenia é um transtorno mental que possui bastante complexidade devido às suas características da sintomatologia. Os sintomas são classificados em positivos, negativos e cognitivos. A farmacoterapia utilizada no tratamento da esquizofrenia consiste no uso de antipsicóticos. O mecanismo de ação é constituído pelo antagonismo dos receptores dopaminérgicos D2. O TAAR1 é um receptor de Amina traco-1 que são substâncias endógenas que são sintetizadas e degradadas na microbiota e podem ser encontradas na alimentação e possui relação com modulação de transmissão dopaminérgica, glutamatérgica e serotoninérgica. Objetivo: Tem como objetivo analisar a diferença do tratamento baseado em TAAR1 dos tratamentos já disponíveis atualmente. Materiais e Métodos: Para execução dessa pesquisa foram adotados como fonte de coleta de dados, pesquisa bibliográfica por meio de livros-online, dissertações publicações em revistas científicas, protocolos e manuais clínicos. Foram utilizados artigos publicados nas fontes de dados, PUBMED, SCIELO, Google Acadêmico, BASE, BIREME, CAPES. Para realização desta pesquisa foi necessário mais de dez artigo sendo excluídos com período acima de cinco anos. Conclusão: Os TAAR1 diferentemente dos antipsicóticos tradicionais não irão apresentar efeitos colaterais como extrapiramidais ou síndrome metabólica e não atuam no mecanismo de bloqueio de receptores de dopamina D2.
Article
Drugs that stimulate the trace amine-associated receptor 1 (TAAR1) are under clinical investigation as treatments for several neuropsychiatric disorders. Previous studies in a genetic mouse model of voluntary methamphetamine intake identified TAAR1, expressed by the Taar1 gene, as a critical mediator of aversive methamphetamine effects. Methamphetamine is a TAAR1 agonist, but also has actions at monoamine transporters. Whether exclusive activation of TAAR1 has aversive effects was not known at the time we conducted our studies. Mice were tested for aversive effects of the selective TAAR1 agonist, RO5256390, using taste and place conditioning procedures. Hypothermic and locomotor effects were also examined, based on prior evidence of TAAR1 mediation. Male and female mice of several genetic models were used, including lines selectively bred for high and low methamphetamine drinking, a knock-in line in which a mutant form of Taar1 that codes for a non-functional TAAR1 was replaced by the reference Taar1 allele that codes for functional TAAR1, and their matched control line. RO5256390 had robust aversive, hypothermic and locomotor suppressing effects that were found only in mice with functional TAAR1. Knock-in of the reference Taar1 allele rescued these phenotypes in a genetic model that normally lacks TAAR1 function. Our study provides important data on TAAR1 function in aversive, locomotor, and thermoregulatory effects that are important to consider when developing TAAR1 agonists as therapeutic drugs. Because other drugs can have similar consequences, potential additive effects should be carefully considered as these treatment agents are being developed.
Article
Full-text available
Trace amine-associated receptor 1 (TAAR1) has emerged as a promising target for addiction treatments because it affects dopamine transmission in the mesolimbic pathway. TAAR1 is involved in the effects of addictive drugs, such as amphetamines, cocaine and ethanol, but the impact of TAAR1 on the effects of nicotine, the psychoactive drug responsible for the development and maintenance of tobacco smoking, has not yet been studied. This study was performed to investigate the possible modulatory action of TAAR1 on the effects of nicotine on locomotor behaviors in rats and mice. Pretreatment with the TAAR1 agonist RO5263397 dose-dependently decreased nicotine-induced hyperlocomotion in rats habituated to locomotor boxes, prevented the development of nicotine sensitization and blocked hypermotility in nicotine-sensitized rats at the highest tested dose (10 mg/kg). The lack of TAAR1 failed to affect the effects of nicotine on the locomotion of mutant mice. Based on the results of the present study, TAAR1 activation attenuates the locomotion-stimulating effects of nicotine on rats. These results further support the previously proposed hypothesis that TAAR1 is a promising target for the prevention and treatment of drug addiction. Further studies aimed at analyzing the effects of TAAR1 agonists on animal models of nicotine addiction are warranted.
Article
Full-text available
Trace amine-associated receptors (TAARs) belong to the class A G-protein-coupled receptors (GPCR) and are evolutionary related to aminergic receptors. TAARs have been identified to mediate effects of trace amines. TAAR1 signaling is mainly mediated via activation of the Gs/adenylyl cyclase pathway. In addition to classical trace amines, TAAR1 can also be activated by the thyroid hormone derivative 3-iodothyronamine (3-T1AM). Pharmacological doses of 3-T1AM induced metabolic and anapyrexic effects, which might be centrally mediated in the hypothalamus in rodents. However, the observed anapyrexic effect of 3-T1AM persists in Taar1 knock-out mice which raises the question whether further GPCRs are potential targets for 3-T1AM and mediate the observed physiological effect. Anapyrexia has been observed to be related to action on aminergic receptors such as the serotonin receptor 1b (5-HT1b). This receptor primarily activates the Gi/o mediated pathway and PLC signaling through the Gβγ of Gi/o. Since the expression profiles of TAAR1 and 5-HT1b overlap, we questioned whether 3-T1AM may activate 5-HT1b. Finally, we also evaluated heteromerization between these two GPCRs and tested signaling under co-expressed conditions. In this study, we showed, that 3-T1AM can induce Gi/o signaling through 5-HT1b in a concentration of 10 μM. Strikingly, at 5-HT1b the ligand 3-T1AM only activates the Gi/o mediated reduction of cAMP accumulation, but not PLC activation. Co-stimulation of 5-HT1b by both ligands did not lead to additive or synergistic signaling effects. In addition, we confirmed the capacity for heteromerization between TAAR1 and 5-HT1b. Under co-expression of TAAR1 and HTR1b, 3-T1AM action is only mediated via TAAR1 and activation of 5-HT1b is abrogated. In conclusion, we found evidence for 5-HT1b as a new receptor target for 3-T1AM, albeit with a different signaling effect than the endogenous ligand. Altogether, this indicates a complex interrelation of signaling effects between the investigated GPCRs and respective ligands.
Article
Full-text available
Alterations in dopamine neurotransmission are generally associated with diseases such as attention-deficit/hyperactivity disorder (ADHD) and obsessive-compulsive disorder (OCD). Such diseases typically feature poor decision making and lack of control on executive functions and have been studied through the years using many animal models. Dopamine transporter (DAT) knockout (KO) and heterozygous (HET) mice, in particular, have been widely used to study ADHD. Recently, a strain of DAT KO rats has been developed (1). Here, we provide a phenotypic characterization of reward sensitivity and compulsive choice by adult rats born from DAT–HET dams bred with DAT–HET males, in order to further validate DAT KO rats as an animal model for preclinical research. We first tested DAT KO rats’ sensitivity to rewarding stimuli, provided by highly appetitive food or sweet water; then, we tested their choice behavior with an Intolerance-to-Delay Task (IDT). During these tests, DAT KO rats appeared less sensitive to rewarding stimuli than wild-type (WT) and HET rats: they also showed a prominent hyperactive behavior with a rigid choice pattern and a wide number of compulsive stereotypies. Moreover, during the IDT, we tested the effects of amphetamine (AMPH) and RO-5203648, a trace amine-associated receptor 1 (TAAR1) partial agonist. AMPH accentuated impulsive behaviors in WT and HET rats, while it had no effect in DAT KO rats. Finally, we measured the levels of tyrosine hydroxylase, dopamine receptor 2 (D2), serotonin transporter, and TAAR1 mRNA transcripts in samples of ventral striatum, finding no significant differences between WT and KO genotypes. Throughout this study, DAT KO rats showed alterations in decision-making processes and in motivational states, as well as prominent motor and oral stereotypies: more studies are warranted to fully characterize and efficiently use them in preclinical research.
Article
Full-text available
Trace amines (TAs), endogenous amino acid metabolites that are structurally similar to the biogenic amines, are endogenous ligands for trace amine-associated receptor 1 (TAAR1), a GPCR that modulates dopaminergic, serotonergic, and glutamatergic activity. Selective TAAR1 full and partial agonists exhibit similar pro-cognitive, antidepressant- and antipsychotic-like properties in rodents and non-human primates, suggesting TAAR1 as a novel target for the treatment of neurological and psychiatric disorders. We previously reported that TAAR1 partial agonists are wake-promoting in rats and mice, and that TAAR1 knockout (KO) and overexpressing mice exhibit altered sleep-wake and EEG spectral composition. Here, we report that locomotor and EEG spectral responses to the psychostimulants modafinil and caffeine are attenuated in TAAR1 KO mice. TAAR1 KO mice and WT littermates were instrumented for EEG and EMG recording and implanted with telemetry transmitters for monitoring locomotor activity (LMA) and core body temperature (Tb). Following recovery, mice were administered modafinil (25, 50, 100 mg/kg), caffeine (2.5, 10, 20 mg/kg) or vehicle p.o. at ZT6 in balanced order. In WT mice, both modafinil and caffeine dose-dependently increased LMA for up to 6 h following dosing, whereas only the highest dose of each drug increased LMA in KO mice, and did so for less time after dosing. This effect was particularly pronounced following caffeine, such that total LMA response was significantly attenuated in KO mice compared to WT at all doses of caffeine and did not differ from Vehicle treatment. Tb increased comparably in both genotypes in a dose-dependent manner. TAAR1 deletion was associated with reduced wake consolidation following both drugs, but total time in wakefulness did not differ between KO and WT mice. Furthermore, gamma band EEG activity following both modafinil and caffeine treatment was attenuated in TAAR1 KO compared to WT mice. Our results show that TAAR1 is a critical component of the behavioral and cortical arousal associated with two widely used psychostimulants with very different mechanisms of action. Together with our previous findings, these data suggest that TAAR1 is a previously unrecognized component of an endogenous wake-modulating system.
Article
Full-text available
Dopamine (DA) controls many vital physiological functions and is critically involved in several neuropsychiatric disorders such as schizophrenia and attention deficit hyperactivity disorder. The major function of the plasma membrane dopamine transporter (DAT) is the rapid uptake of released DA into presynaptic nerve terminals leading to control of both the extracellular levels of DA and the intracellular stores of DA. Here, we present a newly developed strain of rats in which the gene encodingDATknockout Rats (DAT-KO) has been disrupted by using zinc finger nuclease technology. Male and female DAT-KO rats develop normally but weigh less than heterozygote and wild-type rats and demonstrate pronounced spontaneous locomotor hyperactivity. While striatal extracellular DA lifetime and concentrations are significantly increased, the total tissue content of DA is markedly decreased demonstrating the key role of DAT in the control of DA neurotransmission. Hyperactivity of DAT-KO rats can be counteracted by amphetamine, methylphenidate, the partial Trace Amine-Associated Receptor 1 (TAAR1) agonist RO5203648 ((S)-4-(3,4-Dichloro-phenyl)-4,5-dihydro-oxazol-2-ylamine) and haloperidol. DAT-KO rats also demonstrate a deficit in working memory and sensorimotor gating tests, less propensity to develop obsessive behaviors and show strong dysregulation in frontostriatal BDNF function. DAT-KO rats could provide a novel translational model for human diseases involving aberrant DA function and/or mutations affecting DAT or related regulatory mechanisms.
Article
Full-text available
Recent evidence suggests that the trace amine-associated receptor 1 (TAAR1) plays a pivotal role in the regulation of dopamine (DA) transmission and cocaine’s actions. However, the underlying mechanisms through which TAAR1 activation mediates these effects have not yet been elucidated. Here, we used fast-scan cyclic voltammetry to measure DA dynamics and explore such mechanisms. We show, first, that the full TAAR1 agonist, RO5256390, dose-dependently blocked cocaine-induced inhibition of DA clearance in slices of the nucleus accumbens. Second, subthreshold inhibition of PKA or PKC phosphorylation did not prevent TAAR1 suppression of cocaine effects whereas subeffective doses of the DA D2 receptor antagonist, L-741,626, rescued cocaine’s ability to produce changes in DA uptake in the presence of full TAAR1 activation, thus indicating that TAAR1 modulation of cocaine effects requires simultaneous DA D2 receptor activation. Predictably, inhibition of glycogen synthase kinase-3 (GSK-3), which results from activation of D2/TAAR1 heterodimers, fully reproduced the inhibitory effects of TAAR1 activation on cocaine-induced changes in DA transmission. Collectively, the present observations reveal that the ability of TAAR1 to regulate cocaine effects is linked to cooperative interactions with D2 autoreceptors and associated downstream molecular targets converging on GSK-3 and suggest a new mechanism to disrupt cocaine neurochemical actions.
Article
Full-text available
The discovery in 2001 of a G protein-coupled receptor family, subsequently termed trace amine-associated receptors (TAAR), triggered a resurgence of interest in so-called trace amines. Initial optimism quickly faded, however, as the TAAR family presented a series of challenges preventing the use of standard medicinal chemistry and pharmacology technologies. Consequently the development of basic tools for probing TAAR and translating findings from model systems to humans has been problematic. Despite these challenges the last 5 years have seen considerable advances, in particular with respect to TAAR1, which appears to function as an endogenous rheostat, maintaining central neurotransmission within defined physiological limits, in part through receptor heterodimerization yielding biased signaling outputs. Regulation of the dopaminergic system is particularly well understood and clinical testing of TAAR1 directed ligands for schizophrenia and psychiatric disorders have begun. In addition, pre-clinical animal models have identified TAAR1 as a novel target for drug addiction and metabolic disorders. Growing evidence also suggests a role for TAARs in regulating immune function. This review critically discusses the current state of TAAR research, highlighting recent developments and focussing on human TAARs, their functions, and clinical implications. Current gaps in knowledge are identified, along with the research reagents and translational tools still required for continued advancement of the field. Through this, a picture emerges of an exciting field on the cusp of significant developments, with the potential to identify new therapeutic leads for some of the major unmet medical needs in the areas of neuropsychiatry and metabolic disorders.
Article
Nicotine addiction and abuse remains a global health issue. To date, the fundamental neurobiological mechanism of nicotine addiction remains incompletely understood. Trace amine-associated receptor 1 (TAAR1) is thought to directly modulate dopaminergic system and are thought to be a neural substrate underlying addictive-like behaviors. We aimed to investigate the role of TAAR1 in nicotine addictive-like behaviors. TAAR1 expression after nicotine treatment was evaluated by western blotting. c-Fos immunofluorescence and in vivo fast-scan cyclic voltammetry were used to examine the activation of brain regions and dopamine release, respectively. We then thoroughly and systematically examined the role of TAAR1 in mediating nicotine-induced sensitization, nicotine discrimination, nicotine self-administration, nicotine demand curve, and the reinstatement of nicotine-seeking. Local pharmacological manipulation was conducted to determine the role of TAAR1 in the nucleus accumbens (NAcs) in the reinstatement of nicotine-seeking. We found that the expression of TAAR1 protein was selectively downregulated in the NAc, with no change in either dorsal striatum or prefrontal cortex. TAAR1 activation was sufficient to block nicotine-induced c-Fos expression in the NAc, while also reducing nicotine-induced dopamine release in the NAc. Systemic administration of TAAR1 agonists attenuated the expression and development of nicotine-induced sensitization, nicotine self-administration, the reinstatement of nicotine-seeking, and increased the elasticity of nicotine demand curve, while intra-NAc infusions of a TAAR1 agonist was sufficient to attenuate nicotine reinstatement. Moreover, TAAR1-knockout rats showed augmented cue-induced and drug-induced reinstatement of nicotine-seeking. These results indicated that modulation of TAAR1 activity regulates nicotine addictive-like behaviors and TAAR1 represents a novel target towards the treatment of nicotine addiction.
Article
Impulsivity is an important personality trait associated with several clinical syndromes including drug abuse. While repeated drug exposure is known to increase certain behavioral responses, such as locomotion, to subsequent drug exposure, few studies have examined whether such sensitization develops for impulsive behavior. In the current study we tested the effects of methamphetamine acutely, during the course of, and upon discontinuation of chronic methamphetamine treatment on impulsive behavior in two models, the 5-choice serial reaction time task (5-CSRTT) and the delay-discounting task which measure impulsive action and impulsive choice, respectively. We also examined whether the trace amine-associated receptor 1 (TAAR1) agonist RO5263397 attenuated methamphetamine-induced effects in parallel tests. Acute methamphetamine dose-dependently increased premature responses in the 5-CSRTT and shifted the delay function upward in delay discounting. Up to 40 days of methamphetamine treatment did not significantly alter the dose-effect curve of methamphetamine-induced premature responses, but produced a significant effect in the delay-discounting task. RO5263397 attenuated acute methamphetamine-induced premature responses, but this effect became non-significant over the course of chronic treatment. RO5263397 did not significantly alter the delay-discounting performance. Discontinuation of methamphetamine treatment increased premature responses, which was attenuated by RO5263397, but did not significantly alter the delay discounting function. These results suggest that acute discontinuation from prolonged methamphetamine treatment increases impulsivity, which can be reduced by a TAAR1 agonist.