Elsevier

Biological Psychiatry

Volume 82, Issue 9, 1 November 2017, Pages 623-633
Biological Psychiatry

Priority Communication
Trace Amine-Associated Receptor 1 Agonists as Narcolepsy Therapeutics

https://doi.org/10.1016/j.biopsych.2016.10.012Get rights and content

Abstract

Background

Narcolepsy, a disorder of rapid eye movement (REM) sleep, is characterized by excessive daytime sleepiness and cataplexy, a loss of muscle tone triggered by emotional stimulation. Current narcolepsy pharmacotherapeutics include controlled substances with abuse potential or drugs with undesirable side effects. As partial agonists at trace amine-associated receptor 1 (TAAR1) promote wakefulness in mice and rats, we evaluated whether TAAR1 agonism had beneficial effects in two mouse models of narcolepsy.

Methods

In the first experiment, male homozygous B6-Taar1tm1(NLSLacZ)Blt (Taar1 knockout) and wild-type mice were surgically implanted to record electroencephalogram, electromyogram, locomotor activity, and body temperature, and the efficacy of the TAAR1 agonist, RO5256390, on sleep/wake and physiological parameters was determined. In the second experiment, the effects of the TAAR1 full agonist RO5256390 and partial agonist RO5263397 on sleep/wake, locomotor activity, body temperature, and cataplexy were assessed in two mouse narcolepsy models.

Results

RO5256390 profoundly reduced rapid eye movement sleep in wild-type mice; these effects were eliminated in Taar1 knockout mice. The TAAR1 partial agonist RO5263397 also promoted wakefulness and suppressed nonrapid eye movement sleep. Both compounds reduced body temperature in the two narcolepsy models at the highest doses tested. Both TAAR1 compounds also mitigated cataplexy, the pathognomonic symptom of this disorder, in the narcolepsy models. The therapeutic benefit was mediated through a reduction in number of cataplexy episodes and time spent in cataplexy.

Conclusions

These results suggest TAAR1 agonism as a new therapeutic pathway for treatment of this orphan disease. The common underlying mechanism may be the suppression of rapid eye movement sleep.

Section snippets

Methods and Materials

All experimental procedures were approved by the Institutional Animal Care and Use Committee at SRI International and were conducted in accordance with the principles set forth in the Guide for Care and Use of Laboratory Animals. Detailed methods are described in Supplemental Methods.

Protocol 1: Efficacy of Full and Partial TAAR1 Agonists in Taar1 KO and WT Mice

We previously showed that the TAAR1 partial agonist RO5263397 promoted wakefulness and inhibited REM sleep in mice and that these effects were TAAR1 dependent (38). To determine the generality of this response, we evaluated the efficacy of another TAAR1 agonist, RO5256390, on sleep/wake when administered to WT and Taar1 KO mice in the mid-light phase when mice normally spend most time asleep. Although we have previously characterized RO5256390 as a full agonist in rats and monkeys, its

Discussion

Narcolepsy is a disorder of REM sleep, and cataplexy is its pathognomonic symptom. In the present study, two TAAR1 agonists showed a therapeutic benefit in reduction of cataplexy in two different mouse models of narcolepsy. The TAAR1 partial agonist RO5263397 reduced cataplexy in both mouse narcolepsy models (Figure 5B, D, F) and suppressed REM sleep in the Atax narcolepsy model (Figure 3I, L) as well as in WT mice (Figure 1I). By contrast, RO5256390 had a therapeutic benefit on cataplexy (

Acknowledgments and Disclosures

This work was supported by the National Institutes of Health Grant Nos. R01 NS082876, R21 NS083639 and R21 NS085757 (to TSK).

We thank Professor Akihiro Yamanaka of Nagoya University for the C57BL/6-Tg(orexin/tTA; TetO diphtheria toxin A fragment)/Yamanaka mice. We also thank Drs. A. Harmeier, J.-L. Moreau, and J. Wettstein for valuable scientific input and Jeremiah Palmerston and Kelsie Bogyo for technical assistance.

MCH is an employee of F. Hoffmann-La Roche Ltd. Over the past 2 years, TSK has

References (49)

  • C.R. Burgess et al.

    Narcolepsy: Neural mechanisms of sleepiness and cataplexy

    J Neurosci

    (2012)
  • C. Peyron et al.

    A mutation in a case of early onset narcolepsy and a generalized absence of hypocretin peptides in human narcoleptic brains

    Nat Med

    (2000)
  • T.C. Thannickal et al.

    Reduced number of hypocretin neurons in human narcolepsy

    Neuron

    (2000)
  • S. Nishino et al.

    Hypocretin (orexin) deficiency in human narcolepsy

    Lancet

    (2000)
  • D. Gerashchenko et al.

    Relationship between CSF hypocretin levels and hypocretin neuronal loss

    Exp Neurol

    (2003)
  • S. Zhang et al.

    The development of hypocretin (orexin) deficiency in hypocretin/ataxin-3 transgenic rats

    Neuroscience

    (2007)
  • R.M. Chemelli et al.

    Narcolepsy in orexin knockout mice: Molecular genetics of sleep regulation

    Cell

    (1999)
  • L. Lin et al.

    The sleep disorder canine narcolepsy is caused by a mutation in the hypocretin (orexin) receptor 2 gene

    Cell

    (1999)
  • J.T. Willie et al.

    Distinct narcolepsy syndromes in orexin receptor-2 and orexin null mice: Molecular genetic dissection of Non-REM and REM sleep regulatory processes

    Neuron

    (2003)
  • C.T. Beuckmann et al.

    Expression of a poly-glutamine-ataxin-3 transgene in orexin neurons induces narcolepsy-cataplexy in the rat

    J Neurosci

    (2004)
  • J. Hara et al.

    Genetic ablation of orexin neurons in mice results in narcolepsy, hypophagia, and obesity

    Neuron

    (2001)
  • S. Tabuchi et al.

    Conditional ablation of orexin/hypocretin neurons: A new mouse model for the study of narcolepsy and orexin system function

    J Neurosci

    (2014)
  • C.R. Burgess et al.

    Dopaminergic regulation of sleep and cataplexy in a murine model of narcolepsy

    Sleep

    (2010)
  • C. Guilleminault et al.

    Narcolepsy: Diagnosis and management

  • J.P. Wisor et al.

    Dopaminergic role in stimulant-induced wakefulness

    J Neurosci

    (2001)
  • B.K. Madras et al.

    Modafinil occupies dopamine and norepinephrine transporters in vivo and modulates the transporters and trace amine activity in vitro

    J Pharmacol Exp Ther

    (2006)
  • S. Nishino et al.

    Pharmacological aspects of human and canine narcolepsy

    Prog Neurobiol

    (1997)
  • E. Mignot

    Narcolepsy: Pathophysiology and genetic predisposition

  • O.G. Bosch et al.

    Reconsidering GHB: orphan drug or new model antidepressant?

    J Psychopharmacol

    (2012)
  • R. Boscolo-Berto et al.

    Narcolepsy and effectiveness of gamma-hydroxybutyrate (GHB): A systematic review and meta-analysis of randomized controlled trials

    Sleep Med Rev

    (2012)
  • B. Borowsky et al.

    Trace amines: Identification of a family of mammalian G protein-coupled receptors

    Proc Natl Acad Sci U S A

    (2001)
  • J.R. Bunzow 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)
  • L. Lindemann et al.

    Trace amine-associated receptor 1 modulates dopaminergic activity

    J Pharmacol Exp Ther

    (2008)
  • T.D. Wolinsky et al.

    The trace amine 1 receptor knockout mouse: An animal model with relevance to schizophrenia

    Genes Brain Behav

    (2007)
  • Cited by (36)

    • Trace amine-associated receptor 1 modulates motor hyperactivity, cognition, and anxiety-like behavior in an animal model of ADHD

      2022, Progress in Neuro-Psychopharmacology and Biological Psychiatry
      Citation Excerpt :

      Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor (GPCR) activated by trace amines (β-phenylethylamine, p-tyramine, tryptamine, and p-octopamine), 5-hydroxytryptamine (5-HT) and DA (partial agonists), and by a wide spectrum of psychoactive drugs, including amphetamine and its numerous analogs (Borowsky et al., 2001; Bunzow et al., 2001; Xie et al., 2007). TAAR1 has been shown to play a role in animal models of several neuropsychiatric conditions related to aberrant regulation of brain monoamines, such as substance use disorders (Liu and Li, 2018; Revel et al., 2012b), eating disorders (Ferragud et al., 2017; Moore et al., 2018), narcolepsy (Black et al., 2017), mood disorders (Mantas et al., 2021; Revel et al., 2012b, 2013; Zhang et al., 2021), Parkinson's disease (Alvarsson et al., 2015; Gemechu et al., 2018; Sotnikova et al., 2008), and schizophrenia (Kokkinou et al., 2020; Revel et al., 2012b, 2013). Ulotaront (SEP-363856), an agonist of TAAR1 and 5-HT type 1A receptor, is under clinical evaluation as a novel therapeutic agent for schizophrenia (Dodd et al., 2020).

    • Translational approaches to influence sleep and arousal

      2022, Brain Research Bulletin
      Citation Excerpt :

      Activation of P2 receptors in the basal forebrain promotes wakefulness via excitatory effects on cholinergic and cortically projecting GABAergic neurons (Yang et al., 2018), whereas activation of P2X7 receptors promotes sleep (Krueger et al., 2010), possibly by actions on glia in the cortex, which as discussed in Section 4 are emerging as important regulators of sleep. Trace amine receptors are an interesting novel pharmacological target to treat disorders of sleep and wakefulness (Black et al., 2017). Recently, a partial agonist (R05263397) of trace-amine associated receptor increased wakefulness in macaques without any impairment of cognition (Goonawardena et al., 2019).

    View all citing articles on Scopus
    1

    SWB and MDS contributed equally to this work.

    View full text