Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

DTD 5

ARTICLE IN PRESS

Regulatory Peptides xx (2004) xxx – xxx


www.elsevier.com/locate/regpep

Review

Reverse pharmacology of orexin: from an orphan GPCR


to integrative physiology
Takeshi Sakurai*
Department of Pharmacology, Institute of Basic Medical Science, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
ERATO Yanagisawa Orphan Receptor Project, Japan Science and Technology Corporation, Tokyo 135-0064, Japan

Abstract

Orexins, which were initially identified as endogenous peptide ligands for two orphan G-protein coupled receptors (GPCRs), have been
shown to have an important role in the regulation of energy homeostasis. Furthermore, the discovery of orexin deficiency in narcolepsy
patients indicated that orexins are highly important factors for the sleep/wakefulness regulation. The efferent and afferent systems of orexin-
producing neurons suggest interactions between these cells and arousal centers in the brainstem as well as important feeding centers in the
hypothalamus. Electrophysiological studies have shown that orexin neurons are regulated by humoral factors, including leptin, glucose, and
ghrelin as well as monoamines and acetylcholin. Thus, orexin neurons have functional interactions with hypothalamic feeding pathways and
monoaminergic/cholinergic centers to provide a link between peripheral energy balance and the CNS mechanisms that coordinate sleep/
wakefulness states and motivated behavior such as food seeking.
D 2004 Elsevier B.V. All rights reserved.

Keywords: Hypothalamus; Orexin; Arousal; Sleep; Cholinergic; Monoaminergic

Contents

1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2. Identification of orexins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3. Orexin receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4. Orexin-producing neurons . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5. Orexin defficiency in narcolepsy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
6. Mechanisms of regulation of behavioral states by orexins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
7. Regulation of orexin neuronal activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
8. Roles of orexins in the regulation of energy homeostasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
9. Orexin: a link between energy homeostasis and arousal . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

1. Introduction

bReverse pharmacologyQ, i.e., endogenous ligand screen-


ing using cell lines which express orphan G-protein coupled
receptors (GPCRs), combined with genetic engineering
* Tel.: +81 29 853 3277; fax: +81 298 853 3276. techniques, has increased our understanding of novel
E-mail address: stakeshi@md.tsukuba.ac.jp. signaling systems in the body [1]. Perhaps the most
0167-0115/$ - see front matter D 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.regpep.2004.08.006

REGPEP-03271; No of Pages 8
ARTICLE IN PRESS
2 T. Sakurai / Regulatory Peptides xx (2004) xxx–xxx

successful example of such approach is discovery of orexin. Mammalian orexin-A is a 33-amino-acid peptide with
Many works clearly suggested that orexins are highly two intrachain disulfide bonds, while mammalian orexin-B
important molecules in the regulation of sleep/wakefulness is a 28-amino-acid, C-terminally amidated linear peptide
states as well as feeding behavior. This review summarizes (Fig. 1B). Strong homology between orexin-A and orexin-B
recent relevant findings on orexins, and discusses physio- is found in their C terminal halves. Orexin-A and orexin-B
logical roles of these peptides. are both derived from a common precursor (prepro-orexin),
which is encoded by a gene composed of two exons and an
intervening intron located at 17q21 in human [2,3]. Prepro-
2. Identification of orexins orexin is a 130–131-residue (depending on the species)
polypeptide, which has a typical secretory signal sequence
Most neuropeptides work though G protein–coupled at its N-terminus, and is proteolytically cleaved to form
receptor (GPCRs). There are numerous (approximately mature orexin-A and -B (Fig. 1A). An mRNA encoding the
100–150) borphanQ GPCR genes in the human genome; same precursor peptide was independently isolated by de
the cognate ligands for these receptor molecules have not Lecea et al. as a hypothalamus-specific transcript [4]. They
been identified yet. We performed a so-called breverse predicted that this transcript encodes two neuropeptides,
pharmacologyQ that aims to identify ligands for orphan named hypocretin-1 and -2. The names bhypocretinQ and
GPCRs. We expressed orphan GPCR genes in transfected borexinQ are currently used as synonyms.
cells and used them as a reporter system to detect
endogenous ligand substances in tissue extracts that can
activate signal transduction pathways in GPCR-expressing 3. Orexin receptors
cell lines. In this process, we identified orexin-A and orexin-
B as endogenous ligands for two orphan GPCRs found as Two orexin receptor subtypes, which have 64% amino-
human expressed sequence tags [2]. Orexins constitute a acid identity with each other, named orexin-1 receptor
novel peptide family, with no homology with any previously (OX1R) and orexin-2 receptor (OX2R), have been identified
described peptides. in mammals [2]. We initially identified orexin peptides
using cells expressing OX1R, which has a 1-order-of-
magnitude greater affinity for orexin-A compared with
orexin-B. BLAST search for EST data bases with OX1R
sequence as a query led us identification of another subtype
of orexin receptor, OX2R, to which both orexin-A and
orexin-B bind with similar affinity (Fig. 1A). OX1R is
coupled exclusively to the Gq subclass of heterotrimeric G
proteins, whereas OX2R is coupled to both Gi/o and Gq
when expressed in cell lines [5].
In situ hybridization studies have demonstrated that
orexin receptors are expressed in regions in which dense
orexin immunoreactive fibers are observed (discussed
below). OX1R and OX2R show a markedly different and
complementary distribution [6]. For instance, within the
hypothalamus, a low level of OX1R mRNA expression is
observed in the dorsomedial hypothalamus (DMH), while
high level of OX2R mRNA expression is observed in this
region. Other areas of OX2R expression in the hypothal-
amus include the arcuate nucleus, paraventricular nucleus
(PVN), LHA, and most significantly, the tuberomammillary
nucleus (TMN) [6]. In these regions, there is little or no
OX1R signal. In the hypothalamus, OX1R mRNA is
Fig. 1. (A) Schematic representation of the orexin system. Orexin-A and -B
abundant in the anterior hypothalamic area and ventrome-
are derived from a common precursor peptide, prepro-orexin. The actions
of orexins are mediated via two G protein-coupled receptors named orexin- dial hypothalamus (VMH). Outside the hypothalamus, high
1 (OX1R) and orexin-2 (OX2R) receptors. OX1R is selective for orexin-A, levels of OX1R mRNA expression are detected in the tenia
whereas OX2R is a nonselective receptor for both orexin-A and orexin-B. tecta, hippocampal formation, dorsal raphe nucleus, and
OX1R is coupled exclusively to the Gq subclass of heterotrimeric G most prominently, the locus coeruleus (LC). OX2R mRNA
proteins, whereas OX2R may couple to Gi/o, and/or Gq. (B) Structures of
is abundantly expressed in the cerebral cortex, nucleus
mature orexin-A and -B peptides. The topology of the two intrachain
disulfide bonds in orexin-A is indicated above the sequence. Amino acid accumbens, subthalamic nucleus, paraventricular thalamic
identities are indicated by boxes. *Mammalian orexin-A sequences thus far nuclei, anterior pretectal nucleus, and the raphe nuclei.
identified (human, rat, mouse, pig, dog, sheep, cow) are all identical. Within the brain, OX1R is most abundantly expressed in the
ARTICLE IN PRESS
T. Sakurai / Regulatory Peptides xx (2004) xxx–xxx 3

LC, while OX2R is most abundantly expressed in the TMN, imately 90% of patients with narcolepsy show decreased
regions highly important for maintenance of arousal. orexin-A levels in the cerebrospinal fluid [20]. Narcolepsy
is a common sleep disorder characterized by a primary
disorganization of behavioral states. This disorder affects
4. Orexin-producing neurons approximately 1 in 2000 individuals in the United States.
Most cases of human narcolepsy usually start during
Orexin-producing neurons (orexin neurons) are exclu- adolescence. A cardinal symptom of the disorder is
sively localized to the LHA [7–9]. These cells diffusely excessive daytime sleepiness (an insurmountable urge to
project to the entire neuroaxis, excluding the cerebellum [7– sleep), manifested particularly as attacks of somnolence at
9] (Fig. 2). The densest staining of orexin-immunoreactive inappropriate times. Nocturnal sleep is also sometimes
nerve endings in the brain was found in the arcuate nucleus disturbed by hypnagogic hallucinations, vivid dreaming,
of the hypothalamus, raphe nuclei, TMN and LC. Together and sleep paralysis. Narcolepsy patients often suffer from
with the tissue distribution of both orexin receptors, these cataplexy, which is an attack characterized by sudden
observations suggest that these regions are major effector muscle weakness, which can range from jaw dropping and
sites of orexin. speech slurring to a complete bilateral collapse of the
Melanin-concentrating hormone (MCH) neurons show postural muscles. These attacks of muscle weakness are
very similar localization with orexin neurons in the LHA most often triggered by strong emotional stimuli. Con-
[10]. However, orexin and MCH neurons are distinct and sciousness is preserved during cataplexy. The latency for
independent neuronal populations [11,12]. Orexin does not REM sleep is notably reduced in narcolepsy patients, and
colocalize with cocaine and amphetamine-regulated tran- the presence of sleep-onset REM period is a diagnostic
script (CART) or nitric oxide synthase, either [13]. How- criterion for narcolepsy. It is generally accepted that the
ever, orexin colocalizes with dynorphin [14], galanin [15], symptoms of narcolepsy can be considered as a pathological
and glutamate [16]. Recently, Eriksson et al. [17] reported intrusion of factors of sleep, and especially REM sleep-
that dynorphin suppressed GABAergic input and thus related phenomena, into the state of wakefulness. Thus,
disinhibited histaminergic neurons in the TMN. Therefore, narcolepsy can be viewed as a behavioral state boundary
co-localized dynorphin and orexin might synergistically disorder [21].
activate TMN histaminergic neurons [17]. The clues suggesting the possible involvement of orexin
in narcolepsy initially came from animal models; mice
lacking either the orexin gene (prepro-orexin knockout
5. Orexin defficiency in narcolepsy mice) or orexin neurons (orexin/ataxin-3 transgenic mice),
as well as mice and dogs with null mutations in the OX 2 R
The importance of orexin in the regulation of sleep/ gene, all have phenotypes remarkably similar to narcolepsy
wakefulness is highlighted by the discovery of orexin [22–25] (Table 1). Prepro-orexin knockout mice and orexin/
deficiency in human narcolepsy patients [18,19]. Approx- ataxin-3 mice showed a similar phenotype, which is

Fig. 2. Schematic drawing of para-agittal section through the rat brain, summarizing the organization of the orexin neuronal system. Orexin neurons are found
only in the lateral hypothalamic area and project to the entire central nervous system. Abbreviations: 3V, third ventricle; 4V, fourth ventricle; Amyg, amygdala;
VLPO, ventrolateral preoptic area; SCN, suprachiasmatic nucleus; PVN, paraventricular nucleus; VMH, ventromedial hypothalamus; ARC, arcuate nucleus;
DMH, dorsomedial hypothalamus; LH, lateral hypothalamus; TMN, tuberomamillary nucleus; PPN, pedunculopontine nucleus; LC, locus coeruleus.
ARTICLE IN PRESS
4 T. Sakurai / Regulatory Peptides xx (2004) xxx–xxx

Table 1
Rodent narcolepsy models produced by genetic engineering
Phenotype Reference
Prepro-orexin knockout Cataplexy (+), sleep attack (+) [23]
Sleep/wake fragmentation (severe)
OX1R knockout Cataplexy ( ), sleep attack (+) [43]
Sleep/wake fragmentation (mild)
OX2R knockout Cataplexy ( ), sleep attack (+) [25]
Sleep/wake fragmentation (severe)
Orexin/ataxin-3 mouse Cataplexy (+), sleep attack (+) [24]
Sleep/wake fragmentation (severe)
Orexin/ataxin-3 rat Cataplexy (+), sleep attack (+) [56]
Sleep/wake fragmentation (severe)

characterized by behavioral arrest similar to cataplexy, thalamus [18,19], supporting an earlier report showing
occasional direct transition to REM sleep from wakefulness, undetectable CSF orexin-A peptide levels in most narco-
and highly fragmented behavioral states, resulted from lepsy patients [26]. These results suggest either a loss of
behavioral instability [23,24] (Fig. 3). orexin-containing neurons or a lack of orexin production in
Consistently, a postmortem study in narcoleptic subjects these neurons if still present. One of the predisposing factors
showed undetectable levels of orexin peptides in projection is a specific class II HLA haplotype on human chromosome
sites such as the cortex and pons and an 80–100% reduction 6, with HLA DQB1*0602 and DQA1*0102 alleles, which
in the number of orexin-containing neurons in the hypo- were found in more than 85% of all narcoleptic patients

Fig. 3. Summary of vigilance state parameters recorded from orexin/ataxin-3 hemizygous transgenic and wild-type littermate control (WT) mice. Upper panels:
Total time spent in each state (minutes, meanFS.E.M.), itemized separately for light (left panel) and dark (right panel) periods. Lower pannels: Mean episode
duration of each vigilance state observed in light (left panel) and dark (right panel) periods. Significant differences ( Pb0.05; repeated measurements ANOVA)
between Tg and WT mice are indicated with an asterisk.
ARTICLE IN PRESS
T. Sakurai / Regulatory Peptides xx (2004) xxx–xxx 5

[27]. This suggests a possibility that narcolepsy may result the basal forebrain cholinergic neurons and monoaminergic
from selective autoimmune degeneration of orexin neurons. neurons in the brain stem to maintain arousal. In addition,
The decreased CSF orexin-A peptide level in most orexin neurons also appear to act on LDT/PPT cholinergic
narcolepsy patients also suggests that measuring CSF neurons, because orexin neurons project directly to the PPT/
orexin-A might be a definitive diagnostic test [28]. LDT nuclei and direct injection of orexin-A into the LDT of
Since narcolepsy is a disorder of sleep–wake cycle cats results in an increase in wakefulness and a decrease in
organization resulting from absence of orexin, replacement REM sleep [38]. In addition, several reports showed that
therapy using orexin receptor agonists may provide treat- orexin induces long-lasting excitation of cholinergic neu-
ment of narcolepsy. Our recent study showed that chronic rons in the LDT [39]. However, a group of cholinergic
over-production of both orexin-A and orexin-B peptides neurons in the LDT/PPT, which are silent during wakeful-
from an ectopically expressed transgene prevented the ness and active in the REM period, constitute a system to
development of narcolepsy syndrome in orexin neuron- induce and maintain REM sleep [40]. Therefore, orexin
ablated (orexin/ataxin-3) mice [29]. Similarly, acute admin- neurons might not exert a direct excitatory influence on
istration of orexin-A maintained wakefulness, suppressed these cells during wakefulness. Orexin neurons might
sleep, and inhibited cataplectic attacks in narcoleptic mice activate another type of cholinergic neurons in the PPT
[29]. Together, these findings provide strong evidence for a and LDT, which are active in wakefulness as well as the
specific relationship between absence of orexin peptides in REM-sleep period. Recent work also shows that orexin
the brain and the development of narcolepsy syndrome. inhibits cholinergic neurons in the PPT via activation of
GABAergic local interneurons and GABAergic neurons in
the substantia nigra pars reticulata [41]. These results
6. Mechanisms of regulation of behavioral states by suggest that hypothalamic orexin neurons affect the activity
orexins of LDT/PPT cholinergic neurons directly and/or indirectly
to appropriately regulate the activity of these cells to control
The finding of orexin deficiency in narcoleptic patients behavioral states.
suggests that orexin has an important role in the normal Several reports showed that the effect of orexin on
regulation of sleep/wakefulness. Orexin neurons might be wakefulness is largely mediated by activation of the
especially important for stabilization of behavioral states, histaminergic system mediated through OX2R. In rats,
because the major symptom in narcolepsy is inability to i.c.v. injection of orexin during the light period potently
maintain each behavioral state, which results in sleep/ increases the wake period, and this effect is markedly
wakefulness fragmentation. When orexin-A was injected attenuated by the H1 antagonist, pyrilamine [35]. Further-
intracerebroventricularly into rats during the light period, it more, the effect of orexin-A on wakefulness in mice is
caused increased wakefulness time and decreased REM and almost completely absent in H1-receptor deficient mice [42].
non-REM sleep time [30]. In rats, Fos expression of orexin Furthermore, OX 2 R knockout mice exhibit a narcoleptic
neurons is increased during the dark active period [31], and phenotype, while OX 1 R knockout mice show only mild
orexin level in cerebrospinal fluid also peaks during the dark fragmentation of behavioral states [43]. Because OX2R is
period and decreases during the light rest period [32]. These strongly expressed in the TMN, while OX1R is strongly
observations suggest that orexin neurons are active during expressed in the LC, the TMN seems to be an important
the active period and support wakefulness, and are inactive effector site of orexin for sleep/wakefulness regulation.
during the sleep period. The activities of the monoaminergic Orexin neurons also contain an additional neurotransmitter,
neurons in the brain stem are also synchronized and strongly dynorphin [14]. Dynorphin in orexin neurons might inhibit
associated with behavioral states: they fire tonically during GABAergic input to TMN neurons, and thus act in concert
wakefulness, less during non-REM sleep, and not at all with orexin to increase the excitability of these neurons [17].
during REM sleep [33]. This regulation might be at least in However, several findings indicate that signaling through
part by orexin neurons, which are also wake-active, because OX1R is also important for the regulation of wakefulness.
orexin neurons project to and excite histaminergic neurons As mentioned before, OX 2 R knockout mice exhibit charac-
in the TMN, noradrenergic neurons in the LC and teristics of narcolepsy [25]. Interestingly, OX 1 R knockout
serotonergic neurons in the dorsal raphe (DR) [7–9], and mice do not have any overt behavioral abnormalities and
the presence of OX1R in the LC and OX2R in TMN and exhibit only mild fragmentation of behavioral states [43].
both receptors in the DR has been confirmed [6]. Consistent However, the behavioral and electroencephalographic phe-
with this hypothesis, isolated cells from these nuclei are all notype of OX 2 R knockout mice is less severe than that
activated by orexins in vitro [30,34,35]. Orexins also have a found in prepro-orexin knockout mice and double receptor
strong direct excitatory effect on cholinergic neurons of the knockout (OX 1 R- and OX 2 R-null) mice, which appear to
basal forebrain [36], which is hypothesized to play an have the same phenotype as prepro-orexin knockout mice.
important role in behavioral and electrocortical arousal [37]. These observations suggest that OX 1 R also has additional
These observations suggest that orexin neurons are active effects on sleep/wakefulness regulation. These findings
during the wake period, and exert an excitatory influence on suggest that despite the lack of an overt OX 1 R phenotype,
ARTICLE IN PRESS
6 T. Sakurai / Regulatory Peptides xx (2004) xxx–xxx

loss of signaling through both receptor pathways is orexin neurons might also be necessary for maintenance of
necessary for severe narcoleptic phenotype. non-REM sleep.
Willie et al. [25] classified, using behavioral, electro-
physiological, and pharmacological criteria, two distinct
classes of behavioral arrest exhibited by mice deficient in 7. Regulation of orexin neuronal activity
orexin-mediated signaling. Both OX 2 R and prepro-orexin
knockout mice are similarly affected with behaviorally Until recently, little was known about the factors that
abnormal attacks of non-REM sleep (bsleep attacksQ) and influence the activity of orexin neurons. Recent electro-
show similar degrees of disrupted wakefulness. In contrast, physiological studies have identified several activators and
OX 2 R knockout mice are only mildly affected with inhibitors of orexin neurons. By recording from hypothala-
cataplexy-like attacks of REM sleep, whereas orexin mic slices of transgenic mice expressing green fluorescent
knockout mice are severely affected. Absence of OX 2 R protein (GFP) only in orexin neurons, it was shown that
eliminates orexin-evoked excitation of histaminergic neu- agonists of ionotropic glutamate receptors (AMPA and
rons in the hypothalamus, which gate non-REM sleep/ NMDA) excite orexin neurons, whereas glutamate antag-
wake transition. While normal regulation of wake/non- onists (AP-5, CNQX or NBQX) reduce their activity [45].
REM sleep transition depends critically upon OX2R These results indicate that orexin neurons are tonically
activation, the profound dysregulation of REM sleep activated by glutamate. Although orexin has little direct
control unique to the narcolepsy syndrome emerges from effect on the activity of orexin neurons, it increases this
loss of signaling through both OX2R-dependent and glutamate signaling by acting on presynaptic terminals
OX1R-dependent pathways in mice. [45]. This mechanism may reinforce and coordinate the
Thus, orexin neurons may be active during wakefulness, activity of orexin neurons in the LHA.
helping sustain activity in monoaminergic arousal regions Several researchers have hypothesized that monoamines
[31], which in turn send inhibitory input to the ventro- excite orexin neurons, forming positive feedback loops that
lateral preoptic area (VLPO) sleep-active neurons, and would maintain wakefulness [21], but our electrophysio-
thereby further maintain wakefulness [44]. In the absence logical studies showed just the opposite; both noradrena-
of orexin, these arousal regions may have reduced or non- line and serotonin hyperpolarize and inhibit GFP-
synchronized activity, resulting in an inappropriately low expressing orexin neurons [45,46]. Histamine has no effect
threshold for transition into non-REM sleep. Narcoleptic on orexin neurons. It seems strange that wake-active
mice also have fragmented non-REM sleep; orexin- monoaminergic areas would inhibit wake-active orexin
deficient mice have more frequent transitions between all neurons. Therefore, we hypothesize that orexin neurons
states, as has been noted in human narcolepsy. Therefore, might be innervated by monoaminergic cells in regions

Fig. 4. Roles of orexin in coordination of energy and sleep homeostasis. Orexin neurons stimulate the hypothalamic nuclei involved in feeding behavior and
increase cortical arousal and promote wakefulness through the aminergic nuclei and other sleep-related nuclei. Stimulation of dopaminergic, limbic, and
cholinergic centers by orexins can modulate reward systems, motor activity, and emotional arousal. Peripheral metabolic signals, leptin, ghrelin, and glucose
and circadian rhythms influence orexin neuronal activity to coordinate arousal and energy homeostasis.
ARTICLE IN PRESS
T. Sakurai / Regulatory Peptides xx (2004) xxx–xxx 7

other than the wake-active regions. To evaluate this wakefulness throughout the day. These mechanisms may be
hypothesis, precise retrograde mapping study of afferent important in maintenance of prolonged wakefulness during
neurons to orexin neurons is required. the active period, and in the regulation of energy homeo-
stasis that helps to ensure survival in nature, but may
counteract attempts to treat obesity by food restriction.
8. Roles of orexins in the regulation of energy
homeostasis
Acknowledgements
The altered energy homeostasis in human narcolepsy
patients suggests roles of orexin in regulation of energy This study was supported in part by a grant-in-aid for
homeostasis [47,48]. The finding of decreased caloric scientific research from the Ministry of Education,
intake [49] combined with an increased body mass index Culture, Sports, Science and Technology (MEXT) of
[47] suggests that narcolepsy patients have a feeding Japan; University of Tsukuba Project Research; The 21st
abnormality with reduced energy expenditure or low century COE program; the Uehara Memorial Foundation;
metabolic rate, and orexin neurons have a role in the and the ERATO from the Japan Science and Technology
regulation of energy homeostasis. Consistently, orexin Corporation.
neuron-ablated mice show hypophagia and late-onset
obesity [24].
Electrophysiological studies on orexin neurons showed References
that, in addition to monoamines and acetylcholine,
peripheral humoral factors related to energy metabolism [1] Wise A, Jupe SC, Rees S. The identification of ligands at orphan G-
also influence the activity of orexin neurons; activity of protein coupled receptors. Annu Rev Pharmacol Toxicol 2004;44:
43 – 66.
isolated orexin neurons is inhibited by glucose and leptin,
[2] Sakurai T, Amemiya A, Ishii M, Matsuzaki I, Chemelli RM, Tanaka
and stimulated by ghrelin [50]. Consistently, orexin H, et al. Orexins and orexin receptors: a family of hypothalamic
expression of normal and ob/ob mice correlates nega- neuropeptides and G protein-coupled receptors that regulate feeding
tively with changes in blood glucose, leptin, and food behavior. Cell 1998;92:573 – 85.
intake. These findings are consistent with our original [3] Sakurai T, Morigushi T, Furuya K, Kajiwara N, Nakamura T,
idea that orexins have a role in the regulation of feeding Yanagisawa M, et al. Structure and function of human prepro-orexin
gene. J Biol Chem 1999;274:17771 – 6.
and energy homeostasis [2]. [4] de Lecea L, Kilduff TS, Peyron C, Gao X, Foye PE, Danielson PE, et
al. The hypocretins: hypothalamus-specific peptides with neuro-
excitatory activity. Proc Natl Acad Sci U S A 1998;95:322 – 7.
9. Orexin: a link between energy homeostasis and [5] Zhu Y, Miwa Y, Yamanaka A, Yada T, Shibahara M, Abe Y, et al.
arousal Orexin receptor type-1 couples exclusively to pertussis toxin-
insensitive G-proteins, while orexin receptor type-2 couples to both
pertussis toxin-sensitive and -insensitive G-proteins. J Pharmacol Sci
Proper maintenance of arousal during food search and 2003;92:259 – 66.
intake of an animal is essential for its survival. Therefore, [6] Marcus JN, Aschkenasi CJ, Lee CE, Chemelli RM, Saper CB,
the two vital processes, feeding and sleep/wake behavior, Yanagisawa M, et al. Differential expression of orexin receptors 1 and
have to be appropriately coordinated. When faced with a 2 in the rat brain. J Comp Neurol 2001;435:6 – 25.
[7] Date Y, Ueta Y, Yamashita H, Yamaguchi H, Matsukura S, Kangawa
negative energy balance due to reduced food availability, T, et al. Orexins, orexigenic hypothalamic peptides, interact with
mammals respond behaviorally with phases of increased autonomic, neuroendocrine and neuroregulatory systems. Proc Natl
wakefulness and locomotor activity that support food Acad Sci U S A 1999;96:748 – 53.
seeking [51–55]. The discovery that orexin neurons are [8] Nambu T, Sakurai T, Mizukami K, Hosoya Y, Yanagisawa M, Goto K.
regulated by peripheral metabolic cues suggests that Distribution of orexin neurons in the adult rat brain. Brain Res
1999;827:243 – 60.
orexin neurons might have important roles in the [9] Peyron C, Tighe DK, van den Pol AN, de Lecea L, Heller HC,
molecular and physiological basis of this phenomenon. Sutcliffe JG, et al. Neurons containing hypocretin (orexin) project to
During starvation, orexin neurons might be activated by multiple neuronal systems. J Neurosci 1998;18:9996 – 10015.
low leptin and glucose levels, along with high ghrelin [10] Bittencourt JC, Presse F, Arias C, Peto C, Vaughan J, Nahon JL, et al.
The melanin-concentrating hormone system of the rat brain: an
level. These mechanisms may directly modulate activity
immuno- and hybridization histochemical characterization. J Comp
of orexin neurons according to appetite and body energy Neurol 1992;319:218 – 45.
stores. Indeed, we found that transgenic mice, in which [11] Broberger C, De Lecea L, Sutcliffe JG, Hokfelt T. Hypocretin/orexin-
orexin neurons are ablated, fail to respond to fasting with and melanin-concentrating hormone-expressing cells form distinct
increased wakefulness and activity [50]. populations in the rodent lateral hypothalamus: relationship to the
These findings indicate that orexin neurons provide a neuropeptide Y and agouti gene-related protein systems. J Comp
Neurol 1998;402:460 – 74.
crucial link between energy balance and arousal (Fig. 4). [12] Elias CF, Saper CB, Maratos-Flier E, Tritos NA, Lee C, Kelly J, et al.
These properties might allow the orexin neurons to promote Chemically defined projections linking the mediobasal hypothalamus
alertness in a hungry animal and maintain long periods of and the lateral hypothalamic area. J Comp Neurol 1998;402:442 – 59.
ARTICLE IN PRESS
8 T. Sakurai / Regulatory Peptides xx (2004) xxx–xxx

[13] Cutler DJ, Morris R, Evans ML, Leslie RA, Arch JR, Williams G. [34] Nakamura T, Uramura K, Nambu T, Yada T, Goto K, Yanagisawa
Orexin-A immunoreactive neurons in the rat hypothalamus do not M, et al. Orexin-induced hyperlocomotion and stereotypy are
contain neuronal nitric oxide synthase (nNOS). Peptides 2001;22: mediated by the dopaminergic system. Brain Res 2000;873:181 – 7.
123 – 128. [35] Yamanaka A, Tsujino N, Funahashi H, Honda K, Guan JL, Wang
[14] Chou TC, Lee CE, Lu J, Elmquist JK, Hara J, Willie JT, et al. Orexin QP, et al. Orexins activate histaminergic neurons via the orexin- 2
(hypocretin) neurons contain dynorphin. J Neurosci 2001;21:RC168. receptor. Biochem Biophys Res Commun 2002;290:1237 – 45.
[15] Risold PY, Griffond B, Kilduff TS, Sutcliffe JG, Fellmann D. [36] Eggermann E, Serafin M, Bayer L, Machard D, Saint-Mleux B,
Preprohypocretin (orexin) and prolactin-like immunoreactivity are Jones BE, et al. Orexins/hypocretins excite basal forebrain cholinergic
coexpressed by neurons of the rat lateral hypothalamic area. Neurosci neurones. Neuroscience 2001;108:177 – 81.
Lett 1999;259:153 – 6. [37] Alam MN, Szymusiak R, Gong H, King J, McGinty D. Adenosinergic
[16] Abrahamson EE, Leak RK, Moore RY. The suprachiasmatic nucleus modulation of rat basal forebrain neurons during sleep and waking:
projects to posterior hypothalamic arousal systems. NeuroReport neuronal recording with microdialysis. J Physiol 1999;521:679 – 90.
2001;12:435 – 40. [38] Xi M, Morales FR, Chase MH. Effects on sleep and wakefulness of
[17] Eriksson KS, Sergeeva OA, Selbach O, Haas HL. Orexin (hypo- the injection of hypocretin-1 (orexin-A) into the laterodorsal
cretin)/dynorphin neurons control GABAergic inputs to tuberomam- tegmental nucleus of the cat. Brain Res 2001;901:259 – 64.
millary neurons. Eur J Neurosci 2004;19:1278 – 84. [39] Takahashi K, Koyama Y, Kayama Y, Yamamoto M. Effects of orexin
[18] Peyron C, Faraco J, Rogers W, Ripley B, Overeem S, Charnay Y, et al. on the laterodorsal tegmental neurones. Psychiatry Clin Neurosci
A mutation in a case of early onset narcolepsy and a generalized 2002;56:335 – 6.
absence of hypocretin peptides in human narcoleptic brains. Nat Med [40] Koyama Y, Jodo E, Kayama Y. Sensory responsiveness of bbroad-
2000;9:991 – 7. spikeQ neurons in the laterodorsal tegmental nucleus, locus coeruleus
[19] Thannickal TC, Moore RY, Nienhuis R, Ramanathan L, Gulyani S, and dorsal raphe of awake rats: implications for cholinergic and
Aldrich M, et al. Reduced number of hypocretin neurons in human monoaminergic neuron-specific responses. Neuroscience 1994;63:
narcolepsy. Neuron 2000;27:469 – 74. 1021 – 31.
[20] Mignot E, Lammers GJ, Ripley B, Okun M, Nevsimalova S, Overeem [41] Takakusaki K, Takahashi K, Saitoh K, Harada H, Okamura T,
S, et al. The role of cerebrospinal fluid hypocretin measurement in the Koyama Y. Role of orexinergic orijections to the midbrain involved in
diagnosis of narcolepsy and other hypersomnias. Arch Neurol the control of locomotion and postural muscle tone. Abstr. 34th
2002;59:1553 – 62. Meeting for Sci. Neurosci.; 2004.
[21] Saper CB, Chou TC, Scammell TE. The sleep switch: hypothalamic [42] Huang ZL, Qu WM, Li WD, Mochizuki T, Eguchi N, Watanabe T,
control of sleep and wakefulness. Trends Neurosci 2001;24:726 – 31. et al. Arousal effect of orexin A depends on activation of the
[22] Lin L, Faraco J, Li R, Kadotani H, Rogers W, Lin X, et al. The sleep histaminergic system. Proc Natl Acad Sci U S A 2001;98:9965 – 70.
disorder canine narcolepsy is caused by a mutation in the hypocretin [43] Willie JT, Chemelli RM, Sinton CM, Yanagisawa M. To eat or to
(orexin) receptor 2 gene. Cell 1999;98:365 – 76. sleep? Orexin in the regulation of feeding and wakefulness. Annu Rev
[23] Chemelli RM, Willie JT, Sinton CM, Elmquist JK, Scammel TE, Lee Neurosci 2001;24:429 – 58.
C, et al. Narcolepsy in orexin knockout mice: molecular genetics of [44] Gallopin T, Fort P, Eggermann E, Cauli B, Luppi PH, Rossier J, et al.
sleep regulation. Cell 1999;98:437 – 51. Identification of sleep-promoting neurons in vitro. Nature 2000;404:
[24] Hara J, Beuckmann CT, Nambu T, Willie JT, Chemelli RM, Sinton 992 – 995.
CM, et al. Genetic ablation of orexin neurons in mice results in [45] Li Y, Gao XB, Sakurai T, van den Pol AN. Hypocretin/Orexin excites
narcolepsy, hypophagia, and obesity. Neuron 2001;30:345 – 54. hypocretin neurons via a local glutamate neuron-A potential mech-
[25] Willie JT, Chemelli RM, Sinton CM, Tokita S, Williams SC, Kisanuki anism for orchestrating the hypothalamic arousal system. Neuron
YY, et al. Distinct narcolepsy syndromes in orexin receptor-2 and 2002;36:1169 – 81.
orexin null mice: molecular genetic dissection of non-REM and REM [46] Yamanaka A, Muraki Y, Tsujino N, Goto K, Sakurai T. Regulation of
sleep regulatory processes. Neuron 2003;38:715 – 730. orexin neurons by the monoaminergic and cholinergic systems.
[26] Nishino S, Ripley B, Overeem S, Lammers GJ, Mignot E. Hypocretin Biochem Biophys Res Commun 2003;303:120 – 9.
(orexin) deficiency in human narcolepsy. Lancet 2000;355:39 – 40. [47] Schuld A, Hebebrand J, Geller F, Pollmacher T. Increased body-mass
[27] Kadotani H, Faraco J, Mignot E. Genetic studies in the sleep disorder index in patients with narcolepsy. Lancet 2000;1274 – 1275.
narcolepsy. Genome Res 1998;8:427 – 34. [48] Honda Y, Doi Y, Ninomiya R, Ninomiya C. Increased frequency of
[28] Mignot E, Lammers GJ, Ripley B, Okun M, Nevsimalova S, Overeem non-insulin-dependent diabetes mellitus among narcoleptic patients.
S, et al. The role of cerebrospinal fluid hypocretin measurement in the Sleep 1986;9:254 – 9.
diagnosis of narcolepsy and other hypersomnias. Arch Neurol [49] Lammers GJ, Pijl H, Iestra J, Langius JA, Buunk G, Meinders AE.
2002;59:1553 – 62. Spontaneous food choice in narcolepsy. Sleep 1996;19:75 – 6.
[29] Mieda M, Willie JT, Hara J, Sinton CM, Sakurai T, Yanagisawa M. [50] Yamanaka A, Beuckmann CT, Willie JT, Hara J, Tsujino N, Mieda M,
Orexin peptides prevent cataplexy and improve wakefulness in an et al. Hypothalamic orexin neurons regulate arousal according to
orexin neuron-ablated model of narcolepsy in mice. Proc Natl Acad energy balance in mice. Neuron 2003;38:701 – 13.
Sci U S A 2004;101:4649 – 54. [51] Borbely AA. Sleep in the rat during food deprivation and subsequent
[30] Hagan JJ, Leslie RA, Patel S, Evans ML, Wattam TA, Holmes S, et al. restitution of food. Brain Res 1977;124:457 – 71.
Orexin- A activates locus coeruleus cell firing and increases arousal in [52] Danguir J, Nicolaidis S. Dependence of sleep on nutrients’ avail-
the rat. Proc Natl Acad Sci U S A 1999;96:10911 – 6. ability. Physiol Behav 1979;22:735 – 40.
[31] Estabrooke IV, McCarthy MT, Ko E, Chou TC, Chemelli RM, [53] Dewasmes G, Duchamp C, Minaire Y. Sleep changes in fasting rats.
Yanagisawa M, et al. Fos expression in orexin neurons varies with Physiol Behav 1989;46:179 – 84.
behavioral state. J Neurosci 2001;21:1656 – 62. [54] Itoh T, Murai S, Nagahama H, Miyate H, Abe E, Fujiwara H, et al.
[32] Yoshida Y, Fujiki N, Nakajima T, Ripley B, Matsumura H, Yoneda H, Effects of 24-hr fasting on methamphetamine- and apomorphine-
et al. Fluctuation of extracellular hypocretin-1 (orexin A) levels in the induced locomotor activities, and on monoamine metabolism in
rat in relation to the light–dark cycle and sleep–wake activities. Eur J mouse corpus striatum and nucleus accumbens. Pharmacol Biochem
Neurosci 2001;14:1075 – 81. Behav 1990;35:391 – 6.
[33] Vanni-Mercier G, Sakai K, Jouvet M. Neurons specifiques de l’eveil [55] Challet E, Pevet P, Malan A. Effect of prolonged fasting and
dans l’hypothalamus posterieur du chat. CR Acad Sci, III 1984; subsequent refeeding on free-running rhythms of temperature and
298:195 – 200. locomotor activity in rats. Behav Brain Res 1997;84:275 – 84.

You might also like