Vous êtes sur la page 1sur 13

Sleep Medicine Reviews 15 (2011) 269e281

Contents lists available at ScienceDirect

Sleep Medicine Reviews


journal homepage: www.elsevier.com/locate/smrv

PHYSIOLOGICAL REVIEW

Serotonin control of sleep-wake behavior


Jaime M. Monti*
Department of Pharmacology and Therapeutics, School of Medicine Clinics Hospital, Montevideo 11600, Uruguay

a r t i c l e i n f o s u m m a r y

Article history: Based on electrophysiological, neurochemical, genetic and neuropharmacological approaches, it is


Received 28 August 2010 currently accepted that serotonin (5-HT) functions predominantly to promote wakefulness (W) and to
Received in revised form inhibit REM (rapid eye movement) sleep (REMS). Yet, under certain circumstances the neurotransmitter
27 November 2010
contributes to the increase in sleep propensity. Most of the serotonergic innervation of the cerebral cortex,
Accepted 28 November 2010
Available online 2 April 2011
amygdala, basal forebrain (BFB), thalamus, preoptic and hypothalamic areas, raphe nuclei, locus coeruleus
and pontine reticular formation comes from the dorsal raphe nucleus (DRN). The 5-HT receptors can be
classied into at least seven classes, designated 5-HT1e7. The 5-HT1A and 5-HT1B receptor subtypes are
Keywords:
Serotonin
linked to the inhibition of adenylate cyclase, and their activation evokes a membrane hyperpolarization. The
Brain stem actions of the 5-HT2A, 5-HT2B and 5-HT2C receptor subtypes are mediated by the activation of phospholipase
Sleep C, with a resulting depolarization of the host cell. The 5-HT3 receptor directly activates a 5-HT-gated cation
Wakefulness channel which leads to the depolarization of monoaminergic, aminoacidergic and cholinergic cells. The
Slow wave sleep primary signal transduction pathway of 5-HT6 and 5-HT7 receptors is the stimulation of adenylate cyclase
Rapid eye movement sleep which results in the depolarization of the follower neurons. Mutant mice that do not express 5-HT1A or 5-
Dorsal raphe nucleus HT1B receptor exhibit greater amounts of REMS than their wild-type counterparts, which could be related to
5-HT receptors
the absence of a postsynaptic inhibitory effect on REM-on neurons of the laterodorsal and pedunculo-
Laterodorsal and pedunculopontine
pontine tegmental nuclei (LDT/PPT). 5-HT2A and 5-HT2C receptor knock-out mice show a signicant
tegmental nuclei
Basal forebrain increase of W and a reduction of slow wave sleep (SWS) which has been ascribed to the increase of cate-
cholaminergic neurotransmission involving mainly the noradrenergic and dopaminergic systems. Sleep
variables have been characterized, in addition, in 5-HT7 receptor knock-out mice; the mutants spend less
time in REMS that their wild-type counterparts. Direct infusion of the 5-HT1A receptor agonists 8-OH-DPAT
and esinoxan into the DRN signicantly enhances REMS in the rat. In contrast, microinjection of the 5-HT1B
(CP-94253), 5-HT2A/2C (DOI), 5-HT3 (m-chlorophenylbiguanide) and 5-HT7 (LP-44) receptor agonists into
the DRN induces a signicant reduction of REMS. Systemic injection of full agonists at postsynaptic 5-HT1A
(8-OH-DPAT, esinoxan), 5-HT1B (CGS 12066B, CP-94235), 5-HT2C (RO 60-0175), 5-HT2A/2C (DOI, DOM),
5-HT3 (m-chlorophenylbiguanide) and 5-HT7 (LP-211) receptors increases W and reduces SWS and REMS.
Of note, systemic administration of the 5-HT2A/2C receptor antagonists ritanserin, ketanserin, ICI-170,809 or
sertindole at the beginning of the light period has been shown to induce a signicant increase of SWS and
a reduction of REMS in the rat. Wakefulness was also diminished in most of these studies. Similar effects
have been described following the injection of the selective 5-HT2A receptor antagonists volinanserin and
pruvanserin and of the 5-HT2A receptor inverse agonist nelotanserin in rodents. In addition, the effects of
these compounds have been studied on the sleep electroencephalogram of subjects with normal sleep.
Their administration was followed by an increase of SWS and, in most instances, a reduction of REMS. The
administration of ritanserin to poor sleepers, patients with chronic primary insomnia and psychiatric
patients with a generalized anxiety disorder or a mood disorder caused a signicant increase in SWS. The
5-HT2A receptor inverse agonist APD-125 induced also an increase of SWS in patients with chronic primary
insomnia. It is known that during the administration of benzodiazepine (BZD) hypnotics to patients with
insomnia there is a further reduction of SWS and REMS, whereas both variables tend to remain decreased
during the use of non-BZD derivatives (zolpidem, zopiclone, eszopiclone, zaleplon). Thus, the association of
5-HT2A antagonists or 5-HT2A inverse agonists with BZD and non-BZD hypnotics could be a valid alternative
to normalize SWS in patients with primary or comorbid insomnia.
2010 Elsevier Ltd. All rights reserved.

* Tel.: 598 2 710 58 07.


E-mail address: jmonti@mednet.org.uy.

1087-0792/$ e see front matter 2010 Elsevier Ltd. All rights reserved.
doi:10.1016/j.smrv.2010.11.003
270 J.M. Monti / Sleep Medicine Reviews 15 (2011) 269e281

Abbreviations LH posterior lateral hypothalamus


MCH melanin-concentrating hormone
ACh acetylcholine mPRF medial pontine reticular formation
AMPA alpha-amino-3-hydroxy-5-methyl-4-isoxazole MRN median raphe nucleus
propionate NE norepinephrine
BFB basal forebrain NREMS non-REM sleep
BNST bed nucleus of the stria terminalis OX orexin
BZD benzodiazepine PCPA p-chlorophenylalanine
CNS central nervous system PPT pedunculopontine tegmental nucleus
DA dopamine REM rapid eye movement
DRN dorsal raphe nucleus REMS REM sleep
5-HT serotonin s.c. subcutaneous
GABA gamma-aminobutyric acid SNc substantia nigra pars compacta
GLU glutamate SSRI selective serotonin reuptake inhibitor
HA histamine SWS slow wave sleep
i.c.v. intracerebroventricular VLPO ventrolateral preoptic area
IL-1 interleukin-1 VPGM ventral periaqueductal gray matter
i.p. intraperitoneal VTA ventral tegmental area
LC locus coeruleus W wakefulness
LDT/PPT laterodorsal and pedunculopontine tegmental nuclei

Introduction regulating mechanisms are not disrupted in PCPA-treated rats.15


With regard to an alternative hypothesis,16 there is no rm
Within the central nervous system serotonin (5-HT) participates evidence to support the proposal that 5-HT released during W might
in a great number of functions including sleep-wake behavior, act as a neurohormone and induce the synthesis and/or release of
cognition, affect, sexual function, thermoregulation and food hypnogenic factors secondarily responsible for SWS and REM (rapid
intake. It has been established, in addition, that increases or eye movement) sleep (REMS) occurrence.
decreases of 5-HT at central sites correlate with improved or More recently it has been posed that 5-HT should not be
worsened depression. In this respect, most of the drugs used to considered either wake-promoting or SWS-promoting, and that the
treat mood disorders including the selective serotonin reuptake effect of 5-HT on sleep-wake behavior would depend upon the
inhibitors (SSRIs), the tricyclic antidepressants and the monoamine degree to which the serotonergic system is activated (systemic
oxidase inhibitors increase synaptic levels of 5-HT. The SSRIs are administration of low vs. high doses of the 5-HT precursor
also the rst line drugs for the treatment of anxiety disorders such 5-hydroxytryptophan), and the time at which the activation occurs
as generalized anxiety disorder, panic disorder and obsessive (light vs. dark period of the light-dark cycle).17e19 It has been
compulsive disorder1. hypothesized that the 5-hydroxytryptophan-related increase of
Although many questions remain about the complicated role of SWS during the dark period depends upon the synthesis or release
5-HT and its receptors in regulating sleep and wakefulness (W), of as yet to be identied sleep-promoting factors.17 Alternatively,
recent studies have revealed much detailed information about this sleep occurrence might be associated with a 5-HT1A receptor-
process. It should be mentioned that in the 1970s, based on lesion mediated reduction in the activity of cholinergic waking-
studies and neuropharmacological analysis, 5-HT was hypothesized promoting neurons in the basal forebrain (BFB).20,21
to be responsible for the initiation and maintenance of slow wave On the basis of results obtained from genetic, neurochemical,
sleep (SWS).2 However, a series of ndings from several laboratories electrophysiological and neuropharmacological studies conducted
seriously challenged the serotonergic hypothesis of sleep. Thus, the over the past three decades it is proposed that 5-HT functions
ring rate of 5-HT-containing dorsal raphe nucleus (DRN) neurons predominantly to promote W and to inhibit REMS. Notwith-
decreases during SWS relative to W3; reduction of brain 5-HT by p- standing this, under certain circumstances the indolamine seems to
chlorophenylalanine (PCPA) fails to disrupt sleep in humans, and in contribute to the increase in sleep propensity. The evidence that 5-
some studies, also in the rat4,5; 5,7-dihydroxytryptamine, which HT is involved in the regulation of the behavioral state and its
produces a more selective and extensive depletion of brain 5-HT relationship to other sleep-wake regulatory neurotransmitters is
than the raphe lesions, does not signicantly affect SWS6; and the focus of this review.
there is no signicant difference between tryptophan-decient The brain regions and neurotransmitter systems involved in the
and -replete rats in time spent in SWS.7 Concerning those studies regulation of the behavioral state will be described next.
showing that PCPA increases W in the rat,8,9 it should be taken into
consideration that this 5-HT synthesis inhibitor also induces Brain regions and neurotransmitter systems involved in the
increased responsiveness to noxious or neutral environmental regulation of wakefulness
stimuli and enhances motor activity, aggressivity, and sexual
activity.10e13 Therefore, insomnia after PCPA in the rat and the cat The brain regions involved in the promotion of the waking state
could be related to a general hyper-responsiveness to stimuli rather are located in the brain stem, hypothalamus and BFB. The nuclei
than to impairment of sleep regulation itself. Even in the studies in found in the brain stem include serotonin [5-HT: dorsal raphe
which insomnia followed PCPA administration, the effect was nucleus (DRN), median raphe nucleus (MRN)]; norepinephrine [NE:
a transitory one, and SWS returned while 5-HT levels remained locus coeruleus (LC)]; dopamine [DA: ventral tegmental area (VTA),
depleted.14 Moreover, in rats subjected to combined treatment with substantia nigra pars compacta (SNc), ventral periaqueductal grey
PCPA and sleep deprivation, delta activity reached the same high matter (VPGM)]; acetylcholine [ACh: laterodorsal and pedunculo-
levels as after deprivation alone, which tends to indicate that SWS- pontine tegmental nuclei (LDT/PPT); BFB]; and glutamate [GLU:
J.M. Monti / Sleep Medicine Reviews 15 (2011) 269e281 271

Fig. 1. Main neuroanatomical structures that receive afferent projections from the dorsal raphe nucleus (B6 and B7) in the rat CNS. Abbreviations: LDT, laterodorsal tegmental
nucleus; PRF, pontine reticular formation.

medial pontine reticular formation (mPRF); BFB]-containing system participates in the response. In this respect, the DRN
neurons. The structures located in the hypothalamus include contains IL-1 receptors; IL-1b inhibits the ring rates of DRN 5-HT
histamine [HA: tuberomammillary nucleus (TM)]; and orexin [OX: neurons in a slice preparation; and microinjection of IL-1b into the
posterior lateral hypothalamus (LH) around the fornix ]-containing DRN of freely behaving rats increases SWS.27 It has been deter-
cells. The cholinergic and glutamatergic neurons of the BFB involved mined, in addition, that IL-1b inhibits DRN 5-HT cells by potenti-
in the regulation of the behavioral state are located predominantly ating GABAergic inhibitory effects.28
in the medial septum and the diagonal band of Broca.22,23
The neural structures that participate in the regulation of W give
Brain regions and neurotransmitter systems involved in the
rise to mainly ascending projections. In this respect, a) 5-HT-, NE-,
regulation of REM sleep
and HA-containing neurons send long ascending projections to the
thalamus, cerebral cortex, and BFB; b) DA-containing cells project
On the grounds of studies in which the cholinergic agonist
into the basal ganglia (caudate and putamen, external and internal
carbachol was microinjected into rostral pontine structures of the
globus pallidus, subthalamic nucleus) and the frontal cortex;
cat, it has been proposed that the critical areas for REMS generation
c) cholinergic neurons from the midbrain tegmentum (LDT/PPT)
could be one or more of the following: a) the most ventral and
project to the thalamus (ventromedial, intralaminar, and midline
rostral part of the pontine reticular nucleus; b) the perilocus
nuclei) and the BFB, and cholinergic BFB neurons have widespread
coeruleus alpha nucleus (peri-LCa) of the mediodorsal pontine
rostral projections to the cerebral cortex and the hippocampus; d)
tegmentum; c) the rostral part of the rostral pontine tegmentum;
orexin-containing neurons from the LH project to the entire fore-
d) the medial pontine reticular formation which encloses the
brain and brain stem arousal systems; and e) glutamatergic neurons
rostral and caudal part of the pontine reticular nucleus and dorsally
make up the projection neurons of the mPRF and thalamus.23,24 All
reaches the locus coeruleus alpha (LCa), or e) the ventral part of the
these ascending projections into the forebrain follow a dorsal and
pontine reticular nucleus.29e31 The reciprocal interaction hypoth-
a ventral route.23 The dorsal route terminates in nonspecic thalamic
esis of REMS generation identies cholinergic neurons of the LDT/
nuclei, which in turn project to the cerebral cortex. The ventral route
PPT as promoting REMS and posits the inhibition of these neurons
passes through the hypothalamus and continues into the BFB, where
by, among others, serotonergic afferents from the DRN.32 The REMS
cells in turn project to the cerebral cortex and the hippocampus. In
induction region of the mPRF, one of the neuroanatomical struc-
addition, a number of neural structures send descending projections
tures proposed to be responsible for REMS generation, includes
to the spinal cord that modulate muscle tone.
predominantly glutamatergic neurons, which are in turn activated
by efferent connections of the LDT/PPT. The DRN provides the
principal source of 5-HT innervation to the LDT/PPT and the REMS
Brain regions and neurotransmitter systems involved in the
induction zone of the mPRF.33 Accordingly, serotonergic terminals
regulation of slow wave sleep
have been characterized that make synaptic contacts with the soma
and the proximal dendrites of cholinergic tegmental neurons, and
Neurons of the preoptic area, anterior hypothalamus, and
with non-cholinergic, presumptively glutamatergic, neurons of the
adjacent BFB constitute the sleep-inducing system.25 Sleep active
REMS induction zone of the mPRF.34 The DRN also sends ascending
neurons of the preoptic area are mainly located in the ventrolateral
non-serotonergic projections to the LDT/PPT.35,36
preoptic area (VLPO). A majority of these neurons contain g-ami-
nobutyric acid (GABA) and galanin, and project to the BFB and to
brain stem and hypothalamic areas involved in the promotion of W The structure and efferent and afferent connections of the
including the DRN, LC, LDT/PPT and LH neurons. During SWS, the dorsal raphe nucleus
neurons of the VLPO inhibit the W-producing structures. A similar
role has been proposed for the melanin-concentrating hormone Serotonergic neurons of raphe regions of the brain stem are
(MCH). Accordingly, MCH-containing neurons located in the zona regarded as forming caudal and rostral cell groups. The most caudal
incerta, perifornical nucleus and lateral hypothalamic area facilitate nuclei project mainly to the medulla and the spinal cord, whereas
sleep occurrence by inhibiting 5-HT, NE, ACh and OX neurons the most rostral cell aggregates innervate the telencephalon,
involved in the promotion of W.26 Of note, interleukin-1 (IL-1) diencephalon, mesencephalon, and rhombencephalon.37,38 In this
increases SWS in several animal species, and the serotonergic respect, most of the serotonergic innervation of the cerebral cortex,
272 J.M. Monti / Sleep Medicine Reviews 15 (2011) 269e281

Table 1
Efferent and afferent connections of the dorsal raphe nucleus of the rat.

Efferent connections
Telencephalon - Cerebral cortex [frontal, piriform, insular, occipital, entorhinal, cingulate, and infralimbic cortices]
- Limbic system [ventral hippocampus, lateral septal nucleus, amygdala (central, lateral, and basolateral nuclei)]
- Basal forebrain [diagonal band (horizontal and vertical limbs), bed nucleus of the stria terminalis]
- Neostriatum [posteromedial regions of the striatum]
Diencephalon - Thalamus [midline, intralaminar and anterior nuclei]
- Hypothalamus [preoptic area, lateral hypothalamic area, supramammillary nucleus]
Mesencephalon - [Interpeduncular nucleus, substantia nigra pars compacta, central gray, mesencephalic reticular formation]
Rhombencephalon - [Pontine reticular formation, pedunculopontine tegmental nucleus, laterodorsal tegmental nucleus,
nucleus reticularis pontis oralis, nucleus reticularis pontis caudalis, nucleus reticularis gigantocellularis,
locus coeruleus, median raphe nucleus, nucleus raphe pontis]

Afferent connections
Telencephalon - Cerebral cortex [frontal, cingulate, and insular cortices]
- Limbic system [amygdala (central, anterior, medial, and basolateral nuclei), lateral septal nucleus]
- Basal forebrain [medial septal nucleus, ventral pallidum, bed nucleus of the stria terminalis, diagonal band]
Diencephalon - Hypothalamus [medial and lateral preoptic areas; anterior, lateral and posterior hypothalamic areas;
dorsomedial and ventromedial nuclei; tuberomammillary nucleus]
Mesencephalon - [Substantia nigra pars reticulata, interpeduncular nucleus, central gray]
Rhombencephalon - [Raphe nuclei, locus coeruleus, pontine reticular formation]

amygdala, BFB, thalamus, preoptic and hypothalamic areas, raphe postsynaptic receptor induces inhibitory responses on target
nuclei, LC and pontine reticular formation comes from the DRN structures. In this respect, data obtained from in vitro recording in
(Fig. 1). In turn, inputs to the DRN 5-HT cells have been found from the rat bed nucleus of the stria terminalis (BNST) slice have shown
the cerebral cortex, the limbic system, the BFB, the hypothalamus, that exogenous application of 5-HT or a non-selective 5-HT1A
and the cholinergic and monoaminergic nuclei of the brain stem39 receptor agonist (carboxamidotryptamine) evokes a membrane
(Table 1). hyperpolarization in approximately one third and one half of the
The DRN contains 5-HT and non-5-HT neurons. The latter neurons tested, respectively.21 The expression of 5-HT1A receptors
express a variety of substances including DA, GABA and GLU. In by GABAergic interneurons of the DRN raises the possibility that
addition, nitric oxide and a number of neuropeptides have been under certain circumstances their activation could actually result in
characterized in the DRN. Of note, numerous brain areas have facilitation of 5-HT cells. Brain areas rich in 5-HT1A receptors
neurons that project to the DRN and express monoamines (NE, HA), include the telencephalon [cerebral cortex, limbic system (medial
amino acids (GABA, GLU), ACh or neuropeptides (OX, MCH, corti- and lateral septal nuclei, hippocampal formation, amygdala), BFB];
cotropin releasing factor and substance P among others) that the diencephalon [thalamus, hypothalamus (ventromedial and
directly or indirectly, through local circuits, regulate the activity of supraoptic nuclei)]; the mesencephalon (VTA/SNc); and the
5-HT cells.40e44 rhombencephalon (LDT/PPT, DRN, MRN, raphe pallidus, raphe
In summary, the DRN serotonergic neurons receive a prominent obscurus, LC).47
innervation from sleep-active GABAergic and MCHergic neurons in The 5-HT1B receptor is linked to the inhibition of adenylate
the VLPO and other subregions which contribute to their inhibition cyclase and was initially proposed to be located at presynaptic
during sleep. The serotonergic cells receive also an excitatory input (5-HT axon terminals) and postsynaptic (outside the DRN) sites.
from monoamine (NE, DA, HA), OX and ACh-containing neurons in The 5-HT1B receptor has been characterized also in the ventro-
the brain stem and the hypothalamus which reinforce their acti- medial DRN where it is expressed by non-5-HT cells.48 Of note, bath
vation during W. application of the selective 5-HT1B receptor agonist CP 93129
reduced the amplitude of evoked excitatory postsynaptic currents
Serotonin receptors in an in vitro rat BNST slice preparation. The effect of the 5-HT1B
ligand was blocked by the selective 5-HT1B antagonist GR 55562,
Understanding the mode of action and effects of 5-HT receptors and was tentatively ascribed to a reduction of glutamate release at
is fundamental to appreciating the physiological role of 5-HT on the presynaptic sites in the BNST.49 The 5-HT1B receptor has been found
behavioral state. in the telencephalon [cerebral cortex, limbic system (amygdala,
The 5-HT receptors can be classied into at least seven classes, hippocampus, lateral septal nucleus), BFB, basal ganglia]; the
designated 5-HT1e7. The 5-HT1, 5-HT2, 5-HT3 and 5-HT5 classes consist diencephalon [thalamus, hypothalamus (lateral preoptic area,
of ve (5-HT1AeBeDeEeF), three (5-HT2AeBeC) and two (5-HT3AeB and anterior, lateral and dorsal hypothalamic areas, suprachiasmatic
5-HT5AeB) subtypes, respectively, whereas the 5-HT4, 5-HT6 and 5-HT7 nucleus)]; the mesencephalon (central gray, interpeduncular
classes have at present one subtype each.45 Except for the 5-HT3 nucleus, VTA/SNc); and the rhombencephalon (DRN/MRN, LC,
receptor, all other 5-HT receptors are structurally related to the raphe magnus nucleus, cerebellum).50,51
superfamily of G-protein-coupled receptors. The 5-HT2A, 5-HT2B and 5-HT2C receptors exhibit 46e50%
Early studies proposed that the 5-HT1A receptor is exclusively overall sequence identity. They are primarily coupled to Gq and
located on the soma and the dendrites (somatodendritic autor- their actions are mediated by the activation of phospholipase C,
eceptor) of 5-HT neurons and at postsynaptic sites (outside the with a resulting depolarization of the host cell. Receptors of the
DRN). More recently, the 5-HT1A receptor has been found to be 5-HT2 subfamily are located within postsynaptic structures,
expressed also by non-5-HT cells of the DRN. Most of these cells are predominantly on proximal and distal dendritic shafts. The 5-HT2A
GABAergic interneurons.46 The 5-HT1A receptor couples to Gi/o, and the 5-HT2C receptors have been characterized in the telen-
with Gi activation leading to an inhibition of adenylate cyclase. cephalon [cerebral cortex, olfactory system, limbic system (septal
Stimulation of the somatodendritic 5-HT1A receptor inhibits the nuclei, hippocampal formation, amygdala), BFB, basal ganglia]; the
ring rate of serotonergic neurons, whereas activation of the diencephalon [thalamus, hypothalamus (medial and lateral
J.M. Monti / Sleep Medicine Reviews 15 (2011) 269e281 273

preoptic areas, ventromedial and dorsomedial nuclei, mammillary neuronal activity. Notwithstanding the above, numerous presumed
nucleus)]; the mesencephalon (central gray, VTA/SNc); and the DRN 5-HT neurons do not adhere to this pattern of discharge.68
rhombencephalon (LDT/PPT, mPRF, DRN/MRN, LC).52e54 The pres-
ence of 5-HT2B receptors at central sites is restricted to the amyg- The role of 5-HT in regulating wakefulness and REMS
dala, lateral septum, hypothalamus and cerebellum.55
Interestingly, Guo et al.56 have demonstrated that one third and Strategies aimed at determining the role of 5-HT receptors in the
one fourth of neurons of the rat BNST respond to 5-HT application regulation of W and REMS have included: 1. genetic (knock-out)
with depolarization or hyperpolarization/depolarization, respec- technology of several of the molecularly dened receptor subtypes;
tively. Prior application of the selective 5-HT1A receptor antagonist 2. local brain delivery of 5-HT receptor agonists and antagonists; 3.
WAY 100635 blocked the inhibitory response. On the other hand, systemic and intracerebroventricular (i.c.v.) administration of 5-HT
application of the 5-HT2A antagonist MDL 100907 (volinanserin) or receptor ligands.
the 5-HT2C antagonist RS 102221 blocked the excitatory response,
which indicates that 5-HT2A and 5-HT2C receptors intervene in the The 5-HT1A receptor
5-HT-induced excitation of BNST cells.
The 5-HT3 receptor is not coupled to G proteins. It directly Flesinoxan and 8-OH-DPAT, two full agonists of the 5-HT1A
activates a 5-HT-gated cation channel, which leads to the depo- receptor at pre- and post-synaptic sites, the azapirones, buspirone,
larization of monoaminergic, aminoacidergic and cholinergic cells. gepirone and ipsapirone, which behave as full agonists at the
The 5-HT3 receptor is present in cortical and subcortical structures somatodendritic autoreceptor but only as partial agonists at the
including the cerebral cortex, olfactory system, limbic system postsynaptic level, and the selective high-afnity silent antagonists
(hippocampus, amygdala), basal ganglia, central gray, mesence- WAY 100635 and p-MPPI have been used to characterize the role of
phalic reticular formation, and DRN.57,58 At the DRN level the 5-HT3 5-HT1A receptor in the regulation of sleep and W.
receptor is partly expressed by glutamatergic interneurons.59 Mutant mice that do not express 5-HT1A receptors exhibit
The 5-HT6 is a G-protein coupled receptor and its primary signal greater amounts of REMS than their wild-type counterparts while
transduction pathway is the stimulation of adenylate cyclase.60 The W and SWS remain unchanged. As expected, systemic 8-OH-DPAT
5-HT6 receptor is located postsynaptically to 5-HT cells and has has no effect on sleep or W in the mutant mice.69 It can be proposed
been localized in the cerebral cortex, BFB (BNST), limbic system that the increase of the behavioral state in the 5-HT1A receptor
(hippocampus, amygdala), striatum, nucleus accumbens, thalamus, knock-out mice is related to the absence of a postsynaptic 5-HT1A
hypothalamus and mesencephalon of rat and man.61,62 inhibitory effect on REM-on neurons of the LDT/PPT.
The 5-HT7 receptor is positively coupled to adenylate cyclase via Portas et al.70 provided direct evidence that suppression of DRN
GS-proteins. It has been localized in the pyramidal neurons of the serotonergic activity increases REMS. In this study, extracellular
cerebral cortex, BFB (BNST), anterior thalamus, hippocampus (elds 5-HT was measured in the DRN and, simultaneously, behavioral
CA1 and CA3 of Ammons horns), suprachiasmatic nucleus, pontine state changes were determined from polygraphic recordings.
nuclei, brain stem reticular formation, DRN and MRN.60,63 The 5-HT7 Microdialysis perfusion of 8-OH-DPAT into the DRN decreased 5-HT
receptor participates together with 5-HT2A and 5-HT2C receptors in levels by 50% during W and signicantly increased REMS. More-
the activation of BNST cells. Hence, prior application of the selective over, direct infusion of either 8-OH-DPAT or esinoxan into the DRN
5-HT7 receptor antagonist SB 269970 blocked or attenuated the signicantly enhanced REMS in rats, and this effect was prevented
depolarizing response to 5-HT in BNST neurons of the rat.56,64 by local infusion of WAY 100635.71 In contrast, microinjection of
In conclusion, 5-HT receptors with known operational charac- WAY 100635 or p-MPPI into the DRN reduced REMS.71,72 Signi-
teristics exhibit similar regional distribution patterns within the cantly, microinjection of 8-OH-DPAT or esinoxan into the LDT or
CNS, especially throughout the brain stem, hypothalamus, thal- the mPRF, where structures that act to promote and to induce REMS
amus, limbic system, BFB and cerebral cortex. Moreover, 5-HT1A, 5- are located, selectively inhibited REMS in the cat and the rat
HT1B, 5-HT2A, 5-HT2C, 5-HT3 and 5-HT7 receptors are part of the key whereas direct infusion of WAY 100635 into the LDT increased
determinants of 5-HT effects on sleep and W. REMS.73,74
Systemic injection of esinoxan increases W and reduces SWS
and REMS in the rat.74 Similar effects have been observed following
Firing pattern of serotonergic neurons of the DRN the administration of 8-OH-DPAT to vehicle-treated and serotonin-
depleted animals.75e77 Pretreatment with the mixed b-adreno-
Spontaneously active immunohistochemically identied 5-HT ceptor and 5-HT1A/1B receptor antagonist ()pindolol or p-MPPI
cells recorded in a slice preparation of the rat DRN are characterized reversed the effect of 8-OH-DPAT on W and SWS.72,75
by slow, rhythmic activity, a broad action potential, a large after Although it remains unclear how 5-HT1A receptor agonists
hyperpolarization potential, and a decrease of their ring rate in inuence W, it is likely through both inhibition of sleep active
response to the stimulation of the 5-HT1A somatodendritic GABAergic neurons of the VLPO, and activation of arousal systems
receptor.65 Allers and Sharp66 characterized a group of neurons of the located in the BFB (ACh), brain stem (NE, DA) and hypothalamus
DRN immunoreactive for 5-HT or both 5-HT and tryptophan (HA, OX). The inhibition of GABAergic cells that express the 5-HT1A
hydroxylase in urethane-anesthetized rats. The neurons red broad receptor and make synaptic contacts with neurons of the arousal
spikes with low frequency and a highly regular pattern. Systemic systems would result in the increase of W. The reduction of REMS
administration of the 5-HT1A receptor agonist 8-OH-DPAT markedly after systemic esinoxan or 8-OH-DPAT administration would be
inhibited the ring rate of these cells. Electrophysiological recordings related to the activation of postsynaptic 5-HT1A receptors on REM-
in unanesthetized animals allowed McGinty and Harper67 and on neurons of the LDT/PPT, similar to the physiologic effect of 5-HT.
Trulson and Jacobs3 to determine the activity of DRN 5-HT neurons Buspirone, ipsapirone, and gepirone have been shown to
acrcoss the sleep-wake cycle. During quiet W, 5-HT neurons re in increase time awake and to reduce SWS and REMS when given
a slow and regular fashion. During active W, the neuronal activity subcutaneous (s.c.) or intraperitoneal (i.p.) to rats.78,79 Subcuta-
shows a 30e50% increase. As the animal enters SWS the ring rate neous injection of the azapirones into 5,7-dihydroxytryptamine-
decreases to 50% of the quiet W state and is no longer regular. Finally, pretreated rats led also to an increase of W and a reduction of sleep;
during REMS there is a further decrease or even a cessation of this result further supports the dependence of these effects on
274 J.M. Monti / Sleep Medicine Reviews 15 (2011) 269e281

Table 2
The role of serotonin 5-HT1A and 5-HT1B receptors in the regulation of sleep and wakefulness in laboratory animals.

W SWS REMS Ref.


5-HT1A receptor
Mutant mice that do not express 5-HT1A receptors n.s. n.s. 69
5-HT1A receptor agonists (8-OH-DPAT, esinoxan)
Microinjection into the DRN (somatodendritic) n.s. n.s. 71
Microinjection into the LDT or the mPRF (receptor expressed by ACh and GLU neurons) n.s. n.s.  74
Systemic administration (postsynaptic)   74
Partial agonists at postsynaptic sites (buspirone, ipsapirone, gepirone)
Systemic administration   78,79
Selective serotonin reuptake inhibitor (uoxetine)
Microinjection into the LDT n.s. n.s.  86
Systemic administration (postsynaptic) n.s. n.s.  84

5-HT1B receptor
Mutant mice that do not express 5-HT1B receptors n.s. n.s. 87
5-HT1B receptor agonists (CP-94253, CGB 12066B)
Microinjection into the DRN (receptor expressed by GABAergic interneurons) n.s. n.s.  88
Systemic administration (postsynaptic)   90,91

Abbreviations: DRN, dorsal raphe nucleus; LDT, laterodorsal tegmental nucleus; mPRF, medial pontine reticular formation; ACh, acetylcholine; GLU, glutamate; W, wake-
fulness; SWS, slow wave sleep; REM, rapid eye movement; REMS, REM sleep; n.s., non-signicant; , increased; , reduced.

postsynaptic 5-HT1A receptors.79 Similar sleep-disrupting effects of Systemic administration of the selective 5-HT1B receptor
buspirone and ipsapirone have been described in human subjects agonists CGS 12066B and CP-94253 signicantly increased W and
with normal sleep and in patients with insomnia. Thus, buspirone reduced SWS and REMS in the rat.90,91 Pindolol antagonized the
administration increased W, prolonged REMS latency, and reduced increase of W and reduction of SWS induced by CP-94253;
REMS time in insomniac patients,80,81 whereas ipsapirone pro- however, REMS remained suppressed. On the other hand, acute
longed REMS latency and reduced REMS and SWS in healthy pharmacological blockade of the 5-HT1B receptor with GR 125939
subjects.82 The reduction of REMS could be related to the azapir- produced an increase of REMS over 4 hours in wild-type mice.87
ones acting on postsynaptic 5-HT1A receptors in the LDT/PPT nuclei Thus, the limited available evidence indicates that 5-HT1B
to inhibit REM-on neurons. receptor activation facilitates the occurrence of W and negatively
The effects of acute administration of selective serotonin reup- inuences REMS (Table 2).
take inhibitors (SSRIs) on sleep have been studied in laboratory
animals, healthy adults, and depressed patients.83e85 SSRIs are The 5-HT2 receptor
potent REMS suppressors, prolonging the latency to the rst REM
period. The inhibitory effect of SSRIs on REMS has been proposed to 5-HT2A and 5-HT2C receptor knock-out mice show a signicant
result from enhancement of central nervous system (CNS) seroto- increase of W and a reduction of SWS while REMS remains
nergic neurotransmission. In this respect, direct infusion of uox- unchanged.92,93 Systemic administration of non-selective 5-HT2A/2C
etine into the LDT or the mPRF induced a signicant reduction of receptor agonists or a selective 5-HT2C receptor agonist consistently
REMS in the rat; pretreatment with WAY 100635 prevented the increases W and reduces sleep. Thus, opposite effects of gene
reduction of REMS induced by the local administration of uoxetine deletion versus acute pharmacological activation of the same
into the cholinergic nucleus86 (Table 2). protein should have been expected on W and SWS. Although the
basis for this apparent discrepancy is unclear, it has been posed that
The 5-HT1B receptor the greater amounts of W in 5-HT2C receptor knock-out mice and,
by extension, in 5-HT2A receptor knock-out mice, could be related
Mutant mice that do not express the 5-HT1B receptor exhibit to the increase of catecholaminergic neurotransmission involving
greater amounts of REMS than their wild-type counterparts mainly the noradrenergic and the dopaminergic systems.92 5-HT2A
whereas W and SWS show no signicant changes.87 The increase of and 5-HT2C receptors that participate in the control of DA and NE
REMS in 5-HT1B receptor knock-out mice could be tentatively release are expressed by inhibitory GABAergic interneurons.94
related to the absence of a postsynaptic 5-HT1B inhibitory effect on Consequently, the reduction of GABA release at critical sites in the
the cholinergic neurons of the LDT/PPT nuclei. However, this CNS of 5-HT2A and 5-HT2C receptor knock-out mice would be
proposal requires further investigation. indirectly responsible for the increased availability of NE and DA.
Microinjection of the selective 5-HT1B receptor agonist CP- Infusion of the 5-HT2A/2C receptor agonist DOI into the DRN
94253 into the DRN during the light phase caused a reduction of induced a signicant reduction of REMS and of the number of REM
REMS and of the mean duration of REM periods in the rat. periods in the rat. Pretreatment with the selective 5-HT2A or 5-HT2C
Pretreatment with the 5-HT1B receptor antagonist SB-224289 receptor antagonists EMD 281014 (pruvanserin) or SB-243213,
antagonized the CP-94253-induced decrease of REMS and of the respectively, prevented the DOI-induced suppression of REMS,
mean duration of REM periods. Administration of the GABAA which indicates that it was mediated by the 5-HT2A and 5-HT2C
receptor agonist muscimol, which at the doses given did not receptors located in the DRN.95 The serotonin 5-HT2A and 5-HT2C-
signicantly affect sleep variables, prevented also the effect of CP- receptor containing neurons are predominantly GABAergic inter-
94253 on REMS.88 Activation of the 5-HT1B receptor in the DRN has neurons. Systemic or intra-raphe administration of DOI inhibits the
been shown to inhibit the release of GABA.89 Thus, the decrease of ring of serotonergic neurons in the DRN and reduces the extra-
GABA inhibitory tone on 5-HT neurons would result in an incre- cellular concentration of 5-HT96; both effects are reversed by the
ment of 5-HT release at postsynaptic sites and the suppression of 5-HT2A receptor antagonist pruvanserin or the GABAA antagonist
REMS. This conclusion is drawn from the fact that microinjection of picrotoxinin.97 Thus, available evidence does not support the direct
muscimol prevented the CP-94253-induced decrease of REMS. involvement of 5-HT cells in the DOI-suppression of REMS. A
J.M. Monti / Sleep Medicine Reviews 15 (2011) 269e281 275

Table 3 Thus, available evidence tends to indicate that the injection of


The role of serotonin 5-HT2A and 5-HT2C receptors in the regulation of sleep and non-selective 5-HT2A/2C or selective 5-HT2C receptor agonists
wakefulness in laboratory animals.
induces an increase of W and a suppression of REMS in laboratory
W SWS REMS Ref. animals. Presently, there are no data on the effect of selective 5-HT2A
Mutant mice that do not express 5-HT2A/2C receptors  n.s. 92,93 agonists on the sleep-wake cycle. The injection of non-selective and
Non-selective 5-HT2A/2C receptor agonists (DOI, DOM) selective 5-HT2A antagonists or a 5-HT2A receptor inverse agonist
Microinjection into the DRN n.s. n.s.  95 has been shown to consistently increase SWS and to reduce REMS. In
Microinjection into the LDT n.s. n.s.  98 contrast, the administration of selective 5-HT2C receptor antagonists
Systemic administration   76,99 induces inconsistent effects on sleep variables.
Selective 5-HT2C receptor agonist (RO 60-0175) n.s.  108
Is 5-HT directly responsible for the DOI- and RO 60-0175-
Non-selective 5-HT2A/2C receptor antagonists induced increase of W? Systemic administration of DOI inhibits the
Ritanserin, sertindole n.s.  76,102
ring of 5-HT neurons in the DRN and NE neurons in the LC. In
Ketanserin   103
addition, 5-HT and NE levels are diminished at postsynaptic sites.
Selective 5-HT2A antagonists The reduction of the ring rate of 5-HT and LC cells is reverted by
Volinanserin, pruvanserin   93,105
volinanserin and ketanserin, respectively.96,112 On the other hand,
5-HT2A inverse agonist the i.p. injection of DOI signicantly increases the extracellular
Nelotanserin   106 concentration of ACh in the medial prefrontal cortex and the dorsal
Abbreviations: DRN, dorsal raphe nucleus; LDT, laterodorsal tegmental nucleus; hippocampus, and of DA in the medial prefrontal cortex and the
W, wakefulness; SWS, slow wave sleep; REMS, REM sleep; n.s., non-signicant; , VTA of the rat. The effect of DOI on ACh and DA release is blocked by
increased; e, reduced.
volinanserin and the non-selective serotonin 5-HT2 receptor
antagonist LY-53,857, respectively.113e115 Thus, indirect evidence
tends to indicate that ACh and DA are involved, at least in part, in
reasonable hypothesis is that intra-DRN microinjection of DOI the DOI-induced increase of W.
activates GABAergic neurons projecting to the LDT/PPT which, in The effects of the non-selective 5-HT2 receptor antagonists
turn, inhibit cholinergic cells that promote REMS. ritanserin, ketanserin, seganserin and ICI-169,369, the selective
Amici et al.98 locally microinjected DOI or the 5-HT2 receptor 5-HT2A antagonist eplivanserin and the 5-HT2A inverse agonist
antagonist ketanserin into the LDT of rats. DOI signicantly nelotanserin have been studied on the sleep EEG of subjects with
decreased the number of REM episodes, whereas ketanserin normal sleep. The administration of ritanserin in the evening
induced the opposite effect. 5-HT2A/2C receptors are located not on increased SWS [stages 3 and 4 non-REM sleep (NREMS)] and
cholinergic cells but on GABAergic interneurons intermingled with reduced stage 2 sleep and REMS.116 After 2 weeks of ritanserin
mesopontine cholinergic cells which tends to explain the inhibitory treatment SWS remained increased. In addition, ritanserin discon-
effect of DOI on cholinergic LDT neurons and the reduction of REMS tinuation following daily administration for 8 weeks was not asso-
episodes. In can be tentatively proposed that the mechanisms ciated with the occurrence of withdrawal symptoms.117 On the other
involved in the suppression of REMS would be reinforced by the hand, the effects of ketanserin on sleep variables were restricted to
activation of 5-HT2 receptors expressed by GABAergic interneurons an increase of SWS and a reduction of stage 2 sleep.118 During
and projection neurons located in the LDT/PPT and the DRN, nighttime sleep after a nap in the evening, seganserin induced an
respectively. increase in SWS and an enhancement of power density in the delta
Systemic administration of the non-selective 5-HT2 receptor and theta frequencies during NREMS. Compound ICI-169,369 also
agonists DOI and DOM has been shown to reduce SWS and REMS, induced an increase of SWS,119 whereas eplivanserin increased SWS
and to augment W in the rat.95,99 Injection of the 5-HT2A/2C receptor and reduced stage 2 sleep. Analysis of the EEG power spectra showed
antagonists ritanserin, ketanserin, ICI-170,809 or sertindole at the an increase of delta activity and a reduction of spindle frequency
beginning of the light period induced a signicant increase of SWS activity after eplivanserin administration.120
and a reduction of REMS in the rat. Wakefulness was also dimin- The effects of nelotanserin on sleep in healthy subjects were
ished in most of these studies.76,100e103 More recently, the action of assessed using a postnap insomnia model. The duration of SWS and
the selective 5-HT2A receptor antagonists volinanserin and pru- of REMS latency were signicantly increased by treatment with the
vanserin and of the 5-HT2A inverse agonist nelotanserin on sleep 5-HT2A inverse agonist. In addition, the number of stage shifts,
variables was assessed in rats and mice. Systemic injection of the number of awakenings and number of bouts of sleep were dimin-
5-HT2A antagonists or the 5-HT2A inverse agonist signicantly ished, whereas the duration of bouts of sleep was signicantly
increased SWS and reduced W and REMS.93,104e107 Furthermore, increased.106
pruvanserin prevented the DOI induced increase in W and reduc- Thus, agents that act by competition for agonist binding sites (5-
tion in SWS. However, REMS remained suppressed, which tends to HT2A receptor antagonists) or stabilize the receptor in its inactive
indicate that the effect of DOI is not restricted to the 5-HT conformation (5-HT2A receptor inverse agonists) consistently
system.104 increase SWS in subjects with normal sleep. The mechanism
The actions of the selective 5-HT2C receptor agonist RO 60-0175 involved in the increase of SWS is presently unclear. Possible
on sleep-wake stages have been evaluated in the rat. The contributing factors include the inhibition of cell groups involved in
compound provoked an increase of W accompanied by a reduction the occurrence of W, and the activation of GABAergic, MCHergic
of REMS108 (Table 3). Oral administration of the selective 5-HT2C and galaninergic neurons.
receptor antagonist SB-243213 signicantly increased SWS and
reduced REMS during the light period.109 However, with subcuta- The 5-HT3 receptor
neous SB-243213 only a signicant reduction of time spent in REMS
was seen.104 On the other hand, intraperitoneal injection of the The microinjection of the 5-HT3 receptor agonist m-chlor-
5-HT2B antagonist SB-242084 at light onset increased W and ophenylbiguanide (m-CPBG) into the DRN caused a reduction of
reduced SWS in the rst hour.110 Similarly, systemic administration REMS. Pretreatment with the 5-HT3 antagonist ondansetron pre-
of the 5-HT2B antagonist SB-215505 augmented W and decreased vented the m-CPBG-induced suppression of REMS.121 Concerning
SWS and REMS in mice and rats.111 the mechanism involved in the reduction of REMS by m-CPBG, it
276 J.M. Monti / Sleep Medicine Reviews 15 (2011) 269e281

Table 4 On the other hand, no signicant changes of values corresponding


The role of serotonin 5-HT3, 5-HT6 and 5-HT7 receptors in the regulation of sleep and to sleep variables were observed following the injection of RO-
wakefulness in laboratory animals.
4368554 during the dark phase.107 A series of neurochemical
W SWS REMS Ref. studies have ascertained the inuence of 5-HT6 receptor blockade
5-HT3 receptor on various neurotransmitter systems. Accordingly, using in vivo
Selective 5-HT3 receptor agonist (m-chlorophenylbiguanide) microdialysis in the freely moving rat, it has been established that
Microinjection into the DRN n.s. n.s.  121
blockade of the 5-HT6 receptor with SB-271046 or RO-046790
Injection into the left lateral ventricle   125
increases GLU and ACh levels in the frontal cortex and hippo-
5-HT6 receptor campus, and NE and DA concentrations in the prelimbic/infralimbic
Selective 5-HT6 receptor antagonists (SB-399885, RO-4368554)
Systemic administration   107
subregions of the medial prefrontal cortex of the rat.129e131 It
remains to be established whether the enhancement of W and
5-HT7receptor
reduction of SWS and REMS after administration of 5-HT6 receptor
Mutant mice that do not express 5-HT7 receptors n.s. n.s.  132
Selective 5-HT7 receptor agonists (LP-44) antagonists is a consequence of the increase of ACh, GLU, NE and DA
Microinjection into the DRN n.s. n.s.  133 levels at critical sites in the SNC or is an independent event.
Abbreviations: DRN, dorsal raphe nucleus; LDT, laterodorsal tegmental nucleus;
mPRF, medial pontine reticular formation; ACh, acetylcholine; GLU, glutamate; W, The 5-HT7 receptor
wakefulness; SWS, slow wave sleep; REMS, REM sleep; n.s., non-signicant; ,
increased; , reduced. Hedlund et al.132 have characterized sleep variables in 5-HT7
receptor knock-out mice. The mutants spent less time in REMS than
has been shown that the 5-HT3 receptor agonist 2-methyl-5-HT
their wild-type counterparts, mainly during the light period. There
increases the electrically-induced tritium overow from rat
were no differences between the 5-HT7 receptor knock-out mice
midbrain slices containing the raphe nuclei loaded with [3H]
and the wild type animals with respect to W and SWS duration.
5-HT.122 Thus, the m-CPBG-induced suppression of REMS would
The effects of LP-44, a selective 5-HT7 receptor agonist, micro-
depend on the increase of 5-HT in areas relevant to the promotion
injected directly into the DRN during the light period were studied
and maintenance of the behavioral state. The activation of seroto-
in adult rats. Infusion of LP-44 induced a signicant reduction of
nergic cells by m-CPBG could be a direct effect; on the other hand, it
REMS and of the number of REM periods. Pretreatment with the
could be an indirect action dependent on the inuence of a modu-
5-HT7 receptor antagonist SB-269970 averted the LP-44 suppres-
latory system on 5-HT neurons. In this respect, there is evidence in
sion of REMS.133 It was tentatively proposed that the activation by
favour of a role for the glutamatergic system in the 5-HT3 receptor-
LP-44 of projection GABAergic neurons located in the DRN could be
induced suppression of REMS. Glutamatergic neurons have been
responsible for the inhibition of cholinergic cells in the LDT/PPT and
characterized in the DRN; in addition, glutamatergic inputs to the
the decrease of REMS. However, the evidence supporting this
DRN have been described that originate in the brain stem, several
assertion is currently lacking.
hypothalamic nuclei, and the cerebral cortex.123 To date no
Intraperitoneal administration of the 5-HT7 receptor agonist LP-
attempts have been made to determine the effect of serotonin
211 during the light phase signicantly increased W and reduced
5-HT3 receptor activation on GLU release in the DRN. However,
REMS and the number of REM periods in the rat. Pretreatment with
Funahashi et al.124 observed that the application of 5-HT or the
SB-269970 prevented the action of LP-211 on the sleep-wake cycle
5-HT3 agonist phenylbiguanide increased the frequency of spon-
(Monti and Jantos, unpublished observations). As mentioned
taneous excitatory postsynaptic currents in neurons from the rat
earlier, 5-HT depolarizes neurons of the BNST and this effect is
area postrema in vitro, and this effect was prevented by the 5-HT3
antagonized by SB-269970 (Table 4). Thus, the LP-211 activation of
antagonist ICS-205,930 and the alpha-amino-3-hydroxy-5-methyl-
5-HT7 receptors expressed by neurons of the BFB could be partly
4-isoxazole propionate (AMPA) antagonist CNQX, thus indicating
responsible for the increase of W.
that synaptic currents are glutamatergic and not serotonergic. We
have found, in addition, that microinjection of the N-methyl-D-
Clinical context and perspective
aspartate (NMDA) receptor antagonist D-AP5 into the DRN
prevents the m-CPBG-decrease of REMS in the rat (Monti and
Ritanserin has been administered to poor sleepers, patients with
Jantos, unpublished observations).
chronic primary insomnia and psychiatric patients with a general-
m-CPBG injected into the left lateral ventricle increased W and
ized anxiety disorder (GAD) or a mood disorder. Ritanserin 5 mg
REMS latency, whereas SWS, REMS, and the number of REM periods
taken by poor sleepers for 20 days caused a large and signicant
were reduced in the rat.125 In contrast, systemic administration of
increase in SWS during the early and the late drug period compared
the 5-HT3 antagonists MDL-72222 and ondansetron signicantly
to baseline placebo nights. Concomitantly with the increase in SWS,
increased SWS and/or REMS, respectively.125,126 Pretreatment with
there was a reduction in stage 2 sleep and in the frequencies of
MDL-72222 prevented the increase of W and reduction of SWS and
awakenings.134 The administration of ritanserin 10 mg during the
REMS induced by m-CPBG.125 On the basis of a series of neuro-
morning for 5 days to a group of patients with chronic primary
pharmacological studies, it has been proposed that changes of sleep
insomnia increased the duration of SWS without modifying stage 2
variables after i.c.v. administration of m-CPBG are partly related to
sleep or REM sleep.135 The effect of ritanserin was additionally
the increased availability of DA at central sites127,128 (Table 4).
characterized in abstinent alcoholic patients with comorbid
insomnia. The 5-HT2 antagonist was given at a daily dose of 10 mg
The 5-HT6 receptor for 28 nights. The increase of total sleep time was associated with
greater amounts of NREMS, whereas SWS and REMS were not
Not much is known about the role of the 5-HT6 receptor in the notably modied.136 The acute effects of ritanserin 5 mg on sleep
regulation of sleep and W. We have found that systemic adminis- variables in patients with GAD and matched healthy controls was
tration of the selective 5-HT6 receptor antagonists SB-399885 and also examined. Ritanserin produced a signicant increase in SWS
RO-4368554 during the light phase causes an increase of W and together with a reduction in stage 1 sleep and wake time after sleep
a reduction of SWS and REMS in the rat (Table 4). The injection of onset.137 Polysomnographic recordings of dysthymia patients who
SB-399885 during the dark period induced also a decrease of REMS. received ritanserin 10 mg for 4 weeks showed a signicant increase
J.M. Monti / Sleep Medicine Reviews 15 (2011) 269e281 277

in SWS. No other variables were modied by the drug.138 Moreover, instances. Presently available evidence tends to indicate that the
acute administration of ritanserin 5 mg in patients with a diagnosis GABAB receptor is involved in the decrease of REMS. Accordingly,
of major depression induced a signicant increase in SWS without microinjection of the selective GABAB receptor agonist baclofen
changing stage 2 sleep or REMS duration.139 into the pedunculopontine tegmental nucleus (PPT) reduces REMS
The effects of the selective 5-HT2A receptor antagonists voli- in the rat,143 whereas the GABAA agonist muscimol induces the
nanserin, eplivanserin and pruvanserin, and of the 5-HT2A receptor opposite effect through a still unknown mechanism.144,145
inverse agonists APD-125 and pimavanserin have been studied in Although the process by which activation of 5-HT receptors
patients with chronic primary insomnia. However, with the facilitates the state of W is still unknown, it should be mentioned
exception of APD-125, no clinical data have yet been released that many of the GABAergic cells in the BFB, hippocampus and
(ClinicalTrials.gov Identier: NCT00664664). APD-125 (10 and neocortex on which 5-HT1A and 5-HT1B receptors are expressed, are
40 mg) given for 7 days decreased wake time after sleep onset and hyperpolarized by serotonin released from the DRN.146e148 Thus,
the number of nocturnal awakenings while the latency to persis- activation of 5-HT1A and 5-HT1B receptors may attenuate GABAergic
tent sleep remained unchanged. Slow wave sleep showed a signif- input and thereby indirectly increase the release of ACh and GLU at
icant dose-dependent increase.140 cortical and subcortical sites.
Several benzodiazepine (BZD) and non-BZD (modied-release Activation of 5-HT2A and 5-HT2C receptors enhances the release
zolpidem, zopiclone, eszopiclone, modied-release indiplon) of ACh in the medial-prefrontal cortex and the hippocampus, and of
hypnotics improve sleep induction and sleep maintenance. DA in the medial-prefrontal cortex and the VTA of the rat. Thus, the
However, BZD derivatives induce a further reduction of SWS and increased availability of DA and ACh at central sites after 5-HT2A/2C
REMS, whereas SWS and REMS values remain decreased during receptor activation could be responsible, at least in part, for the
non-BZD administration. Thus, the association of a 5-HT2A receptor increased incidence of W.
antagonist or a 5-HT2A receptor inverse agonist with a BZD or Not much is known about the mechanisms subserving the
a non-BZD hypnotic could be a valid alternative to normalize SWS increase of W and reduction of REMS after activation of 5-HT3 and
in patients with primary or comorbid insomnia.141 5-HT7 receptors. The 5-HT3 receptor is well known for stimulating
the release of 5-HT, NE, DA, HA, GLU, and GABA in the brain stem,
Conclusions the limbic system, the BFB, and the cortex, which could tentatively
explain its disruption of sleep variables. However, further studies
Attempts to characterize the role of serotonin receptors on sleep are needed to resolve this issue.
variables have been limited to the 5-HT1A, 5-HT1B, 5-HT2A, 5-HT2B,
5-HT2C, 5-HT3, 5-HT6 and 5-HT7 receptors. Most studies have Limitations of the approaches used to study the role of 5-HT
examined the effect of systemic administration of selective and in the regulation of W and REMS
relatively selective agonists and antagonists on sleep and W in the
rat. Recently, results from several studies have quantied the The selectivity of the knock-out technology has been empha-
spontaneous sleep/waking cycles in serotonin 5-HT1A, 5-HT1B, sized on the grounds of a more specic impairment of 5-HT
5-HT2A, 5-HT2C or 5-HT7 receptor knock-out mice. Much less is receptors than the neuropharmacological approach.149 However,
known about the effect of local microinjection of serotonin receptor the proposal would apply almost exclusively to mutant mice that
ligands into CNS structures relevant to the regulation of sleep and do not express 5-HT1A or 5-HT1B receptor. Accordingly, opposite to
W, including the DRN, the LDT/PPT, the mPRF, the BFB and the animals treated sistemically with selective 5-HT1A or 5-HT1B
VLPO. receptor agonists where REMS is suppressed, knock-out mice show
Steinbusch142 and Lowry et al.44 have shown that 5-HT cells in the an increase of the behavioral state. On the other hand, data
rat DRN appear in topographically organized groups. These obtained from 5-HT2A receptor knock-out mice do not conrm the
subpopulations of neurons differ in their morphological character- ndings generated by selective 5-HT2A receptor antagonists in as
istics, cellular properties, and afferent and efferent connections.39,43 much as mutant mice show an increase of W and a reduction of
In addition, the subpopulations of 5-HT neurons coexpress different SWS. This outcome would be related to the limited specicity of
neurotransmitters and neuromodulators including DA, GLU, GABA, presently available knock-out technology. Hence, in knock-out mice
substance P, and corticotropin releasing factor.41,42,142 Thus, the the 5-HT2A receptor is absent not only in neural structures that
possibility exists that efferent projections from each subpopulation participate in the regulation of SWS but also in brain regions
of 5-HT neurons form synapses with only one subtype of serotonin involved in the regulation of W. Thus, the absence of 5-HT2A
receptor. This would allow different 5-HT subsystems to inuence receptors in GABAergic cells that control the release of DA in the
specically and separately the neurons involved in the regulation of prefrontal cortex and the nucleus accumbens and of NE in
W and REMS. the hippocampus, respectively, signicantly increases the release of
The characterization of subpopulations of 5-HT neurons adds to the monoamines at postsynaptic sites. As a result W is augmented
the appreciation that each 5-HT receptor family makes use of and SWS is decreased. An unexpected picture has emerged also
a particular signal transduction pathway. Accordingly, the 5-HT1 with respect to the 5-HT7 receptor, such that REMS is suppressed in
family, like the 5-HT5 receptor, are negatively coupled to adenylate both mutant mice and animals administered a selective 5-HT7
cyclase. On the other hand, the 5-HT2 family is coupled to phos- receptor agonist. It is proposed that the changes of the sleep-wake
pholipase C whereas the 5-HT4, 5-HT6 and 5-HT7 receptors are cycle in knock-out mice would depend not only on the receptor
positively coupled to adenylate cyclase. As an exception, the 5-HT3 involved but also on the type of cell (excitatory or inhibitory) that
receptor incorporates an ion channel that regulates ion ux in a G- expresses the receptor and on its location in the CNS.
protein-independent manner.37 Adrien150 has pointed out that the technology of inducible
Activation of the postsynaptic 5-HT1A and 5-HT1B receptors knock-outs, or lentiviral vectors applied directly into selective brain
induces the hyperpolarization of target neurons in the LDT/PPT and structures will help to overcome the limitations of presently
the mPRF. On the other hand, activation of the 5-HT2A, 5-HT2C and available techniques.
5-HT7 receptors leads to the depolarization of GABAergic cells that Presently available 5-HT receptor agonists tend to increase W
innervate the structures involved in the promotion and the and to reduce REMS. While 5-HT2A and 5-HT3 receptor antagonists
induction of REMS. The behavioral state is suppressed during both increase SWS and/or REMS, opposite effects have been described
278 J.M. Monti / Sleep Medicine Reviews 15 (2011) 269e281

following the administration of 5-HT1A, 5-HT1B, 5-HT6 and 5-HT7 9. Laguzzi RF, Adrien J. Inversion de linsomnie produit par la chlor-
ophenylalanine chez le chat. Arch Ital Biol 1980;118:109e23.
antagonists. The so called competitive antagonism, a characteristic
10. Tenen S. The effects of p-chlorophenylalanine, a serotonin depletor on
pattern of antagonism, refers to a drug that lacks intrinsic efcacy avoidance adquisition, pain sensitivity and related behavior in the rat.
but retains afnity to compete with the agonist for the binding Psychopharmacology 1967;10:204e19.
site.151 Although selective 5-HT receptor antagonists compete with 11. Tagliamonte A, Tagliamonte P, Gessa G, Brodie B. Compulsive sexual activity
induced by p-chlorophenylalanine in normal and pinealectomized male
the agonists for the binding site, they do not discriminate between rats. Science 1969;166:1433e5.
neural structures with dissimilar functions. In addition, their 12. Sheard MH. The effects of p-chlorophenylalanine on behavior in rats:
afnity for a given 5-HT receptor subtype tends to vary during the relation to brain serotonin and 5-hydroxyindolacetic acid. Brain Res
1969;15:524e8.
light-dark cycle.107 The limitations inherent to the use of presently 13. Borbly AA, Huston JP, Waser PG. Physiological and behavioral effects of
available antagonists will be partly resolved when selective ligands parachlorophenylalanine in the rat. Psychopharmacology 1973;31:131e42.
become available that act at specic sites in the CNS 14. Dement WC, Mitler MM, Henriksen SJ. Sleep changes during chronic
administration of parachlorophenylalanine. Can J Biol 1972;31:239e46.
15. Tobler I, Borbly AA. Sleep regulation after reduction of brain serotonin:
effect of p-chlorophenylalanine combined with sleep deprivation in the rat.
Sleep 1982;5:145e53.
Practice points
16. Jouvet M. Neuromediateurs et facteurs hypnognes. Rev Neurol
1984;140:389e400.
1) All serotonin receptor agonists studied to date share the 17. Imeri L, Mancia M, Bianchi S, Opp MR. 5-Hydroxytryptophan, but not
ability to promote W and to suppress REMS when given l-tryptophan, alters sleep and brain temperature in rats. Neuroscience
systemically or by the i.c.v. route. 2000;95:445e52.
18. Imeri L, Bianchi S, Opp MR. Antagonism of corticotropin-releasing hormone
2) Microinjection of 5-HT1A agonists into the DRN
alters serotonergic-induced changes in brain temperature, but not sleep, of
increases REMS, whereas local administration of 5- rats. Am J Physiol Regul Integr Comp Physiol 2005;289:R1116e1123.
HT1B, 5-HT2A/2C, 5-HT3 and 5-HT7 agonists induces the 19. Morrow JD, Vikraman S, Imeri L, Opp MR. Effects of serotonergic activation
opposite effect. by5-hydroxytryptophan on sleep and body temperature of C57BL/6J and
3) Systemic administration of non-selective 5-HT2A/2C interleukin-6-decient mice are dose and time related. Sleep
2008;31:21e33.
antagonists, selective 5-HT2A antagonists or 5-HT2A
20. Khateb A, Fort P, Alonso A, Jones BE, Mhlthaler M. Pharmacological and
inverse agonists increases SWS in laboratory animals, immunohistochemical evidence for serotonergic modulation of cholinergic
subjects with normal sleep and patients with primary or nucleus basalis neurons. Eur J Neurosci 1993;5:541e7.
comorbid insomnia. 21. Levita L, Hammack SE, Mania I, Li XY, Davis M, Rainnie DG. 5-hydroxy-
tryptamine1A-like receptor activation in the bed nucleus of the stria ter-
minalis: electrophysiological and behavioral studies. Neuroscience
2004;128:583e96.
22. Pace-Schott EF, Hobson JA. Basic mechanisms of sleep: New evidence on
the neuroanatomy and neuromodulation of the NREMeREM cycle. In:
Research agenda
Charney D, Nemeroff C, editors. Neuropsychopharmacology e the fth
generation of progress. Philadelphia: Lippincott: Williams and Wilkins;
1) Evaluation of the effects of systemic and local admin- 2002. p. 1859e77.
istration of selective 5-HT4, 5-HT5 and 5-HT6 receptor *23. Jones BE. Arousal systems. Front Biosci 2003;8:438e51.
agonists on the sleep-wake cycle. *24. Baghdoyan HA, Lydic R. Neurotransmitters and neuromodulators regu-
lating sleep. In: Bazil CW, Marlow BA, Sammaritano MR, editors. Sleep
2) Studies aimed at determining the mechanisms involved
and epilepsy: the clinical spectrum. Amsterdam: Elsevier Science; 2002.
in 5-HT receptor subtypes-induced increase of W and p. 17e44.
suppression of REMS. 25. Szymusiak R, Gvilia I, McGinty D. Hypothalamic control of sleep. Sleep Med
3) Research is needed to better understand the influence 2007;8:291e301.
of 5-HT receptor subtypes on the thalamus, BFB and 26. Lagos P, Torterolo P, Jantos H, Chase MH, Monti JM. Effects on sleep of
melanin-concentrating hormone (MCH) microinjections into the dorsal
cerebral cortex.
raphe nucleus. Brain Res 2009;1265:103e10.
27. Manfridi A, Brambilla D, Bianchi S, Mariotti M, Opp MR, Imeri L. Inter-
leukin1b enhances non-rapid eye movement sleep when microinjected
into the dorsal raphe nucleus and inhibits serotonergic neurons in vitro. Eur
J Neurosci 2003;18:1041e9.
References 28. Brambilla D, Franciosi S, Opp MR, Imeri L. Interleukin-1 inhibits ring of
serotonergic neurons in the dorsal raphe nucleus and enhances GABAergic
1. Bandelow B, Zohar J, Hollander E, Kasper S, Moller HJ. World Federation of inhibitory post-synaptic potentials. Eur J Neurosci 2007;26:1862e9.
Societies of Biological Psychiatry (WFSBP) guidelines for the pharmaco- 29. Vanni-Mercier G, Sakai K, Lin JS, Jouvet M. Mapping of cholinoceptive
logical treatment of anxiety, obsessive-compulsive and post-traumatic brainstem structures responsible for the generation of paradoxical sleep in
stress disorders e rst revision. World J Biol Psychiatry 2008;9:248e312. the cat. Arch Ital Biol 1989;127:133e64.
2. Jouvet M. The role of monoamines and acetylcholine containing neurons in 30. Yamamoto K, Mamelak AN, Quattrochi JJ, Hobson JA. A cholinoceptive
the regulation of the sleepewaking cycle. Ergeb Physiol 1972;64:166e307. desynchronized sleep induction zone in the anterodorsal pontine
3. Trulson ME, Jacobs BL. Raphe unit activity in freely moving cats: correlation tegmentum: locus of the sensitive region. Neuroscience 1990;39:279e93.
with level of behavioral arousal. Brain Res 1979;163:135e50. 31. Reinoso-Surez F, de Andrs S, Rodrigo-Angulo ML, Garzn M. Brain
4. Wyatt RJ, Engelman K, Kupfer DJ, Scott J, Sjoerdsma A, Snyder F. Effects of structures and mechanisms involved in the generation of REM sleep. Sleep
para-chlorophenylalanine on sleep in man. Electroencephalogr Clin Neuro- Med Rev 2001;5:63e77.
physiol 1969;27:529e32. 32. Hobson JA, Stickgold R, Pace-Schott EF. The neuropsychology of REM sleep
5. Rechtschaffen A, Lovell RA, Freedman DX, Whitehead WE, Aldrich M. The dreaming. Neuroreport 1998;9:R1e14.
effect of para-chlorophenylalanine in the rat: some implications for the 33. Steiniger TL, Rye DB, Wainer BH. Serotonergic dorsal raphe nucleus
serotonin-sleep hypothesis. In: Barchas J, Usin E, editors. Serotonin and projections to the cholinergic and noncholinergic neurons of the pedun-
behavior. New York: Academic Press; 1973. p. 401e18. culopontine tegmental region: a light and electron microscopic antero-
6. Ross CA, Trulson ME, Jacobs BL. Depletion of brain 5-HT following intra- grade tracing and immunohistochemical study. J Comp Neurol
ventricular 5,7-dihydroxytryptamine fails to disrupt sleep in the rat. Brain 1997;382:302e22.
Res 1976;114:517e23. 34. Semba K. Aminergic and cholinergic afferents to REM sleep induction
7. Clancy JJ, Caldwell DF, Oberleas D, Sangiah S, Villeneuve MJ. Effects of regions of the pontine reticular formation in the rat. J Comp Neurol
chronic tryptophan dietary deciency on the rats sleep-wake cycle. Brain 1993;330:543e56.
Res Bull 1978;3:83e7. 35. Van Bockstaele EJ, Biswas A, Pickel VM. Topography of serotonin neurons in
8. Mouret J, Bobillier P, Jouvet M. Insomnia following parachlorophenylalanine the dorsal raphe nucleus that send axon collaterals to the rat prefrontal
in the rat. Eur J Pharmacol 1968;5:17e22. cortex and nucleus accumbens. Brain Res 1993;624:188e98.
36. Janowski MP, Sessack SR. Prefrontal cortical projections to the rat dorsal
raphe nucleus: ultrastructural features and association with sero-
tonin and gamma-aminobutyric acid neurons. J Comp Neurol
* The most important references are denoted by an asterisk. 2004;468:518e29.
J.M. Monti / Sleep Medicine Reviews 15 (2011) 269e281 279

37. Cooper JR, Bloom FE, Roth RH. The biochemical basis of neuropharmacology. 63. Gustafson EL, Durkin MM, Bard JA, Zgombick J, Brancheck TA. A receptor
New York: Oxford University Press; 1996. autoradiographic and in situ hybridization analysis of the distribution of
38. Stanford SC. 5-Hydroxytryptamine. In: Webster RA, editor. Neurotrans- the 5-HT7 receptor in rat brain. Br J Pharmacol 1996;117:657e66.
mitters, drugs and brain function. Chichester: John Wiley & Sons; 2001. 64. Hammack SE, Guo JD, Hazra R, Dabrowska J, Myers KM, Rainnie DG. The
p. 187e209. response of neurons of the bed nucleus of the stria terminalis to serotonin:
39. Vertes RP, Linley SB. Efferent and afferent connections of the dorsal and implications for anxiety. Prog Neuropsychopharmacol Biol Psychiatry
median raphe nuclei in the rat. In: Monti JM, Pandi-Perumal SR, Jacobs BL, 2009;33:1309e20.
Nutt DJ, editors. Serotonin and sleep: molecular, functional and clinical 65. Vandermaelen CP, Aghajanian GK. Electrophysiological and pharmacolog-
aspects. Basel: Birkhuser Verlag; 2008. p. 69e102. ical characterization of serotonergic dorsal raphe neurons recorded
40. Jacobs BL, Azmitia EC. Structure and function of the brain serotonin system. extracellularly and intracellularly in rat brain slices. Brain Res
Physiol Rev 1992;72:165e229. 1983;289:1009e19.
41. Charara A, Parent A. Chemoarchitecture of the primate dorsal raphe 66. Allers KA, Sharp T. Neurochemical and anatomical identication of fast-
nucleus. J Chem Neuroanat 1998;15:111e27. and slow-ring neurons in the rat dorsal raphe nucleus using juxtacellular
42. Li YQ, Li H, Kaneko T, Mizuno N. Morphological features and electrophys- labelling methods in vivo. Neuroscience 2003;122:193e204.
iological properties of serotonergic and non-serotonergic projection 67. McGinty DJ, Harper RM. Dorsal raphe neurons: depression of ring during
neurons in the dorsal raphe nucleus. Brain Res 2001;900:110e8. sleep. Brain Res 1976;101:569e75.
43. Abrams JK, Johnson PL, Hollis JH, Lowry CA, et al. Anatomic and functional 68. Sakai K, Crochet S. Differentiation of presumed serotonergic dorsal raphe
topography of the dorsal raphe nucleus. Ann N Y Acad Sci 2004;1018:46e57. neurons in relation to behavior and wakeesleep states. Neuroscience
*44. Lowry CA, Evans AK, Gasser PJ, Hale MW, Staub DR, Shekhar A. Topographic 2001;104:1141e55.
organization and chemoarchitecture of the dorsal raphe nucleus and the *69. Boutrel B, Monaca C, Hen R, Hamon M, Adrien J. Involvement of 5-HT1A
median raphe nucleus. In: Monti JM, Pandi-Perumal S-R, Jacobs BL, Nutt DJ, receptors in homeostatic and stress-induced adaptive regulations of
editors. Serotonin and sleep: molecular, functional and clinical aspects. Basel: paradoxical sleep: studies in 5-HT1A knock-out mice. J Neurosci
Birkhuser Verlag; 2008. p. 25e62. 2002;22:4686e92.
*45. Hannon J, Hoyer D. Molecular biology of 5-HT receptors. In: Monti JM, Pandi- *70. Portas CM, Thakkar M, Rainnie D, McCarley RW. Microdialysis perfusion of
Perumal S-R, Jacobs BL, Nutt DJ, editors. Serotonin and sleep: molecular, 8-hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT) into the dorsal
functional and clinical aspects. Basel: Birkhuser Verlag; 2008. p. 155e82. raphe nucleus decreases serotonin release and increases rapid eye move-
46. Day HE, Greenwood BN, Hammack SE, Watkins LR, Fleshner M, Maier SF, ment sleep in the freely moving rat. J Neurosci 1996;16:2820e8.
et al. Differential expression of 5-HT1A, alpha(1B) adrenergic, CRF-R1, and 71. Monti JM, Jantos H, Monti D. Increased REM sleep after intra-dorsal raphe
CRF-R2 receptor mRNA in serotonergic, gamma-aminobutyric acidergic, nucleus injection of esinoxan or 8-OH-DPAT: prevention with WAY
and catecholaminergic cells of the rat dorsal raphe nucleus. J Comp Neurol 100635. Eur Neuropsychopharmacol 2002;12:47e77.
2004;474:464e78. 72. Srensen E, Grnli J, Bjorvatn B, Ursin R. Sleep and waking following
47. Kia HK, Brisorgueil MJ, Hamon M, Calas A, Verg D. Ultrastructural locali- microdialysis perfusion of the selective 5-HT1A antagonist p-MPPI into the
zation of 5 -hydroxytryptamine1A receptors in the rat brain. J Neurosci Res dorsal raphe nucleus of the freely moving rat. Brain Res 2001;897:122e30.
1996;46:697e708. 73. Sanford LD, Ross RJ, Seggos AE, Morrison AR, Ball WA, Mann GL. Central
48. Clark MS, McDevitt RA, Neumaier JF. Quantitative mapping of tryptophan administration of two 5-HT receptor agonists: effect on REM sleep initia-
hydroxylase-2, 5-HT1A, 5-HT1B and serotonin transporter expression across tion and PGO waves. Pharmacol Biochem Behav 1994;49:93e100.
the anteroposterior axis of the rat dorsal and median raphe nuclei. J Comp 74. Monti JM, Jantos H. Differential effects of the 5-HT1A receptor agonist
Neurol 2006;498:611e23. esinoxan given locally or systemically on REM sleep in the rat. Eur J
49. Guo JD, Rainnie DG. Presynaptic 5-HT1B receptor-mediated serotonergic Pharmacol 2003;478:121e30.
inhibition of glutamate transmission in the bed nucleus of the stria ter- 75. Monti JM, Jantos H. Dose-dependent effects of the 5-HT1A receptor agonist
minalis. Neuroscience 2010;165:1390e401. 8-OHDPAT on sleep and wakefulness in the rat. J Sleep Res 1992;1:169e75.
50. Bruivels AT, Landwehrmeyer B, Gustafson EL, Durkin MM, Mengod G, 76. Monti JM, Orellana C, Boussard M, Jantos H, Labraga P, Olivera S, et al. 5-HT
Branchek K, et al. Localization of 5-HT1B, 5-HT1Da, 5-HT1E and 5-HT1F receptor agonists 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI)
receptor messenger mRNA in rodent and primate brain. Neuropharma- and 8-OH-DPAT increase wakefulness in the rat. Biogen Amines
cology 1994;33:367e86. 1990;7:145e51.
51. Sari Y, Miquel MC, Brisorgueil MJ, Ruiz G, Doucet E, Hamon M, et al. Cellular 77. Monti JM, Jantos H, Silveira R, Reyes-Parada M, Scorza C, Prunell G.
and subcellullar localization of 5-hydroxytryptamine1B receptors in the rat Depletion of brain serotonin by 5,7-DHT: effects on the 8-OH-DPAT-
central nervous system: immunocytochemical, autoradiographic and lesion induced changes of sleep and waking in the rat. Psychopharmacology
studies. Neuroscience 1999;88:899e915. 1994;115:273e7.
52. Abramowski D, Rigo M, Duc D, Hoyer D, Staufenbiel M. Localization of the 78. Lerman JA, Kaitin KI, Dement WC, Peroutka SJ. The effect of buspirone on
5-hydroxytryptamine2C receptor protein in human and rat brain using sleep in the rat. Neurosci Lett 1986;72:64e8.
specic antisera. Neuropharmacology 1995;34:1635e45. 79. Monti JM, Jantos H, Silveira R, Reyes-Parada M, Scorza C. Sleep and waking
53. Cornea-Hbert V, Riad M, Wu C, et al. Cellular and subcellular distribution in 5,7-DHT-lesioned or ()pindolol-pretreated rats after administration of
of the serotonin 5-HT2A receptor in the central nervous system of the adult buspirone, ipsapirone or gepirone. Pharmacol Biochem Behav
rat. J Comp Neurol 1999;409:187e209. 1995a;52:305e12.
54. Clemett DA, Punhani T, Duxon MS, et al. Immunohistochemical localization 80. Seidel WF, Cohen SA, Bliwise NG, Dement WC. Buspirone: and anxiolytic
of the 5-HT2C receptor protein in the rat CNS. Neuropharmacology without sedative effect. Psychopharmacology 1985;87:371e3.
2000;39:123e32. 81. Manfredi RL, Kales A, Vgontzas AN, Bixler EO, Isaac MA, Falcone CM.
55. Bonhaus DW, Bach C, Desouza A, et al. The pharmacology and distribution Buspirone: sedative or stimulant effect? Am J Psychiatry 1991;148:1213e7.
of human 5-hydroxytryptamine(2B) (5-HT2B) receptor gene products e 82. Gillin JC, Jermajczyk W, Valladares-Neto DC, Golshan S, Lardon M, Stahl SM.
comparison with 5.HT2A and 5-HT2C receptors. Br J Pharmacol Inhibition of REM sleep by ipsapirone, a 5-HT1A agonist, in normal
1995;115:622e8. volunteers. Psychopharmacology 1994;116:433e6.
*56. Guo JD, Hammack SE, Hazra R, Levita L, Rainnie DG. Bi-directional modu- 83. Armitage R. Effects of antidepressant treatment on sleep EEG in depression.
lation of bed nucleus of stria terminalis neurons by 5-HT: molecular J Psychopharmacol 1996;10(Suppl. 1):22e5.
expression and functional properties of excitatory 5-HT receptor subtypes. 84. Staner L, Luthringer R, Macher JP. Effects of antidepressant drugs on sleep
Neuroscience 2009;164:1776e93. EEG in patients with major depression. Cent Nerv Syst Drugs
57. Gehlert DR, Gackenheimer SL, Wong DT, et al. Localization of 5-HT3 1999;11:49e60.
receptors in the rat using [3H]LY278584. Brain Res 1991;553:149e54. 85. Oberndorfer S, Saletu-Zyhlarz G, Saletu B. Effects of selective serotonin
58. Morales M, Battenberg E, Bloom FE. Distribution of neurons expressing reuptake inhibitors on objective and subjective sleep quality. Neuro-
immunoreactivity for the 5-HT3 receptor subtype in the rat brain and psychobiology 2000;42:69e81.
spinal cord. J Comp Neurol 1998;402:385e401. 86. Monti JM, Jantos H. A study of the brain structures involved in the acute
59. Laporte AM, Koscielniak T, Ponchant M, Verg D, Hamon M, Gozlan H. effects of uoxetine on REM sleep in the rat. Int J Neuropsychopharmacol
Quantitative autoradiographic mapping of 5-HT3 receptors in the rat CNS 2005;8:75e86.
using [125I]iodo-zacopride and [3H]zacopride as radioligands. Synapse 87. Boutrel B, Franc B, Hen R, Hamon M, Adrien J. Key role of 5-HT1B receptors
1992;10:2771e81. in the regulation of paradoxical sleep as evidenced in 5-HT1B knock-out
60. Ruat M, Traiffort E, Arrang JM, Tardivel-Lacombe J, Diaz J, Leurs R, et al. mice. J Neurosci 1999;19:3204e12.
A novel rat serotonin (5-HT6) receptor - molecular cloning, localization and 88. Monti JM, Jantos H, Lagos P. Activation of serotonin 5-HT1B receptor in the
stimulation of cAMP accumulation. Biochem Biophys Res Comm dorsal raphe nucleus affects REM sleep in the rat. Behav Brain Res
1993;193:268e76. 2010;206:8e16.
61. Roberts JC, Reavill C, East SZ, Harrison PJ, Patel S, Routledge C, et al. The 89. Lemos JC, Pan YZ, Ma X, Lamy C, Akanwa AC, Beck SG. Selective 5-HT1B
distribution of 5-HT6 receptors in rat brain: an autoradiographic binding receptor inhibition of glutamatergic and gabaergic synaptic activity in the
study using the radiolabelled 5-HT6 receptor antagonist [125I]SB-258585. rat dorsal and median raphe. Eur J Neurosci 2006;24:3415e30.
Brain Res 2002;934:49e57. 90. Bjorvatn B, Ursin R. Effects of the selective 5-HT1B agonist CGS 12066B, on
62. Woolley ML, Marsden CA, Fone KCF. 5-HT6 receptors. Curr Drug Targets CNS sleep/waking stages and EEG power spectrum in rats. J Sleep Res
Neurol Disord 2004;3:59e79. 1994;3:97e105.
280 J.M. Monti / Sleep Medicine Reviews 15 (2011) 269e281

91. Monti JM, Monti D, Jantos H, Ponzoni A. Effects of selective activation of the 117. Idzikowski C. The effects of ritanserin and seganserin on human slow wave
5-HT1B receptor with CP-94,253 on sleep and wakefulness in the rat. sleep. In: Wauquier A, Dugovic C, Radulovacki M, editors. Slow wave
Neuropharmacology 1995b;34:1647e51. sleep ephysiological, pathophysiological and functional aspects. New York:
92. Frank MG, Stryker MP, Tecott LH. Sleep and sleep homeostasis in mice Raven Press; 1989. p. 197e215.
lacking the 5-HT2C receptor. Neuropsychopharmacology 2002;27:869e73. 118. Sharpley AL, Elliott JM, Attenburrow MJ, Cowen PJ. Slow wave sleep in
93. Popa D, Lna C, Fabre V, Prenat C, Ginrich J, Escourrou P, et al. Contribution humans: role of 5-HT2A and 5-HT2C receptors. Neuropharmacology
of 5-HT2 receptor subtypes to sleep-wakefulness and respiratory control, 1994;33:467e71.
and functional adaptations in knock-out mice lacking 5-HT2A receptors. 119. Sharpley AL, Solomon RA, Fernando AI, da Roza Davis JM, Cowen PJ. Dose-
J Neurosci 2005;25:11231e8. related effects of selective 5-HT2 receptor antagonists on slow wave sleep in
94. Fink KB, Gthert M. 5-HT receptor regulation of neurotransmitter release. humans. Psychopharmacology 1990;101:568e9.
Pharmacol Rev 2007;59:360e417. 120. Abbas A, Roth BI. Pimavanserin tartrate: a 5-HT2A inverse agonist with
95. Monti JM, Jantos H. Effects of activation and blockade of 5-HT2A/2C recep- potential for treating various neuropsychiatric disorders. Expert Opin
tors in the dorsal raphe nucleus on sleep and waking in the rat. Prog Neuro- Pharmacother 2008;9:3251e9.
Psychopharmacol Biol Psychiatry 2006a;30:1189e95. 121. Monti JM, Jantos H. Activation of the serotonin 5-HT3 receptor in the dorsal
96. Garratt JC, Kidd EJ, Wright IK, Marsden CA. Inhibition of 5-hydroxytryp- raphe nucleus suppresses REM sleep in the rat. Prog Neuro-Psycho-
tamine neuronal activity by the 5-HT agonist, DOI. Eur J Pharmacol pharmacol Biol Psychiatry 2008a;32:9407.
1991;199:349e55. 122. Bagdy E, Solyom S, Harsing LG. Feedback stimulation of somatodendritic
97. Martin-Ruiz R, Puig MV, Celada P, Shapiro DA, Roth BL, Mengod G, et al. serotonin release: a 5-HT3 receptor-mediated effect in the raphe nuclei of
Control of serotonergic function in medial prefrontal cortex by serotonin- the rat. Brain Res Bull 1998;45:203e8.
2A receptors through a glutamate-dependent mechanism. J Neurosci 123. Lee HS, Kim MA, Valentino RJ, Waterhouse BD. Glutamatergic afferent
2001;21:9856e66. projections to the dorsal raphe nucleus of the rat. Brain Res
98. Amici R, Sanford LD, Kearney K, McInemey B, Ross RJ, Horner RL, et al. 2003;963:57e71.
A serotonergic (5-HT2) receptor mechanism in the laterodorsal tegmental 124. Funahashi M, Mitoh Y, Matsuo R. Activation of presynaptic 5-HT3 receptors
nucleus participates in regulating the pattern of rapid-eye-movement facilitates glutamatergic synaptic inputs to area postrema neurons in rat
occurrence in the rat. Brain Res 2004;996:9e18. brain slices. Methods Find Exp Clin Pharmacol 2004;26:615e22.
*99. Dugovic C, Wauquier A, Leysen JE, Janssen PAJ. Functional role of 5-HT2 125. Ponzoni A, Monti JM, Jantos H. The effects of selective activation of the 5-
receptors in the regulation of sleep and wakefulness in the rat. Psycho- HT3 receptor with m-chlorophenylbiguanide on sleep and wakefulness in
pharmacology 1989;97:436e42. the rat. Eur J Pharmacol 1993;249:259e64.
100. Dugovic C, Wauquier A. 5-HT2 receptors could be primarily involved in the 126. Adrien J, Tissier MH, Lanfumey L, Haj-Dahmane S, Jolas T, Franc B, et al.
regulation of slow wave sleep in the rat. Eur J Pharmacol 1987;137:145e6. Central action of 5-HT3 receptor ligands in the regulation of sleep-wake-
101. Tortella FC, Echevarria E, Pastel RH, Cox B, Blackburn TP. Suppressant effects fulness and raphe neuronal activity in the rat. Neuropharmacology
of selective 5-HT2 antagonists on rapid eye movement sleep in rats. Brain 1992;31:519e29.
Res 1989;485:294e300. 127. Ponzoni A, Monti JM, Jantos H, Altier H, Monti D. Increased waking after
102. Coenen AML, Ates N, Skarsfeldt T, Luijtelaar ELJM. Effects of sertindole on intra-accumbens injection of m-chlorophenylbiguanide: prevention with
sleepewake states, electroencephalogram, behavioral patterns, and serotonin or dopamine receptor antagonists. Eur J Pharmacol
epileptic activity in rats. Pharmacol Biochem Behav 1995;51:353e7. 1995;278:111e5.
103. Kirov R, Moyanova S. Age-dependent effect of ketanserin on the 128. Monti JM, Ponzoni A, Jantos H, Lagos P, Silveira R, Banchero P. Effects of
sleepewaking phases in rats. Int J Neurosci 1998;93:257e64. accumbens m-chlorophenylbiguanide microinjection on sleep and waking
*104. Monti JM, Jantos H. Effects of the serotonin 5-HT2A/2C receptor agonist DOI in intact and 6-hydroxydopamine-treated rats. Eur J Pharmacol
and of the selective 5-HT2A and 5-HT2C receptor antagonists EMD 281014 1999;364:89e98.
and SB-243213, respectively, on sleep and waking in the rat. Eur J Pharmacol 129. Dawson LA, Nguyen Q, Li P. In vivo effects of the 5-HT6 antagonist
2006b;553:163e70. SB-271046 on striatal and frontal cortex extracellular concentrations of
105. Morairty SR, Hedley L, Flores J, Martin R, Kilduff TS. Selective 5-HT2A and noradrenaline, dopamine, 5-HT, glutamate and aspartate. Br J Pharmacol
5-HT6 antagonists promote sleep in the rat. Sleep 2008;31:34e44. 2000;130:23e6.
106. Al-Shamma HA, Anderson C, Chuang E, Luthringer R, Grottick AJ, Hauser E, 130. Wolley ML, Marsden CA, Sleight J, Fone K. Reversal of a scopolamine-
et al. Nelotanserin, a novel selective human 5-hydroxytryptamine2A inverse induced decit in object discrimination by a selective 5-HT6 receptor
agonist for the treatment of insomnia. J Pharmacol Exp Ther antagonist, RO-046790, in rats. Br J Pharmacol 2000;129:64P.
2010;332:281e90. 131. Lacroix LP, Dawson LA, Hagan JJ, Heidbreder CA. 5-HT6 receptor antagonist
107. Monti JM, Jantos H. Effects of the 5-HT6 receptor antagonists SB-399885 SB-271046 enhances extracellular levels of monoamines in the rat medial
and RO-4368554 and of the 5-HT2A receptor antagonist EMD 281014 on prefrontal cortex. Synapse 2003;51:158e64.
sleep and wakefulness in the rat during both phases of the lightedark cycle. 132. Hedlund PB, Huitron-Resendiz S, Henriksen SJ, Sutcliffe JG. 5-HT7 inhibition
Behav Brain Res 2011;216:381e8. and inactivation induce antidepressantlike behavior and sleep pattern. Biol
108. Martin JR, Bss M, Jenck JL, Moreau J, Mutel V, Sleight AJ, et al. 5-HT2C Psychiatry 2005;58:831e7.
receptor agonists: pharmacological characteristics and therapeutic poten- 133. Monti JM, Leopoldo M, Jantos H. The serotonin 5-HT7 agonist LP-44
tial. J Pharmacol Exp Ther 1998;286:913e24. microinjected into the dorsal raphe nucleus suppresses REM sleep in the
109. Smith MI, Piper DC, Duxon MS, Upton N. Effect of SB-243213, a selective rat. Behav Brain Res 2008b;191:184e9.
5-HT2C receptor antagonist on the rat sleep prole: a comparison to 134. Adam K, Oswald I. Effects of repeated ritanserin on middle-aged poor
paroxetine. Pharmacol Biochem Behav 2002;33:467e71. sleepers. Psychopharmacology 1989;99:219e21.
110. Kantor S, Jakus R, Molnar E, Gyongyosi N, Toth A, Detari L, et al. Despite 135. Ruiz-Primo E, Haro R, Valencia M. Polysomnographic effects of ritanserin in
similar anxiolytic potential, the 5-hydroxytryptamine2C receptor antago- insomniacs. A crossed double-blind controlled study. Sleep Res 1989;19:72.
nist SB-242084 [6-chloro-5-methyl-1-[2-(2-methylpyrid-3-yloxy)-pyrid-5- 136. Monti JM, Alterwain P, Estvez F, Alvario F, Giusti M, Olivera S, et al. The
yl carbamoyl] indoline] and chlordiazepoxide produced differential effects effects of ritanserin on mood and sleep in abstinent alcoholic patients.
on electroencephalogram power spectra. J Pharmacol Exp Ther Sleep 1993;16:647e54.
2005;315:921e30. 137. da Roza Davies JM, Sharpley AL, Cowen PJ. Slow wave sleep and 5-HT2
111. Kantor S, Jakus R, Balogh B, Benko A, Gagdy G. Increased wakefulness, receptor sensitivity in generalized anxiety disorder. Pschopharmacology
motor activity and decreased theta activity after blockade of the 5-HT2B 1992;108:387e9.
receptor by the subtype-selective antagonist SB-215505. Br J Pharmacol 138. Paiva T, Arriaga F, Wauquier A, Lara E, Largo R, Leitao JN. Effect of ritanserin
2004;142:1332e42. on sleep disturbances in dysthymic patients. Psychopharmacology
112. Chiang C, Aston-Jones GA. 5-hydroxytryptamine2 agonist augments 1988;96:395e9.
gamma-aminobutyric acid and excitatory amino acid inputs to noradren- 139. Staner L, Kempenaers C, Simonnet MP, Fransolet L, Mendlewicz J. 5-HT2
ergic locus coeruleus neurons. Neuroscience 1993;54:409e20. receptor antagonism and slow wave sleep in major depression. Acta Psy-
113. Pehec EA, McFarlane HG, Maguschak K, Price B, Pluto CP. M100,907, chiatr Scand 1992;86:133e7.
a selective 5-HT(2A) antagonist attenuates dopamine release in the rat 140. Seiden DJ, Hull SG, Erman M, Schwartz H, Anderson C, Prosser W, et al.
medial prefrontal cortex. Brain Res 2001;888:51e9. APD125, a selective serotonin 5-HT(2A) receptor inverse agonist, signi-
114. Nair SG, Gudelsky GA. Activation of 5-HT2 receptors enhances the release of cantly improves sleep maintenance in primary insomnia. Sleep
acetylcholine in the prefrontal cortex and hippocampus of the rat. Synapse 2008;31:1663e71.
2004;53:202e7. 141. Landolt HP, Wehrle R. Antagonist of serotonergic 5-HT2A/2C receptors:
115. Bortolozzi A, Diaz-Mataix L, Scorza MC, Celada P, Artigas F. The activation of mutual improvement of sleep, cognition and mood. Eur J Neurosci
5-HT2A receptors in prefrontal cortex enhances dopaminergic activity. 2009;29:1795e809.
J Neurochem 2005;95:1597e607. 142. Steinbusch HW. Serotonin-immunoreactive neurons and their projections
*116. Idzikowski C, Mills FJ, Glennard R. 5-Hydroxytryptamine-2 antagonist in the CNS. In: Bjorklund A, Hokfelt T, editors. Handbook of chemical
ritanserin increases human slow wave sleep. Brain Res 1986;378:164e8. neuroanatomy, vol. 3. New York: Elsevier; 1984. p. 68e125.
J.M. Monti / Sleep Medicine Reviews 15 (2011) 269e281 281

143. Fogel SM, Beninger RJ, Smith CT. Increased GABAergic activity in the 148. Detari L, Rasmusson DD, Semba K. The role of basal forebrain neurons in
region of the pedunculopontine and deep mesencephalic reticular nuclei tonic and phasic activation of the cerebral cortex. Prog Neurobiol
reduces REM sleep and impairs learning in rats. Behav Neurosci 1999;58:249e77.
2010;124:79e86. 149. Adrien J, Alexandre C, Boutrel B, Popa D. Contribution of the knock-out
144. Torterolo P, Morales FR, Chase MH. GABAergic mechanisms in the pedun- technology to understanding the role of serotonin in sleep regulations. Ach
culopontine tegmental nucleus of the cat promote active (REM) sleep. Brain Ital Biol 2004;142:369e77.
Res 2002;944:1e9. 150. Adrien J. Sleep and waking in mutant mice that do not express various
145. Pal D, Mallick BN. GABA en pedunculopontine tegmentum regulates proteins involved in serotonergic neurotransmission such as the seroto-
spontaneous rapid eye movement sleep by acting on GABAA receptors in nergic transporter, monoamine oxidase A, and 5-HT1A, 5-HT1B, 5-HT2A,
freely moving rats. Neurosci Lett 2004;365:200e4. 5-HT2C and 5-HT7 receptors. In: Monti JM, Pandi-Perumal SR, Jacobs BL,
146. Parnavelas JG. Neurotransmitters in the cerebral cortex. In: Uylings HBM, Nutt DJ, editors. Serotonin and sleep: molecular, functional and clinical
Van Eden CG, De Bruin JPC, Corner MA, Feenstra MGP, editors. Progress aspects. Basel: Birkuser; 2008. p. 457e75.
brain research, vol 85. Amsterdam: Elsevier; 1990. p. 13e29. 151. Ross EM, Kenakin TP. Mechanisms of drug action and the relationship
147. Araneda R, Andrade R. 5-Hydroxytryptamine2 and 5-hydroxytryptamine1A between drug concentration and effect. In: Hardman JG, Limbird LE, editors.
receptors mediate opposing responses on membrane excitability in rat The pharmacological basis of therapeutics. 10th ed. New York: McGraw-Hill;
association cortex. Neuroscience 1991;40:399e412. 2001. p. 31e43.

Vous aimerez peut-être aussi