You are on page 1of 14

REVIEW ARTICLE

published: 15 November 2011


doi: 10.3389/fneur.2011.00070

Functional anatomy of non-REM sleep


Isabel de Andrs, Miguel Garzn and Fernando Reinoso-Surez*
Departamento de Anatoma Histologa y Neurociencia, Universidad Autnoma de Madrid, Instituto de Investigacin Hospital Universitario La Paz, Madrid, Spain

Edited by: The state of non-REM sleep (NREM), or slow wave sleep, is associated with a synchro-
Oscar Prospro-Garca, Universidad
nized EEG pattern in which sleep spindles and/or K complexes and high-voltage slow wave
Nacional Autnoma de Mxico,
Mexico activity (SWA) can be recorded over the entire cortical surface. In humans, NREM is sub-
Reviewed by: divided into stages 2 and 34 (presently named N3) depending on the proportions of each
Matthew R. Ebben, Weill Medical of these polygraphic events. NREM is necessary for normal physical and intellectual per-
College of Cornell University, USA formance and behavior. An overview of the brain structures involved in NREM generation
Rene Druckner-Colin, Universidad
shows that the thalamus and the cerebral cortex are absolutely necessary for the most
Nacional Autonoma de Mexico,
Mexico signicant bioelectric and behavioral events of NREM to be expressed; other structures
Javier Velazquez-Moctezuma, like the basal forebrain, anterior hypothalamus, cerebellum, caudal brain stem, spinal cord
Universidad Autonoma Metropolitana, and peripheral nerves contribute to NREM regulation and modulation. In NREM stage 2,
Mexico
sustained hyperpolarized membrane potential levels resulting from interaction between
*Correspondence:
thalamic reticular and projection neurons gives rise to spindle oscillations in the mem-
Fernando Reinoso-Surez,
Departamento de Anatoma brane potential; the initiation and termination of individual spindle sequences depends on
Histologa y Neurociencia, corticothalamic activities. Cortical and thalamic mechanisms are also involved in the gen-
Universidad Autnoma de Madrid, eration of EEG delta SWA that appears in deep stage 34 (N3) NREM; the cortex has
Arzobispo Morcillo 4, Madrid 28029,
Spain.
classically been considered to be the structure that generates this activity, but delta oscil-
e-mail: fernando.reinoso@uam.es lations can also be generated in thalamocortical neurons. NREM is probably necessary to
normalize synapses to a sustainable basal condition that can ensure cellular homeostasis.
Sleep homeostasis depends not only on the duration of prior wakefulness but also on its
intensity, and sleep need increases when wakefulness is associated with learning. NREM
seems to ensure cell homeostasis by reducing the number of synaptic connections to a
basic level; based on simple energy demands, cerebral energy economizing during NREM
sleep is one of the prevalent hypotheses to explain NREM homeostasis.
Keywords: slow wave sleep, sleep need, thalamuscerebral cortex unit, rostral hypnogenic system, caudal
hypnogenic system, NREM sleep homeostasis

INTRODUCTION OVERVIEW OF THE BRAIN STRUCTURES INVOLVED IN NREM


The state of non-REM sleep (NREM), or slow wave sleep, is GENERATION
associated with a synchronized EEG pattern in which specic Recently, in a revision of the publications describing the anatomi-
electrographic events take place. These events are sleep spindles cal connections and effects of lesions and electrical stimulation of
and/or K complexes and high-voltage slow wave activity (SWA) specic brain structures on the sleepwakefulness cycle (SWC) we
within the delta frequency band (between 0.5 and 4.0 Hz) that can outlined the brain structures located at practically all levels of the
be recorded over the entire cortical surface. In humans, NREM is encephalon that participate in the organization of the NREM state
subdivided into stages 2 and 34 depending on the proportions (Reinoso-Surez et al., 2011; Figure 2). Also, different ndings
of each of these polygraphic events. Stage 2, with sleep spindles have shown that impulses from peripheral nerves and spinal cord
and K complexes and less than 20% of SWA in the recording, cor- can modify brain electrical activity increasing EEG synchroniza-
responds, from the behavioral point of view, to a light sleep with tion and producing SWA, as occurs after stimulation of cutaneous
a low-threshold to awakening. Behaviorally, stage 34, with SWA nerves (Pompeiano and Swett, 1962), after repetitive visual stim-
occupying at least 20% of the recording, is a deeper sleep. Recently, uli or diffused and permanent illumination of the retina (Mancia
in the new sleep scoring classication of the AASM (Iber et al., et al., 1959), or after afferent volleys originating in the vagus
2007) the former stage 34 is identied as only one stage, named nerve (Puizillout et al., 1973; Valds-Cruz et al., 2008). Conversely,
N3. In young adults stage 34 (N3) is especially abundant in the decrease of SWA in the EEG (and therefore enhancement of desyn-
rst third of the night (Figure 1). Its proportions show notable sta- chronized EEG) takes place after suppression of trigeminal nerve
bility in long and short sleepers, and it is the rst stage to recover afferents (Vies-Morros, 1959), and after lesion or blockage of the
after sleep deprivation; it is considered to be the main component spinal cord, thus indicating that ascending synchronizing impulses
of the core sleep necessary for normal physical and intellectual can also be generated in the spinal cord (Hodes, 1964; De Andrs
performance and behavior. It usually decreases in the elderly. et al., 1976).

www.frontiersin.org November 2011 | Volume 2 | Article 70 | 1


de Andrs et al. Non-REM sleep functional anatomy

When the medulla and the caudal pons are separated from more medullary or midpontine cats (Reinoso-Surez et al., 2011). How-
cranially located brainstem structures, active and resting periods ever, lesion and stimulation experiments in our and other labo-
are still organized in a manner that can resemble very elemental ratories demonstrated that EEG synchronizing hypnogenic inu-
waking and sleep states (Siegel et al., 1986). But, in the absence ences on the rostral brainstem and prosencephalon originate in the
of inuence from higher brainstem structures, the medulla, and caudal pons and medulla at the level of the nucleus of the solitary
caudal pons are unable to generate true NREM or REM sleep signs tract (Magnes et al., 1961; Berlucchi et al., 1964; Reinoso-Barbero
since the full expression of these states does not occur in chronic and De Andrs, 1995) and caudal pontine reticular nucleus (Batini
et al., 1958; Cordeau and Mancia, 1959; Reinoso-Surez et al.,
1962; Rossi et al., 1963; Camacho-Evangelista and Reinoso-Surez,
1964; Zarranz and Reinoso-Surez, 1971; Reinoso-Surez and De
Andrs, 1976; Garzn, 1996). These ndings, together with the
rostral projections of these lower brainstem structures (Reinoso-
Surez et al., 1977), support the assertion by Moruzzi (1972) that
the deactivating structures of the lower brainstem may inhibit the
ascending reticular system and counteract its inuence at the level
of the diencephalon.
Our group reported that lesions in the long ascending tracts
at the level of the oral pontine tegmentum produce EEG desyn-
chronization; bilateral desynchronization of the EEG also fol-
lowed uni- or bilateral lesions in the superior cerebellar pedun-
cle that destroyed the brachium conjunctivum tract (Camacho-
Evangelista and Reinoso-Surez, 1964, 1965). This led us to infer
that these lesions suppressed ascendant hypnogenic synchroniz-
FIGURE 1 | Nocturnal hypnogram of a young man. Modied from ing impulses from more caudal levels of the brainstem and the
Reinoso-Surez et al. (2010).
deep cerebellar nuclei. In the case of lesion to the brachium

FIGURE 2 | Schematic representation of the encephalic NREM that these structures are necessary for the behavioral and bioelectric signs
sleep-promoting structures. A parasagittal section of cat brain shows a that characterize NREM sleep. AC, anterior commissure; CC, corpus
shaded area representing the brain structures where lesion decreases NREM callosum; DCN, deep cerebellar nuclei; Fo, fornix; G7, genu of the facial nerve;
sleep and/or EEG synchronization or stimulation increases NREM sleep. The IC, inferior colliculus; IO, inferior olive; LV, lateral ventricle; MRF; midbrain
main connections between these structures and the structures responsible of reticular formation; MT, medullary tegmentum; OCh, optic chiasm; PGS,
the organization of wakefulness (encompassed in red line) or REM sleep (blue periaqueductal gray substance; RPC, caudal pontine reticular nucleus; RPO,
lines) with the exception of the efferent hypothalamic and midbrain oral pontine reticular nucleus; SC, superior colliculus; SC and PN, spinal cord
connections and the brainstem connections from the basal forebrain are and peripheral nerves; SN, solitary nucleus; TB, trapezoid body. Modied from
shown. The thalamuscerebral cortex complex or unit is darker to emphasize Reinoso-Surez et al. (2011).

Frontiers in Neurology | Sleep and Chronobiology November 2011 | Volume 2 | Article 70 | 2


de Andrs et al. Non-REM sleep functional anatomy

conjunctivum tract, the suppression of the synchronizing impulses THE THALAMUS AND THE CEREBRAL CORTEX
was associated to a bilateral increase of desynchronized EEG but The thalamus and the cerebral cortex work as an indivisible unit
also to the generation of bursts of intermediate- to high-voltage in brain functions. The cerebral cortex is the main afferent and
theta band EEG activity recorded in cortical areas (motor and pari- efferent structure for the thalamus, and, in turn, the thalamus is
etal cortices) that are targets of the thalamic nuclei where cerebellar the main subcortical afferent and efferent structure for the cere-
projections end (Camacho-Evangelista and Reinoso-Surez, 1965; bral cortex (Reinoso-Surez et al., 2011; Figures 3 and 4). In both
Reinoso-Surez, 1992, 1993). In agreement with these results, uni- cases these connections are made of two types of projection neu-
lateral lesions of the brachium conjunctivum in cats increased rons (Jones, 2001; Rubio-Garrido et al., 2009; Figures 3 and 4): the
wakefulness and drowsiness while decreasing NREM and REM thalamus sends projections from core- (C) and matrix- (M) type
sleep (De Andrs and Reinoso-Surez, 1979). thalamocortical neurons and the cerebral cortex sends projections
Sleep spindles and delta SWA cannot be observed in decer- from layer VI and layer V neurons.
ebrate animals in which the brainstem has been released from The axons of both types (M and C) of thalamocortical projec-
prosencephalic inuences (Villablanca, 1966; De Andrs and Cor- tion neurons provide collateral branches to the thalamic reticular
pas, 1991), even though, these preparations show the behavioral nucleus when traversing it on their way to the cerebral cortex.
and bioelectric REM sleep signs. On the other hand, the isolated The GABAergic thalamic reticular neurons that receive these col-
forebrain (or cerveau isol preparation) of cats transected at the laterals project back, modulating both the thalamic projection
intercollicular level, which lacks brainstem inuences, is able to neurons that provide the thalamocortical axons as well as the
express the full bioelectric activity patterns of NREM sleep (Vil- related interneurons (Figure 3). Thalamic innervation in cortical
lablanca, 1965; Corpas and De Andrs, 1991). In addition, when
the third cranial nerves and nuclei are not affected by the transec-
tion in the cerveau isol preparation, the bioelectric EEG activities
in the isolated forebrain are coupled to pupil size changes, that
is, myosis during synchronized EEG and mydriasis during desyn-
chronized EEG, that occur in intact cats during NREM sleep and
wakefulness, respectively (Villablanca, 2004). Furthermore, pros-
encephalic mechanisms are sufcient for homeostatic regulation
of NREM sleep: pharmacologic deprivation of this state in the
cerveau isol preparation is followed as in intact animals by an
NREM sleep rebound (Corpas and De Andrs, 1991).
In addition, clinical, and lesion and stimulation experiments
made it possible to describe a rostral NREM hypnogenic system
that includes the basal forebrain, and preoptic and anterior hypo-
thalamic regions (von Economo, 1930; Hess, 1931; Nauta, 1946;
Hernndez-Pen, 1962; Sterman and Clemente, 1962; Madoz and
Reinoso-Surez, 1968; McGinty and Sterman, 1968; Szymusiak
and McGinty, 1986). The ventral lateral preoptic area and the
median preoptic nucleus show a higher density of neurons with
increased activity during NREM episodes than do other anterior
hypothalamic regions (Szymusiak et al., 1998).
Also, lesion, stimulation, and clinical studies demonstrate that
the thalamus is necessary to generate the electrophysiological and
behavioral manifestations that characterize NREM sleep (Hess,
1931, 1968; Morrison and Dempsey, 1942; Villablanca, 1974; Ste-
riade et al., 1985; Sforza et al., 1995). The chronic athalamic cat
shows wakefulness and REM sleep but not typical NREM sleep
(Villablanca, 2004). Also Villablanca (1972, 2004) afrmed that
true NREM sleep is absent in chronic cats without telencephalon
(diencephalic cats) because neither delta SWA nor sleep spindles
could be recorded in the thalamus. FIGURE 3 | Thalamic nuclei, cell types, and thalamocortical
In summary, it appears that the thalamus and the cerebral relationships. The lower part of the gure represents a schematic coronal
section of the primate diencephalon and the upper drawing depicts a
cortex are absolutely necessary for the expression of the most sig- schematic section of the primary and association cerebral cortices.
nicant bioelectric and behavioral events of NREM sleep. Other Thalamic lateral posterior (LP) and ventral posterior (VP) nuclei are
structures like the basal forebrain, cerebellum and caudal brain represented as examples of high-order and rst-order thalamic nuclei,
stem, and impulses from the spinal cord and peripheral nerves respectively. C, core-type cells; Intr, thalamic intralaminar nuclei; It,
modulate NREM sleep but are not required for the actual gen- GABAergic interneurons; M, matrix-type cells; MD, medial dorsal thalamic
nucleus; NR, thalamic reticular nucleus; IVI, cortical layers. Modied from
eration of the bioelectric and behavioral signs accompanying the Reinoso-Surez et al. (2011).
expression of NREM sleep (Figure 2).

www.frontiersin.org November 2011 | Volume 2 | Article 70 | 3


de Andrs et al. Non-REM sleep functional anatomy

FIGURE 4 | Schematic representation of the fiber and neuronal and subcortical origins are represented in different colors and specic
organization of the cerebral cortex. The projection neurons, pyramidal distributions. The wide distributions of dopaminergic (DA), serotonergic
neurons in layers IIIII, V, and VI, are represented in different colors (5-HT), noradrenergic (NA), histaminergic (His), orexinergic (Orx), and
according to their origin and targets. The two types of interneurons are GABAergic (GABA) bers originating in brainstem, diencephalic, and basal
represented in different colors: (1) the excitatory interneuron, a spiny prosencephalic structures are represented by their terminals, as is the
stellate cell, is in pink; and (2) the inhibitory interneurons are in light green. topographically organized terminals of the basal forebrain cholinergic (Ach)
There are four specic examples of inhibitory interneuron: two bers. Thalamocortical bers targeting cortical layers I (M-type) and IV
dendrite-and tuft-targeting cells [Cajal-Retzius (C-R) and Martinotti (M) (C-type) are also shown. I to VI, cortical layers one to six. Open triangles,
neurons], one dendrite targeting cell [double bouquet (DB) neuron], and excitatory terminals; solid triangles, inhibitory terminals. From
one axon targeting cell [chandelier (C) neuron]. Afferent bers from cortical Reinoso-Surez et al. (2011).

layer I is at least as heavy as that in layer IV; every part of corti- deafferentized reticular thalamic nucleus. Bal and McCormick
cal layer I receives combined input from multiple thalamic nuclei (1993) demonstrated that reticular nucleus neurons have an
(Rubio-Garrido et al., 2009). intrinsic rhythmic activity at spindle frequencies (712 Hz) in
Since the 1980s, the thalamocortical and corticothalamic cel- slice preparations. The thalamic reticular nucleus is a relatively
lular bases of sleep spindles and delta SWA generation have been thin sheet of neurons that surrounds the anterior, lateral and,
extensively studied by Steriade et al. (1985, 1988, 1991, 1993). In to some extent, ventral surfaces of the dorsal thalamus, and it
a series of experiments these authors showed that the combina- is entirely composed of GABAergic cells. Because of its anatomical
tion of the intrinsic electrophysiological properties of the thalamic position the thalamic reticular nucleus is traversed by virtually
and cortical neurons, together with their synaptic connections, is all axons connecting the dorsal thalamus with the neocortex,
responsible for the generation of sleep spindles and delta SWA giving the nucleus its reticulated appearance and name; it also
oscillations in the EEG during NREM sleep (for review see Nez- receives collateral branches from corticothalamic bers originat-
Molina and Amzica, 2004; Fuentealba and Steriade, 2005). Thus, ing in cortical layer VI (Figures 3 and 5). Thalamic reticular
the EEG activities, which appear during stages 2 and 34 (N3) of nucleus neurons do not project directly to the cortex although
NREM sleep are essentially generated by the activity of neurons in they widely project to the relay thalamic nuclei. Simultaneous
the thalamic reticular nucleus, in the thalamocortical projecting intracellular recordings in reticular, thalamocortical, and cortical
nuclei, and in the cerebral cortex. neurons (Steriade and Deschenes, 1988) indicated that sleep spin-
Steriade et al. (1985) proposed, early on, that the neurons of dle generation was produced by a chain of events with three main
the thalamic reticular nucleus are pacemakers for sleep spindles, consecutive steps: (1) repetitive spike-bursts at spindle frequency
based on the absence of the sleep spindle oscillation in thalam- generated by GABAergic thalamic reticular nucleus neurons; (2)
ocortical systems when they are disconnected from the thalamic transfer of this activity to relay thalamocortical nuclei producing
reticular nucleus and the presence of spindle rhythmicity in the rhythmic (at spindle frequency) inhibitory postsynaptic potentials

Frontiers in Neurology | Sleep and Chronobiology November 2011 | Volume 2 | Article 70 | 4


de Andrs et al. Non-REM sleep functional anatomy

(Morrison and Dempsey, 1942); these responses resemble the


EEG activity recorded in supercial layer I of the cortex during
sleep spindles (Hess, 1968). The frontal and posterior parietal
cortices are the cortical regions that are the prime location for
recruiting responses and spontaneous spindling in the cat (Morri-
son and Dempsey, 1942; Starzl and Magoun, 1951; Reinoso-Surez,
1954). The supercial thalamocortical projection system to layer I
in the frontal and posterior parietal cortex of the cat does not arise
from intralaminar nuclei but from the paralaminar region of ven-
tral medial, ventral anterior, and ventral lateral nuclei (Oka et al.,
1982; Avendao et al., 1990). In addition, the M-type cells of these
nuclei innervate wider zones of layer I of the cerebral hemisphere,
as it also happens in rats (Rubio-Garrido et al., 2009). Conse-
quently this layer I projection system to the frontal and posterior
parietal cortices may be an important leading path for recruit-
ing responses and spontaneous spindling activities (Oka et al.,
1982; Avendao et al., 1990). In relation with the possible thalam-
ocortical route for sleep spindle generation, Jimnez-Castellanos
and Reinoso-Surez (1985), Velayos et al. (1989), and Avendao
et al. (1990) proposed a circuit that involves the thalamic reticular
nucleus and its projection to the paralaminar ventral medial, ven-
tral anterior, and ventral lateral nuclei (Figure 5). The thalamic
reticular nucleus GABAergic pacemaker neurons of the spindle
FIGURE 5 | Schematic representation of the anatomical bases for the
oscillations would impose their rhythmicity on the paralaminar
sleep spindles of NREM sleep and the intermediate- to high-voltage M-type neurons that project to layer I of the frontal and pari-
theta EEG activity of drowsiness. From Reinoso-Surez et al. (2011). etal cortices (Reinoso-Surez et al., 2011; Figures 3 and 5). For
this to occur, as happens in stage 2 of NREM sleep, it is neces-
sary to decrease the brainstem activating system action on both
(IPSPs) in thalamocortical neurons; and (3) transfer to the cerebral the thalamic reticular nucleus neurons, which consequently re at
cortex of postinhibitory rebound spike-bursts of the thalamocor- their intrinsic burst activity, and the paralaminar neurons, which
tical cells producing excitatory postsynaptic potentials (EPSPs) then transfer the slow burst activity to the cortex. Altogether this
in cortical cells at spindle frequency. The repetitive spike-bursts permits the thalamic paralaminar M-type neurons projecting to
of the reticular nucleus neurons are Na+ action potentials gen- layer I of the cerebral cortex, in their turn, impose this activity on
erated by low-threshold Ca+ spikes at spindle frequency (Bal the cortex, which has also been released from the inuence of the
and McCormick, 1993). Since the reticular nucleus projection is ascending activating system (Figure 5; Reinoso-Surez, 1992, 1993;
GABAergic, the reticular nucleus activities are transmitted to the Reinoso-Surez et al., 2011). Recently Bonjean et al. (2011) have
thalamocortical relay nuclei generating rhythmic IPSPs in these concluded that spindle oscillation initiation depends on activity by
cells. Thalamocortical relay cells are able to generate Ca+ spikes cortical pyramidal neurons on thalamic reticular nucleus neurons,
at membrane hyperpolarizing levels (Jahnsen and Llins, 1984), which would then re synchronizing discharges. The intermediate
these Ca2+ spike makes it possible to generate Na+ potentials in phase would be the interaction of thalamic reticular nucleus neu-
the re-polarizing phase of the IPSPs that can reach the cerebral cor- rons with the thalamic projection neurons as we have described
tex and produce rhythmic EPSPs in cortical neurons at the sleep above. Spindle ring is terminated by the combination of I h cur-
spindle frequencies (Steriade and Deschenes, 1988) since the thal- rent and corticothalamic input depolarizing actions. Consequently
amic afferents to the cerebral cortex are glutamatergic (Figures 3 corticothalamic activity is involved not only in the long-range syn-
and 5). Finally, in relation with the cellular basis of the sleep chronization of spindles but also in the initiation and termination
spindles, it is noteworthy that spindle oscillations in the mem- of individual spindle sequences, and this controls spindle duration.
brane potential of the reticular and thalamocortical neurons arise Andrade et al. (2011) have investigated human hippocampal
from sustained hyperpolarized membrane potential levels (Llins functional connectivity with neocortex in wakefulness and during
and Steriade, 2006), and that thalamocortical or corticothalamic NREM sleep. They found that the strongest functional connec-
collaterals to the thalamic reticular nucleus can reinforce the gener- tivity between the hippocampal formation and neocortex was
ation of sleep spindles. Synaptic activation of the thalamic reticular observed in stage 2 sleep spindles. The hippocampal structure
nucleus neurons by stimulation of these afferents invariably deter- dominating functional connectivity to frontal neocortex regions
mines the generation of a sequence of spindle waves crowned with during sleep stage 2 was the subiculum. This increased connectiv-
spike-bursts. ity between the hippocampal formation and neocortical regions
The recruiting responses are the neurophysiological activity suggests an increased capacity for possibly global information
that can be experimentally elicited in the cat cortex after low- transfer in sleep stage 2 and would support the hypothesis of
frequency stimulation of the intralaminar and related nuclei sleep-dependent memory consolidation.

www.frontiersin.org November 2011 | Volume 2 | Article 70 | 5


de Andrs et al. Non-REM sleep functional anatomy

Cortical and thalamic mechanisms are also involved in the gen- membrane oscillations were accompanied by a slower <1 Hz
eration of EEG delta SWA that appear in deep NREM sleep stage membrane rhythm (Dossi et al., 1992). This oscillation consists
34. EEG with SWA can be registered in the cortex of cats without of a depolarized (UP) state in which neurons re followed by a
thalamus (Villablanca, 1974); therefore, the cortex has classically hyperpolarized (DOWN) state with neuronal silence. There is a
been considered to be the structure that generates this activity. close temporal relationship between the >1 Hz cellular oscillation
However, Steriade et al. (1991, 1993) showed that delta oscilla- and the 1- to 4-Hz delta EEG waves since the negative component
tions can also be generated in thalamocortical neurons. These cells of the EEG signal corresponds to the DOWN state of the corti-
express this property intrinsically when their membrane potential cal neurons (Contreras and Steriade, 1995). The cortical origin of
is at more strongly hyperpolarized levels than during sleep spin- these slow <1 Hz rhythm waves was demonstrated by the presence
dles. In thalamic cells, delta is an intrinsic oscillation consisting of of the oscillation in the cerebral cortex after thalamectomy (Ste-
low-threshold spikes alternating with post-hyperpolarizing poten- riade et al., 1993) and its absence in the thalamus of decorticated
tials. Action potentials at delta frequency that can be transferred to animals (Timofeev and Steriade, 1996). The cortical slow <1 Hz
the cortex are generated in the depolarizing phase of the Ca2+ low- activity is expressed synchronously in wide areas of the cerebral
threshold spikes. Therefore, there is a degree of incompatibility in cortex; disconnection of intracortical synaptic linkages does not
simultaneously generating thalamic spindles and delta rhythms; abolish the <1 Hz activity, but it does determine the loss of its
this may explain the distinct prevalences of these two types of oscil- synchronization between the different cortical areas (Amzica and
lations during the different stages of NREM [stages 2 and 34 (N3) Steriade, 1995). These ndings indicate a possible role for <1 Hz
respectively] with synchronization of the electroencephalogram cortical activity in synchronous excitation of wide brain territo-
(Nuez et al., 1992). ries (cortical and thalamic) during NREM, which would allow the
In fact, the thalamus and the cerebral cortex are close structures simultaneous expression of the same EEG activities in all territories
and abundantly interconnected, and they form a non-divisible unit (Nez-Molina and Amzica, 2004). Interestingly, the generation
in brain functions. During NREM sleep oscillating cortical activ- of K complexes during stage 2 of NREM sleep seems to be related
ities can be transferred to specic thalamic structures via cortical with the <1 Hz rhythm of the cortical pyramidal cells as the K
efferents from layer VI and V pyramidal neurons; in turn, thalamic complexes can be generated by stimulation of a cortical area other
oscillating activities can reach cortical neurons principally through than the one in which they are recorded. Also, K complexes can be
M-type, but also C-type, thalamocortical neurons, since the for- evoked by sensory stimuli arriving at the cerebral cortex (Nez-
mer innervate extensive zones of layer I of the cerebral cortex and Molina and Amzica, 2004). K complexes, during NREM stage 2,
can convey oscillatory information activity to the distal apical tufts are usually followed by a sleep spindle; in these circumstances, it
of pyramidal neurons from layers II, III, and V, neurons which are is possible that the <1 Hz cortical activity would be sufcient to
source for corticocortical and subcortical connections. induce thalamic reticular nucleus neuron oscillation at the sleep
The different oscillatory activities and spike-bursting mode in spindle frequencies, since, as we have indicated above, stimulation
thalamic and cortical neurons that we have just described are com- of corticothalamic afferents to the reticular nucleus is one the most
pletely absent in wakefulness and/or REM where the associated efcient stimuli in evoking rhythmic activity in reticular thalamic
EEG is desynchronized. During the later states, thalamic and cor- cells at sleep spindle frequencies.
tical neurons are depolarized and show a tonic ring discharge
pattern (Llins and Steriade, 2006). The level of the membrane BASAL FOREBRAIN ANTERIOR HYPOTHALAMIC
potential of thalamic neurons is mainly controlled by cholinergic STRUCTURES
input and, in wakefulness by additional glutamatergic and aminer- Beside thalamus and cerebral cortex, the preoptic region and the
gic inputs, from the mesopontine cholinergic, glutamatergic, and anterior hypothalamus are other prosencephalic regions involved
aminergic cell groups. Stimulation of cholinergic mesopontine in NREM mechanisms (Figure 2). As we have mentioned before,
nuclei produces an opposite effect in thalamic cells to the effects of von Economo (1930) was the rst to suggest that the anterior
corticothalamic volleys, readily suppressing their slow oscillatory hypothalamus was a sleep-promoting structure (Reinoso-Surez
bursts due to the generation of excitatory depolarizing discharges et al., 2011); later on in the sixties, the importance of this region
that produce a tonic ring pattern of activity in thalamic neurons in the mechanisms for NREM generation was conrmed after
(Steriade et al., 1991). The ring rate of the cholinergic laterodorsal behavioral sleep associated with synchronized EEG was obtained
and pedunculopontine mesopontine nuclei neurons is increased with electrical stimulation of the preoptic region and of the basal
during wakefulness and REM sleep in comparison with ring rate forebrain (Sterman and Clemente, 1962) and by the insomnia
during NREM sleep (Steriade et al., 1982); therefore, when the that followed electrolytic or diathermocoagulation lesions in these
activity of the cholinergic mesopontine, hypothalamic and basal areas (Madoz and Reinoso-Surez, 1968; McGinty and Sterman,
forebrain, and glutamatergic and aminergic mesopontine struc- 1968). Also, selective neurotoxic lesions of the preoptic neurons
tures decreases during NREM, the membrane potential of thalamic induce insomnia (Szymusiak and McGinty, 1986) thus indicating
and cortical cells is hyperpolarized. This allows the generation of that the loss of local neurons rather than of bers of passage is
oscillatory patterns in the thalamic and cortical neurons, leading to critical for sleep control. Single unit recordings have shown that
expression of the specic EEG events of the NREM stages (Steriade the preoptic region as well as the basal forebrain there hold neu-
and McCarley, 1990). rons whose discharge rate is higher during NREM than in W; in
Finally, intracellular recording experiments of cortical and thal- addition, many preoptic- and diagonal band-sleep active neurons
amic neurons during synchronized EEG states detected that delta are also heat-sensitive (Alam et al., 1995). The same local thermal

Frontiers in Neurology | Sleep and Chronobiology November 2011 | Volume 2 | Article 70 | 6


de Andrs et al. Non-REM sleep functional anatomy

stimuli that activate preoptic heat-sensitive neurons produce a hypothalamic and midbrain structures could suppress activity by
sleep propensity and NREM with delta SWA (see Szymusiak et al., preoptic NREM sleep-promoting cells as well as REM-promoting
2007 for a review of the close interactions between sleep regulation neurons (Reinoso-Surez et al., 2010) and facilitate the switch to
and thermoregulation together with the numerous factors that wakefulness. Finally, the involvement of the preoptic region in
modulate the activity of the sleep-sensitive neurons in the anterior homeostatic sleep regulation has been reported in recent experi-
hypothalamus). Clusters of neurons exhibiting sleep-related c-Fos ments examining the patterns of c-Fos immunoreactivity in pre-
protein immunostaining were found in the ventral lateral preop- optic neurons following acute total sleep deprivation or selective
tic area and in the median preoptic nucleus (Sherin et al., 1996). REM sleep restriction (Gvilia et al., 2006a,b). These studies indi-
Neuronal density with increased activity during NREM episodes cate that the GABAergic ventral lateral preoptic neurons are more
is higher in these regions than in other anterior hypothalamic strongly activated in response to increasing sleep amounts during
sites (Szymusiak et al., 1998). Neurons of the ventral lateral pre- sleep rebound, whereas GABAergic cells of the median preoptic
optic area contain the inhibitory neuromodulator galanin and the nucleus are more strongly activated in response to sleep pres-
inhibitory neurotransmitter GABA (Sherin et al., 1998); GABA is sure during sleep deprivation. Thus, within the prosencephalon,
also found in most cells in the median preoptic nucleus (Gong the preoptic region seems to be deeply involved in homeostatic
et al., 2004). Anatomical tracer studies have shown that the ven- NREM and REM sleep mechanisms. Prosencephalic mechanisms
tral lateral preoptic area and the median preoptic nucleus project underlying sleep homeostasis have been indicated by the capac-
to arousal-related regions in the posterior hypothalamus and ity of the isolated forebrain to present NREM sleep rebound after
midbrain including the histaminergic tuberomammillary nucleus pharmacological deprivation (Corpas and De Andrs, 1991) and
(Sherin et al., 1998), the hypocretinergic/orexinergic perifornical the absence of REM rebound in REM sleep-deprived decerebrate
hypothalamic area (Uschakov et al., 2006; Yoshida et al., 2006), the animals (De Andrs and Corpas, 1991; De Andrs et al., 2003).
serotonergic dorsal raphe, and the noradrenergic locus coeruleus
(Steininger et al., 2001). All of these ndings indicate that the pre- BRAINSTEM AND CEREBELLAR STRUCTURES
optic area may promote sleep onset and maintenance by producing As we have mentioned in the introduction, early work with brain-
an inhibitory modulation of multiple arousal systems (Szymusiak stem transections or lesions below the oral pontine tegmentum
et al., 2007). In fact, GABA-mediated inhibitory actions in his- indicated that sleep-generating structures were located in the lower
taminergic tuberomammillary neurons were demonstrated after brainstem. NREM with EEG synchronization could be obtained
electrical stimulation of the ventral lateral preoptic area (Yang in behaving cats by low-frequency electrical stimulation of the
and Hatton, 1997). Also, local warming of the preoptic region medullary reticular formation at the level of the solitary tract
(a stimulus that as we have mentioned before activates pre- nucleus (Magnes et al., 1961), but lesion and stimulation studies in
optic warmth-sensitive neurons and generates NREM) causes the peripheral nerves, spinal cord, caudal pontine tegmentum, and
suppression of waking related ring in both dorsal presumedly deep cerebellar nuclei can also induce hypnogenic synchronizing
serotonergic raphe nucleus neurons (Guzmn-Marn et al., 2000) impulses (Figure 2).
and in hypothalamic perifornical neurons (Krilowicz et al., 1994). The solitary tract nucleus region in the dorsal medulla is
Recently, it has been demonstrated that sleep-regulatory molecules thought to provide a link between visceral activities such as res-
like interleukin-1, tumor necrosis factor, and growth hormone- piratory, cardiovascular and gastrointestinal functions, and the
releasing hormone, stimulate NREM sleep by acting on neurons in sleepwakefulness states. Biochemically, the solitary tract nucleus
the preoptic region. In addition, NREM sleep-promoting actions is a rather heterogeneous region where most viscerosensory affer-
also take place through interleukin-1 and tumor necrosis factor ent bers from the VIIth, IXth, and Xth cranial nerves terminate;
inhibitory actions on locus coeruleus neurons and by interleukin- therefore, this region has important roles in the central regula-
1 action on serotonergic raphe cells. What is more, the facilitation tion of visceral functions, but it also inuences sleepwakefulness
of NREM sleep can occur when the lateral hypothalamic perifor- states and electrical activity of the cerebral cortex. Not only does
nical area is subject to a endogenous adenosinergic inhibition that electrical stimulation of the solitary tract nucleus result in behav-
restrains the wakefulness-generating hypocretinergic/orexinergic ioral sleep with slow-EEG waves, these responses have also been
neurons (Krueger et al., 2008; Rai et al., 2010). obtained after the local administration of a variety of substances
Anatomical studies have demonstrated that most of the pos- including serotonin (Laguzzi et al., 1984), morphine and other
terior hypothalamic- and monoaminergic- arousal systems send opiates agonists of mu or delta opioid receptors (Reinoso-Barbero
feedback projections to the ventral lateral preoptic area (Chou and De Andrs, 1995; Figure 6), and glutamate (Golanov and
et al., 2002; Reinoso-Surez et al., 2011). In vitro experiments Reis, 2001). These latter ndings indicate that it is the activa-
have indicated that the wake-promoting neurotransmitters sero- tion of intrinsic solitary tract nucleus neurons and not of passing
tonin, noradrenalin, and acetylcholine inhibit identied preop- bers that is responsible for the sleep and EEG responses. Unit
tic GABA neurons (Gallopin et al., 2000); therefore, mutually activity studies have shown that there is a population of neu-
inhibitory interactions between the sleep-promoting preoptic rons in the solitary tract nucleus with an increased ring rate
region and the arousal-related hypothalamic and midbrain struc- during NREM sleep (Eguchi and Satoh, 1980). The solitary tract
tures may provide a substrate for a sleepwakefulness switch nucleus does not directly project to the cerebral cortex (Saper,
(McGinty and Szymusiak, 2000; Saper et al., 2001). Thus, activa- 1995), although it does project to several brainstem, thalamic, and
tion of preoptic sleep-promoting cells could lead to sleep onset hypothalamic areas that innervate the cortex and can mediate EEG
by inhibiting arousal structures; in turn, activation of arousal and sleep responses. These areas include: locus coeruleus, raphe

www.frontiersin.org November 2011 | Volume 2 | Article 70 | 7


de Andrs et al. Non-REM sleep functional anatomy

FIGURE 6 | Top: microphotographs illustrating the location of the saline


and opioid microinjections in two cats injected in the medial nucleus
(arrow) of the solitary tract (SOL). Bottom: Bars show the mean duration
in minutes spent in the different sleepwakefulness states during the rst
hour of recordings in controls (saline, 40 nl) and after morphiceptin (mu
opioid agonist) or DPDPE (delta opioid agonist) microinjections (2.4 nmol,
40 nl). Respiratory rate (triangles) in breaths per minute and heart rate
(circles) in beats per minute are also shown for each sleepwakefulness
state during the rst hour of recordings. Note the decreased respiratory and
heart rates in sleep states in comparison with values during wakefulness in
the control saline experiments; both opioid treatments produced a further
decrease in both autonomic variables. W, wakefulness; D, drowsiness;
NREM, non-REM sleep; REM, REM sleep. (After Reinoso-Barbero, 1993).

FIGURE 7 | Activating and synchronizing EEG centers in the pontine


and parabrachial nuclei, lateral hypothalamus, and nuclei of the
tegmentum. (A,CE) show diagrams of the combined extent of lesions in
midline thalamus. the pontine tegmentum of cats producing either EEG activation (diagonal
Results from our groups early work with lesions showed the dashed lines encircled by heavy dashed lines) or EEG synchronization
existence of ascending EEG synchronizing inuences arising from (horizontal lines encircled by heavy solid lines). (A) Parasagittal section
the rostral and intermediate parts of the nucleus reticularis pontis 2 mm from the midline; circled numbers indicate frontal plane levels on the
Reinoso-Surez (1961) atlas corresponding to sections (CE), respectively.
caudalis. Lesions in these areas decreased SWA in the cerebral
(B) Graph showing the percent of increase in activation (dashed line) and
cortex and the bioelectrical and behavioral signals of NREM decrease in activation (solid line) relative to the pre-lesion control-based
and REM sleep (Figure 7; Camacho-Evangelista and Reinoso- zero for each animal. Note that lesions that produced EEG synchronization
Surez, 1964; Zarranz and Reinoso-Surez, 1971; Reinoso-Surez are located at the level of the oral pontine reticular nucleus and those that
and De Andrs, 1976). More recent experiments microinjecting produced EEG activation are located caudal, dorsal, and lateral to this
nucleus. BC, brachium conjunctivum; BP, brachium pontis; CP, colliculus
small amounts of a carbachol solution in rostral and intermediate inferior; FP, fasciculus longitudinalis medialis; LM, lemniscus medialis; MV,
parts of the nucleus reticularis pontis caudalis produced a synchro- tractus mesencephalicus nervi trigemini; NLL, nucleus lemnisci lateralis;
nized EEG pattern associated with some REM sleep signs such as NP, nuclei pontis; NR, nucleus ruber; NRV, nucleus reticularis ventralis; NTR,
isolated and/or clustered PGO waves; the most rostral microin- nucleus corporis trapezoidei; OS, oliva superior; P, tractus pyramidalis; RPC,
jections were also associated with muscular atonia (Garzn, 1996; nucleus reticularis pontis caudalis; RPO, nucleus reticularis pontis oralis;
SCV, tractus spinocerebellaris anterior; TD, nucleus tegmenti dorsalis; TR,
Reinoso-Surez et al., 2001). Power spectra analyses indicated that corpus trapezoideum; TF, tractus tectospinalis. From Camacho-Evangelista
the synchronized EEG pattern obtained in these experiments did and Reinoso-Surez (1964).
not consist of delta waves, instead, the high-voltage SWA activity
was produced by enhancement of theta and alpha band frequencies
(Garzn, 1996). Therefore, this cortical electrical pattern does not synchronized EEG without delta wave enhancement. These results
correspond to fully developed NREM sleep; instead, these electri- indicate that not only is the pontine tegmentum the neural sub-
cal features are more similar to those expressed in the EEG during strate for REM sleep and desynchronized EEG mechanisms, but
the transitional period between NREM and REM sleep (Gottes- that its caudal region especially the rostral and intermediate areas
mann, 1996). Recently we have reported (Moreno-Balandrn et al., of the nucleus reticularis pontis caudalis; together with the dorsal
2008) that low-doses and small carbachol microinjections deliv- part of the oral region seems to be involved in generating the
ered in the dorsal part of the oral pontine tegmentum in cats, pattern of bioelectrical activity that announces REM onset from
at the level of the perilocus coeruleus alpha, produce a similar NREM sleep.

Frontiers in Neurology | Sleep and Chronobiology November 2011 | Volume 2 | Article 70 | 8


de Andrs et al. Non-REM sleep functional anatomy

The major role of the cerebellum in motor functions has under- in SWC mechanisms through a functional antagonism in the
valued its possible involvement in sleep regulation. Cerebellar regulation of the proportions of sleep and wakefulness between
participation in sleep can be related to motor activities during cerebellar cortex and deep cerebellar nuclei; that is, like the changes
sleep but also to intrinsic sleep mechanisms. The former activities occurring after the brachium conjunctivum tract lesions, increased
involve postural adjustment and control of muscle tone and move- wakefulness, and decreased NREM and REM sleep were reported
ments during sleep that, as in wakefulness, must be controlled by after bilateral lesions of the fastigial nuclei (Giannazzo et al., 1968).
the cerebellum; in fact, early experimental studies showed the exis- However, these effects were just the opposite after lesions in the
tence of cerebellar components in muscle atonia and phasic activ- middle cerebellar peduncle (Raffaele et al., 1971) or in the cor-
ities of the eyes lateral rectus muscle during REM (Jouvet, 1962; tex of vermis and cerebellar hemispheres that produced increased
Guglielmino and Strata, 1971; Gadea-Ciria and Fuentes, 1976; NREM and REM sleep (De Andrs and Reinoso-Surez, 1979;
Cunchillos and De Andrs, 1982). Together with these actions, Garca-Ura et al., 1980). Cerebellectomized animals present all
the cerebellum has important inuences on intrinsic sleep mech- SWC states (Jouvet, 1962; Paz et al., 1982). However and sur-
anisms as was rst indicated by the changes observed in cortical prisingly, in spite of the severe neurological decits produced by
and thalamic electrical activity that follow electrical stimulation cerebellectomy, the quantitative alterations in the proportions of
and lesion of different cerebellar structures. SWC states in cerebellectomized animals are smaller than those
Moruzzi (1958) reported that diffuse EEG effects could be produced by localized subcortical or cortical cerebellar lesions.
obtained through the efferent of the nucleus dentatus, this effect In our experiments (Cunchillos and De Andrs, 1982) with cere-
was also obtained with low-frequency electrical stimulation of bellectomized cats, the main change after cerebellectomy was an
the nucleus fatigius, while high frequency stimulation of this increase in drowsiness that persisted during the entire survival
nucleus produced diffuse EEG activation (Fadiga et al., 1968). time (5 weeks) and that was associated with small decreases in
Specic supercial thalamocortical responses (surface negative- both W and NREM sleep as well as an increase in REM sleep.
deep positive potentials) in the frontal and parietal cortices were These results conrm cerebellar involvement in mechanisms of
also produced by stimulation of the cerebellar nuclei (Sasaki et al., synchronized EEG and support the hypothesis that the cerebel-
1976). Suppression of the impulses from the deep cerebellar nuclei lum, through dual opposite cortical and subcortical effects, would
through lesion of the brachium conjunctivum tract produce, have an important regulatory action in the ne adjustments in the
together with a bilateral activation of the EEG, the appearance of proportions of SWC states for maintaining the sleepwakefulness
synchronized bursts of intermediate- to high-voltage EEG activ- equilibrium (Cunchillos and De Andrs, 1982).
ity in the theta band (713 Hz, bigger of 250 mv) that can be A recent study (Dang-Vu et al., 2008) with functional magnetic
recorded in frontal and parietal cortical areas that are targets of the resonance imaging has reported that the slow oscillation <1 Hz
projections from the thalamic nuclei that receive cerebellar affer- rhythm of NREM sleep is associated with signicantly increased
ents (Camacho-Evangelista and Reinoso-Surez, 1965; Reinoso- local activity in specic cerebral regions: the parahippocampal
Surez, 1992, 1993). The bioelectrical features of these synchro- gyrus and cerebellum (in both cerebellar hemispheres and ver-
nized bursts are similar to the relaxation rhythms described in mis). On the basis of parallel changes that occur in forebrain and
cats during periods of relaxed wakefulness or drowsiness (Ursin cerebellum during NREM sleep, Andre and Arrighi (2003) have
and Sterman, 1981). A similar EEG synchronization, most con- proposed that forebrain and cerebellum may cooperate strongly
spicuously recorded in the ipsilateral frontal and parietal cortices, in information processing during sleep. As yet no further stud-
appears especially after the lesion of the brachium conjunctivum ies have examined cerebellar participation at the cellular level in
tract at the level of the red nucleus (Reinoso-Surez, 1954). This the generation of the slow <1 Hz NREM sleep rhythm, but the
intermediate- to high-voltage SWA in the theta band can also be Dang-Vu et al. (2008) ndings may be indicative of parallel oper-
recorded in the thalamic paralaminar nuclei that receive afferents ating capabilities in the cerebellum and hippocampal formation
from the cerebellum; it looks as if suppression of the cerebellar during NREM sleep. The growing data on hippocampal activation
afferents releases these nuclei and allows them to re at a pos- during NREM sleep after memory task training (Peigneux et al.,
sible intrinsic theta frequency, modulated by thalamic and other 2004; Ji and Wilson, 2007; Rasch et al., 2007) and recent com-
extrathalamic connections that, projecting to layer I, impose their pelling evidence that slow <1 Hz oscillations have a causal role
bioelectrical activity on the frontal and parietal cortices (Figure 5; in consolidating memories during NREM sleep (Marshall et al.,
Reinoso-Surez, 1992, 1993; Reinoso-Surez et al., 2011). The two 2006) open the possibility of cerebellarforebrain cooperation in
bioelectric phenomena after the lesion on the superior cerebel- memory consolidation processes during NREM sleep.
lar peduncle (increased EEG activation and synchronized bursts
of EEG activity in the theta band) are coherent with the SWC NREM SLEEP HOMEOSTASIS
modications after lesion of the superior cerebellar peduncle that The current dominant theory in sleep research considers that the
signicantly increase wakefulness and drowsiness while decreas- connections between structures putatively responsible for each of
ing REM and NREM sleep (De Andrs and Reinoso-Surez, 1979). the three phases of the SWC (wakefulness, REM, and NREM sleep)
The sleep suppression was a consequence of the decrease in both would provide the anatomical basis for the central organization
the duration and the number of the NREM episodes (Figure 8). of reciprocal interactions among these structures and result in
Also, changes in sleepwakefulness proportions occur after fasti- the alternation of the different phases of the SWC under the cir-
gial nuclei, cortical, or middle cerebellar peduncle lesions. All these cadian control of the suprachiasmatic nucleus (Reinoso-Surez
studies point to an important participation by the cerebellum et al., 2001). In addition, we would note that, in humans and

www.frontiersin.org November 2011 | Volume 2 | Article 70 | 9


de Andrs et al. Non-REM sleep functional anatomy

FIGURE 8 | Modifications in the total duration of wakefulness, (Slow Wave) and REM (Paradoxical) sleep. In contrast, the cats with
drowsiness, NREM (slow sleep), and REM (paradoxical) sleep, as lesions in the cerebellar cortex and white matter of the anterior
well as their episode mean duration and number per recording vermis showed a signicant decrease of drowsiness and a signicant
after different cerebellar lesions. The cats with lesions in the increase in REM sleep. Modied from De Andrs and Reinoso-Surez
brachium conjunctivum showed a signicant decrease of NREM (1979).

other mammals, sleep is not only regulated by the circadian clock, Neuronal activity during wakefulness increases the synthesis of
but also by sleep homeostasis. Humans are awake in the morning NREM sleep regulating molecules; this increase, as occurs with
because sleep pressure is low after a night of rest (Mignot and the effects of adenosine on the A1-receptor, leads to the inhibi-
Huguenard, 2009). Throughout the day increased wakefulness- tion of neuronal activity, thereby increasing sleep pressure, and
promoting signals, partly driven by the circadian clock, counter the nally NREM begins (Vassalli and Dijk, 2009). Although NREM-
growing sleep debt or sleep pressure, keeping the subject awake. An promoting substances act on basal forebrain and hypothalamic
opposite interaction occurs at night. Sleep would be necessary to NREM sleep regulating circuits (Gvilia et al., 2006a,b), they can
normalize the synapses to a basal condition that is sustainable and also act locally on the cerebral cortex and thalamus to change the
ensures cellular homeostasis (Cirelli and Tononi, 2008). NREM electrical properties of thalamic and cortical neurons thus alter-
sleep homeostasis depends not only on the duration of prior wake- ing their inputoutput relationships (Krueger et al., 2008). These
fulness but also on intensity, and sleep intensity increases when observations help explain the capacity of the isolated forebrain to
wakefulness is associated with learning (Cirelli, 2009). exhibit NREM sleep rebound (Corpas and De Andrs, 1991).
The metabolic factors that initiate sleep are molecules produced Currently, NREM and REM sleep are considered behavioral
by neuronal activity during wakefulness and they increase propor- states involving the whole organism and governed by central con-
tionally with the duration and intensity of arousal. The accumu- trol mechanisms. Although accepting this point of view, there
lation of metabolites up to a critical level would be involved in the is increasing evidence that NREM sleep (and thus sleep) might
initiation of sleep specically promoting NREM sleep. During the be a fundamental property of local neuronal networks since it
course of sleep metabolite levels would be reduced from the criti- can be initiated at small brain assemblies in response to their
cal concentration to basal levels (Datta and MacLean, 2007). Many use, and only later on be consolidated by general central mech-
molecules have been implicated in the processes of sleep regula- anisms (Krueger et al., 2008; Rector et al., 2009). One of the
tion, but only a few meet the criteria for being considered NREM smallest sleep units could be the individual cortical columns in
sleep-regulatory substances. Among these substances are: adeno- which sensory stimulation produces uctuating high and low
sine, nitric oxide, prostaglandin d2, interleukin-1, tumor necrosis amplitude evoked responses depending on the behavioral state.
factor, and growth hormone-releasing hormone (Krueger et al., In sleeping animals, the sleep-like response in cortical columns is
2008). These molecules are involved in regulating the intensity and characterized by evoked response potentials with a greater ampli-
duration of NREM sleep and they act in biochemical cascades. tude that the evoked responses during wakefulness (Rector et al.,

Frontiers in Neurology | Sleep and Chronobiology November 2011 | Volume 2 | Article 70 | 10


de Andrs et al. Non-REM sleep functional anatomy

2005). However, the sleep-/wake-like response also seems to be between hyperpolarized (DOWN) and depolarized (UP) periods
an intrinsic property of the individual cortical columns since, in which neurons are respectively silent or active (Dossi et al.,
independently of the behavioral state of the whole-animal, the 1992; Contreras and Steriade, 1995). More recently, Vyazovskiy
probability of nding a column in a sleep-like state depends on et al. (2009) have shown that during NREM, barrel cortex multi-
the length of time that column has previously spent in the wake- unit activity expresses cyclic (<1 Hz) changes with silent periods
like state (Rector et al., 2005), suggesting that in different cortical (OFF) alternating with others (ON) in which there is a sustained
regions cortical columns can be sleeping while others are awake ring activity; these ON/OFF cortical multiunit activities closely
during both whole-animal wakefulness and sleep. Thus, these nd- resemble the neuronal membrane potential events of UP/DOWN
ings suggest that NREM sleep may be regulated at local neuronal states. In addition, Vyazovskiy et al. (2009) also showed that the
assembly levels and this regulation appears to be a fundamental multiunit OFF periods in barrel cortical neurons are temporally
property of the neural networks and it depends on prior activity in correlated with the negative component of the EEG delta waves as
each network (Krueger et al., 2008). The NREM sleep of each local are the DOWN states that occur at cellular level (Contreras and Ste-
cortical column can be initiated by metabolically driven changes riade, 1995). During the rst NREM sleep after an uninterrupted
derived from the manufacture of molecules of sleep-regulatory waking period, neuronal barrel cortex population ON periods are
substances produced by neuronal activity during wakefulness-like short and frequent, while neuronal silence periods (DOWN states)
activity. The process may involve increased electrical activity, blood are long and frequent. DOWN states (accompanied by an overall
ow, extracellular levels of ATP and extra-and intracellular levels of decrease in neuronal activity) are a phenomenon that is thought to
adenosine, all of which would decrease the intracellular ATP level, be associated with energy savings. After a sustained sleep period,
and this would drive individual columns to enter a sleep-like activ- which reduces the sleep debt, barrel cortex OFF periods decrease
ity state. Once a nal sleep state was reached this process would be and the duration of the ON periods increases. In addition, the
reversed: reduced electrical activity, blood ow, extracellular levels changes in ring patterns in NREM sleep correlate positively with
of ATP and extra-and intracellular levels of adenosine would raise changes in SWA intensity (Vyazovskiy et al., 2009). Therefore, the
the intracellular ATP level in individual local cortical columns, and systematic increase of ring during wakefulness is balanced by
thus, the cortical column would be prepared for wakefulness. The staying asleep.
sleep or wakefulness states of individual cortical columns could Recent data (Jones et al., 2008) indicate that sleep reduces the
be synchronized through humoral and electrical connections and number of synaptic connections to a basic level that is augmented
therefore the sleep or wakefulness of the whole organism would during waking experience that is, NREM sleep seems to ensure
occur as an emergent property of the interaction of individual cell homeostasis; simultaneously during sleep the expression of
networks (Krueger et al., 2008). Nowadays there is an emerging molecules involved in synaptic reorganization is increased. Based
understanding of the role of astrocytes as potential mediators of on simple energy demands and since the cerebral cortexthalamus
the known effect of adenosine in sleep regulation and this medi- unit is a large part of the human and mammal brain, being awake
ation would involve both neuroenergetic and synaptic plasticity for longer and longer periods of time seems to be very expen-
roles (Jones, 2009). sive, hence energy economy by the brain during NREM sleep is
The power of delta SWA in the EEG is considered to be an one of the prevalent hypotheses to explain NREM sleep pressure
indicator of NREM sleep intensity, and delta SWA is homeosta- (Mignot and Huguenard, 2009). However, during NREM sleep
tically regulated since its power increases after wakefulness and neurons must down-re in synchrony rather than simply stay
returns to baseline during sleep. The magnitude of delta SWA is silent; for some authors this circumstance supports the need to
the best known correlation of sleep homeostasis or sleep pres- transfer memory to different cortical areas, as has been demon-
sure. Indeed, experiments have shown that the power of SWA strated to occur between the hippocampus and visual cortex. To
reects the duration of prior wakefulness regardless of the cir- Vyazovskiy et al. (2009), wakefulness is associated with learning,
cadian time. Delta SWA seems to reect a form of restorative and enhancing long-term potentiation strengthens glutamater-
process for cortical local circuits: SWA delta power is increased gic synapses, a process with an unsustainable energy require-
in cortical areas that have been most active during the day. Also, ment, whereas during sleep only robust connections remain intact,
imaging techniques have shown that there is a localized decrease reducing the energy requirements to the levels necessary for main-
in metabolic activity at night in areas that have been activated taining critical learned circuits. This reduction of synapses during
during the day (Mignot and Huguenard, 2009). SWA homeosta- sleep increases the signal to noise ratio for the remaining con-
sis may reect synaptic changes underlying a cellular need for nections, improving performance (Mignot and Huguenard, 2009;
sleep. Nevertheless, Huber et al. (2004) have indicated that sleep Vyazovskiy et al., 2009).
homeostasis has a local component, one that can be triggered in The close reciprocal connections between the thalamus and the
specic human brain regions by a learning task; these authors cerebral cortex would force NREM sleep homeostatic processes to
also showed that the local increase in SWA after learning corre- take place simultaneously in the two structures. The thalamus
lates with improved performance of the task after sleep. Thus, cerebral cortex is a strictly necessary unit in the expression
sleep homeostasis can be induced on a local level and can improve of the most signicant bioelectric and behavioral events of
performance. NREM sleep and the thalamuscerebral cortex unit is con-
As we have indicated above delta EEG waves are synchronized in nected to subcortical NREM sleep-promoting structures and
the different cortical areas in correlation with the neuronal events with all the subcortical structures involved in the organiza-
that express cyclic potential membrane uctuations (<1 Hz) tion of the other phases of SWC (Reinoso-Surez et al., 2011).

www.frontiersin.org November 2011 | Volume 2 | Article 70 | 11


de Andrs et al. Non-REM sleep functional anatomy

The former connections would act on the thalamuscerebral ACKNOWLEDGMENTS


cortex unit to facilitate the NREM sleep neurophysiological Grant BFU2009-06991/BFI from the Spanish Ministry of Science
and homeostatic processes and also through neurophysiologi- and Innovation supported this work. We acknowledge very valu-
cal and homeostatic mechanisms, the latter subcortical connec- able suggestions from Dr. C. de la Roza on the NREM sleep home-
tions would inhibit the other phases of SWC: REM sleep and ostasis section. We thank Ms. M. Callejo for technical assistance
wakefulness. and Ms. C. F. Warren for revision of English language usage.

REFERENCES Camacho-Evangelista, A., and Reinoso- De Andrs, I., and Corpas, I. (1991). sleep-wakefulness cycle after surgi-
Alam, M. N., McGinty, D., and Szymu- Surez, F. (1965). Effects on Morphine effects in brainstem- cal removal of the cerebellar cortex,
siak, R. (1995). Neuronal discharge the EEG of lesions in the trac- transected cats: II. Behavior and in Sleep 1978, eds L. Popoviciu, B.
of preoptic/anterior hypothalamic tus brachium conjunctivum, in sleep of the decerebrate cat. Behav. Asgian, and G. Badiu (Basel: Karger
thermosensitive neurons: relation to 6th International Congress Electroen- Brain Res. 44, 2126. Medical and Scientic Publishers),
NREM sleep. Am. J. Physiol. 269, cephalography Clinical Neurophysiol- De Andrs, I., Garzn, M., and Vill- 289292.
R1240R1249. ogy, Vienna, 467470. ablanca, J. R. (2003). The discon- Garzn, M. (1996). Estudio morfofun-
Amzica, F., and Steriade, M. (1995). Dis- Chou, T. C., Bjorkum, A. A., Gaus, S. E., nected brain stem does not support cional de los ncleos reticular oral y
connection of intracortical synaptic Lu, J., Scammell, T. E., and Saper, C. rapid eye movement sleep rebound reticular caudal del tegmento pon-
linkages disrupts synchronizatrion B. (2002). Afferents to the ventrolat- following selective deprivation. Sleep tino como regiones generadoras de
of a slow oscillation. J. Neurosci. 15, eral preoptic nucleus. J. Neurosci. 22, 26, 419425. sueo paradjico. Doctoral Thesis,
46584677. 977990. De Andrs, I., Gutirrez-Rivas, E., Universidad Autnoma de Madrid,
Andrade, K. C., Spoormaker, V. I., Cirelli, C. (2009). The genetic and mol- Nava, E., and Reinoso-Surez, F. Madrid.
Dresler, M., Wehrle, R., Holsboer, ecular regulation of sleep: from fruit (1976). Independence of sleep- Giannazzo, E., Manzoni, T., Raffaele, R.,
F., Smann, P. G., and Czisch, M. ies to humans. Nat. Rev. Neurosci. wakefulness cycle in an implanted Sapienza, S., and Urbano, A. (1968).
(2011). Sleep spindles and hip- 10, 549560. head encephale isole. Neurosci. Lett. Changes in the sleep-wakefulness
pocampal functional connectivity in Cirelli, C., and Tononi, G. (2008). Is 2, 1318. cycle induced by chronic fastigial
human NREM sleep. J. Neurosci. 31, sleep essential? PLoS Biol. 6, e216. De Andrs, I., and Reinoso-Surez, F. lesions in the cat. Brain Res. 11,
1033110339. doi:10.1371/journal.pbio.0060216 (1979). Participation of the cerebel- 281284.
Andre, P., and Arrighi, P. (2003). Hipnic Contreras, D., and Steriade, M. (1995). lum in the regulation of the sleep- Golanov, E. V., and Reis, D. J. (2001).
modulation of cerebellar informa- Cellular basis of EEG slow rhythms: wakefulness cycle through the supe- Neurons of nucleus of the solitary
tion processing: implications for the a study of dynamic corticothala- rior cerebellar peduncle. Arch. Ital. tract synchronize the EEG and ele-
cerebro-cerebellar dialogue. Cerebel- mic relationships. J. Neurosci. 15, Biol. 117, 140163. vate cerebral blood ow via a novel
lum 2, 8495. 604622. Dossi, R. C., Nuez, A., and Steriade, M. medullary area. Brain Res. 892, 112.
Avendao, C., Stepniewska, I., Rausell, Cordeau, J. P., and Mancia, M. (1959). (1992). Electrophysiology of a slow Gong, H., McGinty, D., Guzman-Marin,
E., and Reinoso-Surez, F. (1990). Evidence for the existence of an elec- (0.5-4 Hz) intrinsic oscillation of cat R., Chew, K. T., Stewart, D., and
Segregation and heterogeneity of troencephalographic synchroniza- thalamocortical neurones in vivo. J. Szymusiak, R. (2004). Activation of
thalamic cell populations pro- tion mechanism originating in the Physiol. 447, 215434. c-fos in GABAergic neurones in
jecting to supercial layers of lower brain stem. Electroencephalogr. Eguchi, K., and Satoh, T. (1980). Con- the preoptic area during sleep and
the posterior parietal cortex: a Clin. Neurophysiol. 3, 551564. vergence of sleep-wakefulness sub- in response to sleep deprivation. J.
retrograde tracing study in the Corpas, I., and De Andrs, I. (1991). systems onto single neurons in the Physiol. 556, 935946.
cat and monkey. Neuroscience 39, Morphine effects in brainstem tran- region of cats solitary tract nucleus. Gottesmann, C. (1996). The transition
547559. sected cats: I. EEG and sleep- Arch. Ital. Biol. 118, 331345. from slow-wave sleep to paradoxi-
Bal, T., and McCormick, D. A. (1993). wakefulness in the isolated fore- Fadiga, E., Manzoni, T., Sapienza, S., and cal sleep: evolving facts and concepts
Mechanisms of oscillatory activ- brain. Behav. Brain Res. 44, 1119. Urbano, A. (1968). Synchronizing of the neurophysiological processes
ity in guinea-pig nucleus reticu- Cunchillos, J. D., and De Andrs, I. and desynchronizing fastigial inu- underlying the intermediate stage of
laris thalami in vitro: a mammalian (1982). Participation of the cere- ences on the electrocortical activity sleep. Neurosci. Biobehav. Rev. 20,
pacemaker. J. Physiol. (Lond.) 468, bellum in the regulation of the of the cat, in acute experiments. Elec- 367387.
669691. sleep-wakefulness cycle. Results in troencephalogr. Clin. Neurophysiol. Guglielmino, S., and Strata, P. (1971).
Batini, C., Moruzzi, G., Palestini, M., cerebellectomized cats. Electroen- 24, 330342. Cerebellum and atonia of the desyn-
Rossi, G. F., and Zanchetti, A. (1958). cephalogr. Clin. Neurophysiol. 53, Fuentealba, P., and Steriade, M. (2005). chronized phase of sleep. Arch. Ital.
Persistent patterns of wakefulness 549558. The reticular nucleus revisited: Biol. 109, 210217.
in the pretrigeminal midpontine Dang-Vu, T. T., Schabus, M., Desseilles, intrinsic and network properties of Guzmn-Marn, R., Alam, M. N., Szy-
preparation. Science 128, 3032. M., Albouy, G., Boly, M., Darsaud, a thalamic pacemaker. Prog. Neuro- musiak, R., Drucker-Coln, R., Gong,
Berlucchi, G., Maffei, L., Moruzzi, G., A., Gais, S., Rauchs, G., Sterpenich, biol. 75, 125141. H., and McGinty, D. (2000). Dis-
and Strata, P. (1964). EEG and V., Vandewalle, G., Carrier, J., Moo- Gadea-Ciria, M., and Fuentes, J. (1976). charge modulation of rat dorsal
behavioral effects elicited by cooling nen, G., Balteau, E., Degueldre, C., Analysis of phasic activities in the raphe neurons during sleep and
of medulla and pons. Arch. Ital. Biol. Luxen, A., Phillips, C., and Maquet, lateral rectus muscle of the eyes waking: effects of preoptic/basal
102, 372392. P. (2008). Spontaneous neural activ- (PALRE) during paradoxical sleep forebrain warming. Brain Res. 875,
Bonjean, M., Baker, T., Lemieux, M., ity during human slow wave sleep. in chronic cerebellectomized cats. 2334.
Timofeev, I., Sejnowski, T., and Proc. Natl. Acad. Sci. U.S.A. 105, Brain Res. 111, 416415. Gvilia, I., Turner, A., McGinty, D., and
Bazhenov, M. (2011). Corticothal- 1516015165. Gallopin, T., Fort, P., Eggermann, E., Szymusiak, R. (2006a). Preoptic area
amic feedback controls sleep spin- Datta, S., and MacLean, R. R. (2007). Cauli, B., Luppi, P. H., Rossier, J., neurons and the homeostatic regula-
dle duration in vivo. J. Neurosci. 31, Neurobiological mechanisms for the Audinat, E., Mhlethaler, M., and tion of rapid eye movement sleep. J.
91249134. regulation of mammalian sleep- Seran, M. (2000). Identication of Neurosci. 26, 30373044.
Camacho-Evangelista, A., and Reinoso- wake behavior: reinterpretation of sleep-promoting neurons in vitro. Gvilia, I., Xu, F., McGinty, D., and
Surez, F. (1964). Activating and historical evidence and inclusion of Nature 404, 992995. Szymusiak, R. (2006b). Homeostatic
synchronizing centers in cat brain: contemporary cellular and molecu- Garca-Ura, J., De Andrs, I., Cun- regulation of sleep: a role for pre-
electroencephalograms after lesions. lar evidence. Neurosci. Biobehav. Rev. chillos, J. D., and Reinoso-Surez, optic area neurons. J. Neurosci. 26,
Science 146, 268270. 31, 775824. F. (1980). Modication of the 94269433.

Frontiers in Neurology | Sleep and Chronobiology November 2011 | Volume 2 | Article 70 | 12


de Andrs et al. Non-REM sleep functional anatomy

Hernndez-Pen, R. (1962). Sleep Laguzzi, R., Reis, D. J., and Talman, Nuez, A., Curr Dossi, R., Contreras, del tracto solitario. Efectos sobre el
induced by localized electrical or W. T. (1984). Modulation of cardio- D., and Steriade, M. (1992). Intra- ciclo vigilia-sueo e interaccin con
chemical stimulation of the fore- vascular and electrocortical activity cellular evidence for incompatibility efectos vegetativos. Doctoral Thesis,
brain. Electroencephalogr. Clin. Neu- through serotonergic mechanisms in between spindle and delta oscilla- Universidad Autnoma de Madrid,
rophysiol. 14, 423424. the nucleus tractus solitarius of the tions in thalamocortical neurons of Madrid.
Hess, W. R. (1931). Le sommeil. C. R. rat. Brain Res. 304, 321328. cat. Neuroscience 48, 7585. Reinoso-Barbero, F., and De Andrs, I.
Soc. Biol. Paris 107, 13331360. Llins, R., and Steriade, M. (2006). Nez-Molina, A., and Amzica, F. (1995). Effects of opioid microin-
Hess, W. R. (1968). Psychologie in biol- Bursting of thalamic neurons and (2004). Mecanismos de generacin jections in the nucleus of the soli-
ogisher Sicht, 2 Au. Stuttgart: G. states of vigilance. J. Neurophysiol. de las oscilaciones lentas del elec- tary tract on the sleep-wakefulness
Thieme. 95, 32973308. troencefalograma durante el sueo. cycle states in cats. Anesthesiology 82,
Hodes, R. (1964). Electrocortical desyn- Madoz, P., and Reinoso-Surez, F. Rev. Neurol. 39, 628633. 144152.
chronization resulting from spinal (1968). Inuence of lesions in pre- Oka, H., Ito, J., and Kawamura, M. Reinoso-Surez, F. (1954). Die
block: evidence for synchronizing optic region on the states of sleep (1982). Identication of thalamo- Auswirkungen der Ausschal-
inuences in the cervical cord. Arch. and wakefulness. Proc. XXIV Int. cortical neurons responsible for cor- tung eines Nucleus ruber auf
Ital. Biol. 102, 183196. Cong. Physiol. Sci. 7, 276. tical recruiting and spindling activi- die Hirnrinde (Electroencephalo-
Huber, R., Ghilardi, M. F., Massimini, Magnes, J., Moruzzi, G., and Pom- ties in cats. Neurosci. Lett. 33, 1318. graphische Untersuchungen an der
M., and Tononi, G. (2004). Local peiano, O. (1961). Synchronization Paz, C., Reygadas, E., and Fernndez- Katze). Dtsch. Z. Nervenheilk 172,
sleep and learning. Nature 430, of the EEG produced by low- Guardiola, A. (1982). Sleep alter- 201219.
7881. frequency electrical stimulation of ations following total cerebellec- Reinoso-Surez, F. (1961). Topographis-
Iber, C., Ancoli-Israel, S., Chesson, A., the region of the solitary tract. Arch. tomy in cats. Sleep 5, 218226. cher Hirnatlas der Katze fr Exper-
Quan, S. F. (eds). (2007). The Ital. Biol. 99, 3367. Peigneux, P., Laureys, S., Fuchs, S., imental Physiologische Untersuchun-
AASM manual for the scoring of Mancia, M., Meulders, M., and San- Collette, F., Perrin, F., Reggers, J., gen. Darmstadt: Merck AG.
sleep and associated events: rules, tibanez, H. G. (1959). Synchronisa- Phillips, C., Degueldre, C., Del Fiore, Reinoso-Surez, F. (1992). Considera-
terminology, and technical specica- tion de llectroencphalogramme G., Aerts, J., Luxen, A., and Maquet, tions on thalamic and pontine sleep
tion,1st Edn. Westchester, IL: Amer- provoque par la stimulation P. (2004). Are spatial memories mechanisms, in Fundamental Neu-
ican Academy of Sleep Medicine. visuelle rptitive chez le chat strengthened in the human hip- robiology, ed. R. Velluti (Montev-
Jahnsen, H., and Llins, R. (1984). mdiopontin prtrigminal. Arch. pocampus during slow wave sleep? ideo: Work), 5971.
Electrophysiological properties of Int. Physiol. Biochem. 67, 661670. Neuron 44, 535545. Reinoso-Surez, F. (1993). Algunas con-
guinea-pig thalamic neurons: and Marshall, L., Helgadttir, H., Mlle, M., Pompeiano, O., and Swett, J. E. (1962). sideraciones sobre la anatoma del
in vitro study. J. Physiol. (Lond.) 349, and Born, J. (2006). Boosting slow EEG behavioural manifestation of sueo. An. Anat. 39, 165181.
205226. oscillations during sleep potentiates sleep induced by cutaneous nerve Reinoso-Surez, F., and De Andrs, I.
Ji, D., and Wilson, M. A. (2007). Coor- memory. Nature 444, 610613. stimulation in normal cats. Arch. (1976). Brain structures and sleep.
dinated memory replay in the visual McGinty, D., and Szymusiak, R. (2000). Ital. Biol. 100, 311342. Trab. Inst. Cajal Invest. Biol. 68,
cortex and hippocampus during The sleep-wake switch: a neuronal Puizillout, J. J., Ternaux, J. P., Foutz,A. S., 3968.
sleep. Nat. Neurosci. 10, 100107. alarm clock. Nat. Med. 6, 510511. and Dell, P. (1973). Slow wave sleep Reinoso-Surez, F., De Andrs, I., and
Jimnez-Castellanos, J., and Reinoso- McGinty, D. J., and Sterman, M. B. with phasic discharges. Triggering by Garzn, M. (2011). Functional
Surez, F. (1985). Topographical (1968). Sleep suppression after basal vago-aortic stimulation. Rev. Elec- anatomy of the sleep-wakefulness
organization of the afferent connec- forebrain lesions in the cat. Science troencephalogr. Neurophysiol. Clin. 3, cycle: wakefulness. Adv. Anat.
tions of the principal ventro-medial 160, 12531255. 2137. Embryol. Cell. Biol. 208, 1128.
thalamic nucleus in the cat. J. Comp. Mignot, E., and Huguenard, J. R. Raffaele, R., Sapienza, S., Urbano, A., Reinoso-Surez, F., De Andrs, I.,
Neurol. 236, 297314. (2009). Resting our cortices by and Ventura, M. (1971). Changes in Rodrigo-Angulo, M. L., and Garzn,
Jones, B. E. (2009). Glia, adenosine, and going DOWN to sleep. Neuron 63, the sleep-wakefulness rhythm after M. (2001). Brain structures and
sleep. Neuron 61, 156157. 719721. chronic bilateral interruption of the mechanisms involved in the gener-
Jones, E. G. (2001). The thala- Moreno-Balandrn, E., Garzn, M., middle cerebellar peduncles in the ation of REM sleep. Sleep Med. Rev.
mic matrix and thalamocortical Bdalo, C., Reinoso-Surez, F., cat. Brain Res. 26, 195199. 5, 6378.
synchrony. Trends Neurosci. 24, and De Andrs, I. (2008). Sleep- Rai, S., Kumar, S., Alam, M. A., Reinoso-Surez, F., de la Roza, C.,
595601. wakefulness effects after microinjec- Szymusiak, R., McGinty, D., and Rodrigo-Angulo, M. L., de Andrs,
Jones, S., Pster-Genskow, M., Benca, tions of hypocretin 1 (orexin A) in Alam, M. N. (2010). A1 receptor I., Nez, A., and Garzn, M. (2010).
R. M., and Cirelli, C. (2008). Mol- cholinoceptive areas of the cat oral mediated adenosinergic regulation GABAergic mechanisms in the ven-
ecular correlates of sleep and wake- pontine tegmentum. Eur. J. Neurosci. of perifornical-lateral hypothalamic tral oral pontine tegmentum the
fulness in the brain of the white- 28, 331341. area neurons in freely behaving rat. REM sleep induction site- in the
crowned sparrow. J. Neurochem. 105, Morrison, R. S., and Dempsey, E. Neuroscience 167, 4048. modulation of sleep-wake states,
4662. W. (1942). A study of thalamo- Rasch, B., Bchel, C., Gais, S., and in GABA and Sleep, ed. J. M.
Jouvet, M. (1962). Research on the cortical relations. Am. J. Physiol. 135, Born, J. (2007). Odor cues during Monti, S. R. Pandi-Perumal, and H.
neural structures and responsible 282302. slow-wave sleep prompt declarative Mhler (Basel: Birkhauser-Verlag),
mechanisms in different phases of Moruzzi, G. (1958). Relations between memory consolidation. Science 315, 233252.
physiological sleep. Arch. Ital. Biol. the cerebellum and other central 14261429. Reinoso-Surez, F., Fairn, A., Martnez-
100, 125206. structures. Effects on the electro- Rector, D. M., Schei, J. L., Van Don- Moreno, E., Nava, B. E., and Pasquier,
Krilowicz, B. L., Szymusiak, R., and corticogram, in The Physiology and gen, H. P., Belenky, G., and Krueger, D. A. (1977). Ascending projections
McGinty, D. (1994). Regulation Pathology of the Cerebellum, eds H. R. J. M. (2009). Physiological markers from the brain stem. Med. Biol. Pap.
of posterior lateral hypothalamic Dow and G. Moruzzi (Minneapolis: of local sleep. Eur. J. Neurosci. 29, 1, 233246.
arousal related neuronal discharge The University of Minnesota Press), 17711778. Reinoso-Surez, F., Sierra, G., and
by preoptic anterior hypothalamic 323333. Rector, D. M., Topchiy, I. A., Carter, K. Camacho, A. (1962). Efectos de
warming. Brain Res. 668, 3038. Moruzzi, G. (1972). The sleep-waking M., and Rojas, M. J. (2005). Local lesiones en formacin reticular
Krueger, J. M., Rector, D. M., Roy, S., cycle. Ergeb. Physiol. 64, 1165. functional state differences between protuberancial e istmo ponto-
Van Dongen, H. P., Belenky, B., and Nauta, W. J. H. (1946). Hypothala- rat cortical columns. Brain Res. 1047, mesenceflico sobre el EEG del gato
Panksepp, J. (2008). Sleep as a funda- mic regulation of sleep in rats. An 4555. (Su participacin en el mecanismo
mental property of neuronal assem- experimental study. J. Neurophysiol. Reinoso-Barbero, F. (1993). Microestim- del sueo). Rev. Med. Univ. Navarra
blies. Nat. Rev. Neurosci. 9, 910919. 9, 285316. ulacin con opiaceos en la region 6, 119.

www.frontiersin.org November 2011 | Volume 2 | Article 70 | 13


de Andrs et al. Non-REM sleep functional anatomy

Rossi, G. F., Minobe, K., and Candia, monoaminergic cell groups. J. thalamus of intact-cortex and decor- forebrain and of the brainstem in the
O. (1963). An experimental study Comp. Neurol. 429, 638653. ticated cats. J. Neurophysiol. 76, control of the sleep-waking system.
of the hypnogenic mechanisms of Steriade, M., and Deschenes, M. (1988). 41524168. J. Sleep Res. 13, 179208.
the brain stem. Arch. Ital. Biol. 101, Intrathalamic and brainstem- Ursin, R., and Sterman, M. B. (1981). Vies-Morros, R. (1959). Vias trigem-
470492. thalamic networks envolved in A Manual for Standardized Scor- inales y actividad bioelctrica cere-
Rubio-Garrido, P., Prez-de-Manzo, F., resting and alert status, in Cellular ing of Sleep and Waking States in bral. An. Anat. 10, 507527.
Porrero, C., Galazo, M. J., and Clasc, Thalamic Mechanisms, eds M. the Adult Cat. Los Angeles: Univer- von Economo, C. (1930). Sleep as a
F. (2009). Thalamic input to distal Bentivoglio and R. Spreaco (New sity of California Brain Information problem of localization. J. Nerv.
apical dendrites in neocortical layer Cork: Elsevier), 3762. Service. Ment. Dis. 71, 249259.
1 is massive and highly convergent. Steriade, M., Deschnes, M., Domich, Uschakov, A., Gong, H., McGinty, D., Vyazovskiy, V. V., Olcese, U., Lazimy,
Cereb. Cortex 19, 23802395. L., and Mulle, C. (1985). Abolition and Szymusiak, R. (2006). Sleep- Y. M., Faraguna, U., Esser, S. K.,
Saper, C. B. (1995). Central autonomic of spindle oscillations in thalamic active neurons in the preoptic area Williams, J. C., Cirelli, C., and
system, in The Rat Nervous System, neurons disconnected from nucleus project to the hypothalamic par- Tononi, G. (2009). Cortical ring
ed. G. Paxinos (San Diego: Academic reticularis thalami. J. Neurophysiol. aventricular nucleus and periforni- and sleep homeostasis. Neuron 63,
Press), 107135. 54, 14731497. cal lateral hypothalamus. Eur. J. Neu- 865878.
Saper, C. B., Chou, T. C., and Scam- Steriade, M., Dossi, R. C., and Nuez, rosci. 23, 32843296. Yang, Q. Z., and Hatton, G. I.
mell, T. E. (2001). The sleep switch: A. (1991). Network modulation Valds-Cruz, A., Magdaleno-Madrigal, (1997). Electrophysiology of exci-
hypothalamic control of sleep and of a slow intrinsic oscillation of V. M., Martnez-Vargas, D., tatory and inhibitory afferents to
wakefulness. Trends Neurosci. 24, cat thalamocortical neurons impli- Fernndez-Mas, R., and Almazn- rat histaminergic tuberomammil-
726731. cated in sleep delta waves: corti- Alvarado, S. (2008). Long-term lary nucleus neurons from hypothal-
Sasaki, K., Kawaguchi, S., Oka, H., Sakai, cally induced synchronization and changes in sleep and electroen- amic and forebrain sites. Brain Res.
M., and Mizuno, N. (1976). Elec- brainstem cholinergic suppression. cephalographic activity by chronic 773, 162172.
trophysiological studies on the cere- J. Neurosci. 10, 32003217. vagus nerve stimulation in cats. Yoshida, K., McCormack, S., Espaa, R.
bello cerebral projections in mon- Steriade, M., and McCarley, R. W. Prog. Neuropsychopharmacol. Biol. A., Crocker, A., and Scammell, T. E.
keys. Exp. Brain Res. 24, 495507. (1990). Brainstem Control of Wake- Psychiatry 32, 828834. (2006). Afferents to the orexin neu-
Sforza, E., Montagna, P., Tinuper, fulness and Sleep. New York: Plenum. Vassalli, A., and Dijk, D.-J. (2009). Sleep rons of the rat brain. J. Comp. Neurol.
P., Cortelli, P., Avoni, P., Fer- Steriade, M., Nuez, A., and Amz- function: current questions and new 494, 845861.
rillo, F., Petersen, R., Gambetti, P., ica, F. (1993). Intracellular analysis approaches. Eur. J. Neurosci. 29, Zarranz, J., and Reinoso-Surez, F.
and Lugaresi, E. (1995). Sleep-wake of relations between the slow (< 18301841. (1971). The pontine tegmentum
cycle abnormalities in fatal familial 1 Hz) neocortical oscillation and Velayos, J. L., Jimnez-Castellanos, J. and sleep-wakefulness states. XXV
insomnia. Evidence of the role of the other sleep rhythms of the elec- Jr., and Reinoso-Surez, F. (1989). Internat. Cong. Physiol. Sci. Munich,
thalamus in sleep regulation. Elec- troencephalogram. J. Neurosci. 13, Topographical organization of the 618.
troencephalogr. Clin. Neurophysiol. 32663283. projections from the reticular thal-
94, 398405. Steriade, M., Oakson, G., and Ropert, N. amic nucleus to the intralami- Conict of Interest Statement: The
Sherin, J. E., Elmquist, J. K., Torrealba, (1982). Firing rates and patterns of nar and medial thalamic nuclei authors declare that the research was
F., and Saper, C. B. (1998). Innerva- midbrain reticular neurons during in the cat. J. Comp. Neurol. 279, conducted in the absence of any
tion of histaminergic tuberomam- steady and transitional states of the 457469. commercial or nancial relationships
millary neurons by GABAergic and sleep-waking cycle. Exp. Brain Res. Villablanca, J. R. (1965). The elec- that could be construed as a potential
galaninergic neurons in the ventro- 46, 3751. trocorticogram in the chronic conict of interest.
lateral preoptic nucleus of the rat. J. Sterman, M. B., and Clemente, C. cerveau isol of the cat. Electroen-
Neurosci. 18, 47054721. D. (1962). Forebrain inhibitory cephalogr. Clin. Neurophysiol. 19, Received: 27 July 2011; paper pend-
Sherin, J. E., Shiromani, P. J., McCar- mechanisms: sleep patterns induced 576586. ing published: 17 August 2011; accepted:
ley, R. W., and Saper, C. B. (1996). by basal forebrain stimulation in Villablanca, J. R. (1966). Behavioral and 26 October 2011; published online: 15
Activation of ventrolateral preoptic the behaving cat. Exp. Neurol. 6, polygraphic study of sleep and November 2011.
neurons during sleep. Science 271, 103117. wakefulnessin chronic decerebrate Citation: de Andrs I, Garzn M and
216219. Szymusiak, R., Alam, N., Steininger, T. cats. Electroencephalogr. Clin. Neuro- Reinoso-Surez F (2011) Functional
Siegel, J. M., Tomaszewsky, K. S., L., and McGinty, D. (1998). Sleep- physiol. 21, 562577. anatomy of non-REM sleep. Front. Neur.
and Nienhuis, R. (1986). Behavioral waking discharge patterns of ventro- Villablanca, J. R. (1972). Permanent 2:70. doi: 10.3389/fneur.2011.00070
states in the chronic medullary and lateral preoptic/anterior hypothala- reduction in sleep after removal of This article was submitted to Frontiers in
midpontine cat. Electroencephalogr. mic neurons in rats. Brain Res. 803, cerebral cortex and striatum in cats. Sleep and Chronobiology, a specialty of
Clin. Neurophysiol. 63, 274288. 178188. Brain Res. 36, 463468. Frontiers in Neurology.
Starzl, T. E., and Magoun, H. W. (1951). Szymusiak, R., Gvilia, I., and McGinty, Villablanca, J. R. (1974). Role of the Copyright 2011 de Andrs, Garzn and
Organization of the diffuse thalamic D. (2007). Hypothalamic control of thalamus in sleep control: sleep- Reinoso-Surez. This is an open-access
projection system. J. Neurophysiol. sleep. Sleep Med. 8, 291301. wakefulness studies in chronic dien- article subject to a non-exclusive license
14, 133146. Szymusiak, R., and McGinty, D. (1986). cephalic and athalamic cats, in Basic between the authors and Frontiers Media
Steininger, T. L., Gong, H., McGinty, Sleep suppression following kainic Sleep Mechanisms, eds O. Petre- SA, which permits use, distribution and
D., and Szymusiak, R. (2001). acid-induced lesions of the basal Quadens and J. Schlag (New York: reproduction in other forums, provided
Subregional organization of pre- forebrain. Exp. Neurol. 94, 598614. Academic), 5181. the original authors and source are cred-
optic area/anterior hypothalamic Timofeev, I., and Steriade, M. (1996). Villablanca, J. R. (2004). Counter- ited and other Frontiers conditions are
projections to arousal-related Low-frequency rhythms in the pointing the functional role of the complied with.

Frontiers in Neurology | Sleep and Chronobiology November 2011 | Volume 2 | Article 70 | 14

You might also like