Anda di halaman 1dari 11

REVIEWS

SLEEP

Sleep as a fundamental property


of neuronal assemblies
James M. Krueger*‡, David M. Rector‡, Sandip Roy§, Hans P. A. Van Dongen‡,
Gregory Belenky‡ and Jaak Panksepp‡
Abstract | Sleep is vital to cognitive performance, productivity, health and well-being. Earlier
theories of sleep presumed that it occurred at the level of the whole organism and that it was
governed by central control mechanisms. However, evidence now indicates that sleep might
be regulated at a more local level in the brain: it seems to be a fundamental property of
neuronal networks and is dependent on prior activity in each network. Such local-network
sleep might be initiated by metabolically driven changes in the production of sleep-regulatory
substances. We discuss a mathematical model which illustrates that the sleep-like states of
individual cortical columns can be synchronized through humoral and electrical
connections, and that whole-organism sleep occurs as an emergent property of
local-network interactions.

The scientific community has yet to reach a consensus regulation and organization. We also discuss a quantita-
as to exactly what it is, in body and brain, that sleeps, tive model of how synchrony of cortical-column states
and why we sleep. This lack of consensus confuses our within cortical regions is achieved and how it enables the
understanding and confounds discussions of sleep. emergence of whole-organism sleep. Finally, the implica-
How can we explain the regulation of sleep if we do tions of our theory for sleep function and consciousness
not know, or have testable theories of, how the brain are briefly presented.
is organized to produce sleep and exactly what sleeps?
Traditionally, sleep has been considered a property of Sleep is a local process
the whole organism: an animal was either awake or Clinical evidence suggests that sleep can be a property
drowsy or asleep. This idea was derived in part from of something less than the whole brain. Mahowald and
the observation that sleep is homeostatic: prolonged Schenck7 describe patients with parasomnias, such as
wakefulness is followed by rebound sleep. Indeed, there sleep walking, and propose that these individuals are
are global ways to induce sleep-like states in animals simultaneously awake (evidenced by their ability to
through brain stimulation1 or through the induction negotiate objects) and asleep (indicated by their lack
*Department of VCAPP,
College of Veterinary of global metabolic2 or neurochemical3 changes in the of awareness of their actions). Studies of strokes and
Medicine, Washington State body (BOX 1). In the past few years, however, evidence other brain lesions indicate that if a human or animal
University, PO BOX 646520, has emerged that supports a new theory of sleep: that it survives acute brain damage for more than a few days,
Pullman, Washington is local and use-dependent 4,5. According to this theory, sleep reemerges: despite there being millions of cases,
99164-6520, USA.

Sleep and Performance
the homeostatic regulation of sleep is not at the level we know of no instance in which there was complete
Research Center and of the whole organism: it occurs in any brain region in insomnia following survivable acute brain lesions. This
Programs in Neuroscience; response to use. finding suggests that sleep is a robust fundamental
and §Electrical Engineering, In this Review, we present a comprehensive mecha- self-organizing property of any group of neurons.
Washington State University,
nistic view of the brain organization of sleep. First, we Naturally occurring patterns of normal sleep also
Pullman and Spokane,
Washington 99164-6520, discuss the recent evidence that directly demonstrates suggest that parts of the brain can be awake while other
USA. that a sleep-like state can exist in cortical columns6, parts are asleep. For example, dolphins do not exhibit
Correspondence to J.M.K. show that such local sleep-like states depend on the prior high-amplitude electroencephalogram (EEG) delta
e-mail: Krueger@vetmed. activity of the columns, and discuss the role of sleep- waves simultaneously in both cerebral hemispheres 8.
wsu.edu
doi:10.1038/nrn2521
regulatory substances (SRSs) in the regulation of local High-amplitude EEG delta waves are a defining charac-
Published online sleep. We then present a hypothesis that mechanisti- teristic of non-rapid-eye-movement sleep (NREMS) (BOX 1).
5 November 2008 cally links metabolism and brain activity to local-sleep Other species of whales, other marine mammals such

910 | DECEMBER 2008 | VOLUME 9 www.nature.com/reviews/neuro


REVIEWS

Cortical columns as seals, and birds also exhibit unihemispheric sleep9. Cortical columns are anatomically well-defined
Collections of highly In humans, as NREMS epochs unfold, the EEG delta- examples of neuronal networks and are also called neu-
interconnected neurons that wave power increases in the frontal cortex earlier than ronal assemblies; they are thought to be a basic process-
often focus on particular tasks. in more posterior cortical areas, suggesting differential ing unit of the waking brain15. Direct evidence indicates
Columns are layered structures,
and the individual layers are
sleep intensities in different brain regions10. Similarly, that cortical columns oscillate between functional states
concerned with different measurements of cerebral blood flow show regional dif- as defined by changing input–output relationships: if rat
functions, such as the receipt of ferences during NREMS and rapid-eye-movement sleep cortical columns are probed using afferent sensory stim-
afferent input and producing an (REMS)11. Collectively, these data suggest that sleep — ulation and the subsequent amplitudes of the induced
output. There is more
or at least correlates of sleep or sleep intensity, such as evoked response potentials are measured, awake-like and
intercellular connectivity within
columns than between columns.
EEG delta-wave power and blood flow — is a regional sleep-like states can be distinguished6. The sleep-like
This makes each column a phenomenon in the brain. state is characterized by evoked response potentials of
functional unit. In humans, each In cats, if cortical islands are prepared surgically in greater magnitude than the evoked response potentials
column contains 1,000–10,000 such a way that they retain their blood flow but not that occur during waking. During whole-animal sleep,
cells and there are ~100,000
cortical columns.
their thalamic inputs, the local field potentials from most of the cortical columns are in this sleep-like state.
those islands wax and wane over periods of 10–20 min- Conversely, when an animal is awake, most of the cortical
Parasomnias utes through episodes with high-amplitude EEG delta columns are in an awake-like state that is characterized
States in which behavioural or waves12. More-recent data also indicate that there is an by low-amplitude evoked response potentials. However,
physical phenomena that are
EEG slow-wave component (0.5–1.5 Hz) that originates the findings that columns can exist in a sleep-like state
characteristic of waking occur
while parts of the body seem
locally in the cortex 13. Such findings indicate that high- during whole-animal wake episodes and, conversely, that
to be asleep. Sleep walking is a amplitude EEG delta waves are a local property of corti- columns can exist in awake-like states during whole-
well-known example of a cal tissue. In monkeys that are at the transition between animal sleep suggest that sleep is a property of individual
parasomnia. wakefulness and sleep, some neurons in the visual cortex cortical columns6.
display the characteristic sleep patterns of hyperpolariza- The cortical-column sleep-like state, like whole-
tion followed by bursting action potentials even when animal sleep, is predictable and homeostatically regu-
the animal is performing a visual discrimination task14. lated: the probability of finding a given column in the
The coordination of such firing patterns contributes sleep-like state is dependent on the length of time that
to the sleep phenotype of EEG delta-wave power 13. the column has spent in the awake-like state. In other
Although the animal is awake, the cellular processes words, the longer a column is in an awake-like state,
that are characteristic of NREMS are evident, suggest- the higher the likelihood that it will make the transition
ing that there are local islands of sleep. Thus, we infer to the sleep-like state6. Such changes also manifest in
that these islands can integrate sleep, waking and behavioural consequences and thereby can be defined
vigilance transition states locally. as functional-state changes. Rats that are subjected to
a conditioned-learning paradigm in which they are
trained to lick in response to stimulation of a single
Box 1 | Sleep
facial whisker show greater incidence of errors of com-
Sleep sustains physical and cognitive performance, productivity, health and well-being; mission (licking when the whisker is not stimulated) and
even mild sleep restriction degrades performance over a few days. However, it is omission (failure to lick in response to whisker stimu-
difficult to define sleep as there are no direct measures of it. Instead, sleep is inferred lation) if the cortical column that receives the whisker
from a variety of measures, including electrical activity in the brain (through electro- input is in the sleep-like state16. Thus, cortical columns
encephalograms (EEGs)) and in muscles (through electromyograms), brain
might be the minimal unit of brain that can manifest a
temperature, behaviour, posture and heart rate. Using those measures one
characterizes epochs (usually less than one minute) as being in a particular state and
sleep-like state4,6. It is possible that individual cells sleep
then determines parameters, such as EEG power, that correlate with the state. independently of other cells, but this will be difficult to
In almost all mammals there are two forms of sleep: non-rapid-eye-movement sleep demonstrate because we currently cannot causally link
(NREMS) and rapid-eye-movement sleep (REMS). In humans, NREMS occupies ~80% the electrical or metabolic activity of individual brain
and REMS ~20% of sleep time. These two forms alternate with a periodicity of ~90–100 cells and sleep, despite the fact that there are numer-
minutes, with more NREMS occurring during the first half of the night and more REMS ous examples in the literature of correlations between
occurring during the second half. Despite this complex architecture, sustaining single-neuron activity and sleep13,14.
performance during waking is dependent on total sleep time. Central sleep-regulatory In summary, the data reviewed in this section
circuits seem to consolidate sleep into one or two daily bouts in humans. Circuits that indicate that sleep can be a property of local neuro-
regulate NREMS include the preoptic anterior hypothalamus, within which are found
nal assemblies. Even though there are central global
the ventrolateral preoptic area and the median preoptic area, and the basal forebrain57.
REMS and the alternation between NREMS and REMS are also under central control.
coordinators of sleep–waking states, such as the clock
The areas that are responsible for these functions include the pedunculopontine mechanisms of the suprachiasmatic nuclei17, we suggest that
tegmental nucleus and the laterodorsal tegmental nucleus in the brainstem. There are global coordination of NREMS is not due to a single
also multiple wake-promoting networks, including the hypocretin/orexin system in the sleep generator: it might largely reflect an emergent
lateral hypothalamus, the degeneration of which is responsible for the clinical property of loosely coupled local processes. This may
condition of narcolepsy. help to explain the common observation in human
The currently dominant paradigm in sleep research views whole-organism sleep as sleep laboratories that EEG indices of NREMS do not
being initiated and regulated centrally by the interactions of specialized sleep- and always correspond to the subjective experience of sleep
wake-promoting neuronal networks1,13,17,57. Notwithstanding this central regulatory quality. For instance, at times some patients report that
control, it is the position of this Review that sleep is initiated locally as a consequence
they have barely had any sleep, whereas EEG indicators
of use, and only then consolidated by central mechanisms.
suggest otherwise.

NATURE REVIEWS | NEUROSCIENCE VOLUME 9 | DECEMBER 2008 | 911


REVIEWS

Sleep depends on past activity There is a consensus that adenosine, nitric oxide
If sleep occurs locally in cortical columns, then what (NO), prostaglandin D2, tumour-necrosis factor (TNF),
determines whether a cortical column is in a sleep-like interleukin-1 (IL1) and growth-hormone-releasing hor-
or an awake-like state? Accumulating evidence suggests mone (GHRH) are involved in the regulation of NREMS
that brain areas enter a sleep-like state after a prolonged duration and intensity; each has met all or most of the
Non-rapid-eye-movement period of activity. For example, in humans, EEG delta- criteria33. These substances work in the biochemical
sleep wave power is higher in the left somatosensory cortex cascades that form the NREMS homeostat (FIG. 1). For
(NREMS). A type of sleep than in the right during the first NREMS episode after instance, IL1 and TNF induce each other’s production
characterized by
prolonged right-hand stimulation before sleep onset18. and activate nuclear factor-KB (NF-KB), which in turn
high-amplitude (up to 500 MV)
electroencephalographic slow In the past 14 years, many other studies have supported induces the production of NO and adenosine. There are
waves (delta waves). In humans these findings. In mice, rats, chickens, pigeons, cats separate substances that regulate REMS and waking,
it is often divided into four and humans, if a localized area is disproportionately although if a substance affects the duration of one state,
stages, the first two of which stimulated during waking, EEG delta-wave power is other sleep–wake states are necessarily also affected.
are characterized less by delta
waves than by a transition from
increased in the stimulated areas during subsequent REMS-promoting substances (such as vasoactive intes-
fast to slower NREMS19–25. Conversely, if afferent activity to a localized tinal polypeptide and prolactin) and wake-promoting
electroencephalographic area is reduced during waking, EEG delta-wave power substances (such as orexin, ghrelin, adrenocorticotropin
activity. In intermediate stages is reduced during NREMS26. There are also several find- hormone and corticotropin-releasing hormone) are not
of NREMS, thalamic cells
ings which show that during sleep cerebral blood flow discussed in this article because the evidence from SRS
produce 8–12 Hz bursts
known as spindles, which also is targeted to, and highest in, areas that were dispropor- studies that has bearing on sleep as a use-dependent
characterize lighter stages of tionately stimulated during prior waking 27,28. Finally, local process is limited to NREMS-promoting SRSs.
NREMS. findings from the developmental-plasticity literature29–31
indicate that changes in the sleep EEG occur in areas that SRSs act on neuronal networks. Several SRSs act on
Delta-wave power
The calculated power (in MV2)
were activated during prior waking. In summary, sleep subcortical sleep-regulatory circuits33. Adenosine acts
of delta waves, obtained from intensity, a characteristic of sleep that is determined on basal forebrain neurons to promote sleep38; GHRH,
fast fourier transformations of from EEG delta-wave power, is dependent on prior use TNF and IL1 act directly on hypothalamic preoptic
the EEG. Delta waves are from and is highest in areas that were disproportionately used neurons to promote NREMS33. In fact, inhibition of
0.5–3.5 Hz. Delta-wave power
during prior wakefulness. either GHRH or TNF by microinjection of GHRH or
is higher during sleep after
sleep deprivation and is Measurements of cortical-column activity also TNF inhibitors into the preoptic hypothalamus inhibits
considered an indicator of support the conclusion that sleep-like states depend NREMS33. TNF and IL1 also act on the locus coeruleus39,
NREMS intensity. Delta waves on prior activity of the cortical column. As described and IL1 acts on raphe serotonergic neurons to promote
are generated by the above, in rats afferent activity induced by twitching a sleep40. IL1 acts in the hypothalamus on GHRH-receptive
synchronous and cyclical
switching of cortical cells
facial whisker promotes in the relevant cortical column GABA (G-aminobutyric acid)-ergic neurons41. SRSs also
between a hyperpolarized and a sleep-like state, as determined from the amplitudes act locally in the cortex to enhance sleep phenotypes.
a depolarized state. Although of evoked-response potentials32. Moreover, if one col- If either IL1 (REF. 42), TNF43 or GHRH44 is applied to
this rhythm is present in umn is stimulated at twice the rate of another there is a the cortex unilaterally, EEG delta-wave power during
isolated cortical slices, similar
higher probability that it will enter the sleep-like state32. NREMS, but not during waking or REMS, is increased
rhythms occur in the thalamus
and might drive the cortical In summary, these data provide support for the idea ipsilaterally for several hours. If inhibitors of either IL1
cells into alternating that state oscillations in cortical columns are dependent or TNF are applied unilaterally to the cortex, the increase
hyperpolarized and on the amount of afferent activity. Sleep at this level of in EEG delta-wave power that normally occurs during
depolarized states. organization is auto-regulatory, in the sense that activity NREMS after sleep deprivation is attenuated ipsilaterally.
Rapid-eye-movement sleep
in the network determines the probability of the network Similarly, if the production of TNF is inhibited unilat-
A state characterized by a entering the sleep-like state. erally by the microinjection of a TNF small interfering
relatively fast waveform EEG RNA onto the surface of the cortex, EEG delta-wave
and a flat electromyogram in Role of sleep-regulatory substances power is inhibited ipsilaterally for days45. At the cortical-
postural muscles. This state is
We have known for almost 100 years that cerebrospinal column level of analysis, similar microinjections of TNF
also well known for the vivid
dreams that can be fluid contains substances that accumulate during wake- onto the cortex increase the probability of a cortical col-
remembered. fulness and, when transferred to the cerebrospinal fluid umn being in the sleep-like state for 1–2 hours46. These
of recipient animals, elicit sleep 33. Many substances results are consistent with the view that SRSs act both
Evoked response potentials have now been implicated in sleep regulation. However, on sleep-regulatory circuits and locally in the cortex and
The field potentials measured
from electrodes on the surface
because all physiological variables vary concurrently with brainstem to promote sleep.
of the cerebral cortex or scalp sleep, it is not possible to isolate sleep as the independent It is clear that SRSs act locally to change the electri-
that occur in response to variable. For that reason, investigators have developed cal property of neurons and thereby alter their input–
sensory afferent stimulation. lists of criteria that a substance should fulfil before it can output relationships. The direct evidence for this is
Synchronous synaptic
be termed an SRS34–37. Briefly, an SRS should enhance that the application of TNF to the surface of the cortex
activation of many cells in a
cortical column generates an sleep; inhibition of an SRS should reduce spontaneous sleep enhances the probability of cortical-column sleep 46.
electrical potential gradient and sleep rebound after sleep loss; the brain levels of an Moreover, evidence from several studies from different
that can be detected by the SRS should vary with an animal’s sleep–wake history; fields of research shows that SRSs such as cytokines and
electrodes. By using an SRS should act on sleep-regulatory circuits such as the neurotrophins alter network properties47. For example,
multiple electrodes and/or
discrete sensory stimuli, these
anterior hypothalamus; and the levels of an SRS should IL1 enhances presynaptic GABA-mediated inhibition
potentials can be localized to be altered in pathological states that are associated with of hypothalamic glutamatergic transmission48. IL1 also
individual cortical columns. enhanced sleepiness33. increases the activity of hypothalamic sleep-active neurons

912 | DECEMBER 2008 | VOLUME 9 www.nature.com/reviews/neuro


REVIEWS

Hours Seconds Milliseconds

Cell electrical and Extracellular


metabolic activity ATP Adenosine
Excitatory
neurons Neuronal-
(Glu) assembly
NOS NO
P2R sleep state
NF-B
Adenosine
Circadian
CRY1, 2 A1AR
clock COX2 PGs Inhibitory
PER1, 2 neurons
Sleep- and plasticity- (GABA)
regulatory substances:
IL1, TNF, EGF
BMAL, GHRH
CLOCK

Inhibitors:
IL4, IL10, CRH,
sTNFR, sIL1R,
IL1RA, TGF
Gene transcription and translation Blood flow Synaptic transmission

Figure 1 | Molecular networks of sleep-regulatory substances make up the non-rapid-eye-movement sleep


homeostat. The sleep-regulatory substances (SRSs) shown (yellow boxes) are produced in response to cellular activity and
metabolism. These SRSs, as well as others not illustrated here, are influenced by positive- and negative-feedback signals,
Nature Reviews | Neuroscience
including transcription-enhancer substances such as nuclear factor-KB (NF-KB)33. The fruitfly homologue of NF-KB, Relish, is
implicated in state control in those insects71. The transcription and translation events that are needed to produce the
proteins shown in the left-hand part of this figure occur over periods of hours or longer. Thus, they offer a mechanism by
which the brain can track its sleep–wake history. SRSs with direct sleep-promoting actions, such as nitric oxide (NO) and
adenosine, are labile substances with short half-lives (middle part of the figure). In the two-process model of sleep34, it is the
output of this homeostat, as estimated from electroencephalogram (EEG) delta-wave power, that is modelled as ‘process S’.
Several of the SRSs — for example, interleukin-1 (IL1), tumour-necrosis factor-A (TNFA) and adenosine — enhance EEG
delta-wave power. A variety of experimental manipulations affects the homeostat and sleep in predictable ways. For
instance, acute mild increases in ambient temperature enhance sleep duration and TNF levels98. Infection enhances sleep
duration and increases the concentration of almost every SRS in the brain99. Excessive food intake enhances brain IL1 levels
and sleep duration. Sleep loss increases levels of IL1, TNFA and adenosine33, whereas administration of antagonists of these
substances before sleep deprivation attenuates the expected rebound of non-rapid-eye-movement sleep33. We propose
that this biochemical network operates in cortical columns and other neuronal assemblies to affect state. Many of these
substances also promote whole-organism sleep if they are applied directly to sleep-regulatory networks in subcortical
Clock mechanisms of the networks33, suggesting that the homeostat operates at every level of the neural axis. Such findings also suggest that there is
suprachiasmatic nuclei a high degree of evolutionary conservation of this fundamental mechanism. A1AR, adenosine A1 receptor; COX2,
In mammals the
cyclooxygenase 2; CRH, corticotropin-releasing hormone; CRY, cryptochrome; EGF, epidermal growth factor; GABA,
suprachiasmatic nuclei of
the hypothalamus house the
G-aminobutyric acid; GHRH, growth-hormone-releasing hormone; IL1RA, interleukin-1 receptor antagonist; NOS, nitric
cellular and molecular oxide synthase; P2R, purine p2 receptor; PER, period; PGs, prostaglandins; sIL1R, soluble type-1 IL receptor; sTNFR, soluble
mechanisms that are TNF receptor; TGF, transforming growth factor. Figure modified, with permission, from REF. 60 ‘ (2007) Elsevier Science.
responsible for circadian
rhythms. If the
suprachiasmatic nuclei are and decreases that of wake-active neurons49. From the determined by Fos expression58,59. IL1 applied in this
lesioned, circadian rhythms
fever literature, we know that IL1 and TNF alter the manner also activates prefrontal cortical neurons, ventral
that are normally entrained
by light are lost. In the sensitivity of hypothalamic neurons to temperature50. lateral preoptic neurons and medial preoptic neurons58.
absence of light–dark cues, From other studies we know that IL1 receptors colocal- It seems likely that sleep-regulatory circuits integrate
normal animals exhibit ize with GHRH receptors on GABAergic hypothalamic information that is related to locally induced cortical-
rhythms that approximate neurons41. It is well known that other SRSs, such as the column states with information that is important for the
24 hours through the actions
of the free-running molecular
neurotrophins, alter the electrical properties of neu- determination of whole-animal sleep–wake states, such
oscillators in the rons51. It has also been demonstrated that SRSs such as as the time of day, sensory input, mental activity and
suprachiasmatic cells. TNF alter K+ channels in neurons52,53, glia54,55 and other information relating to emotion and disease.
cell types56. Whether such actions are related directly
Sleep-active neurons
to sleep remains unknown, but the studies cited above Neuronal activity increases SRS levels. The production
Neurons with activity that
increases during sleep or just suggest multiple routes by which SRSs can alter network of every well-characterized NREMS SRS is enhanced
before sleep onset. responsiveness in both the short and the long term. by neuronal activity. For example, if activity is experi-
NREMS is also influenced by the activity of subcor- mentally enhanced by epileptic episodes, levels of IL1
Wake-active neurons tical sleep-regulatory circuits57. These sleep-regulatory and TNF in the brain increase33. Neuronal expression
Neurons with activity that
increases during wakefulness
circuits are kept informed of cortical-column states. of TNF and IL1 is also enhanced in response to affer-
or just before wakefulness For example, application of either TNF or IL1 onto the ent activity. For instance, excessive stimulation of rat
onset. cerebral cortex activates reticular thalamic neurons, as facial whiskers for 2 hours enhances IL1 and TNF

NATURE REVIEWS | NEUROSCIENCE VOLUME 9 | DECEMBER 2008 | 913


REVIEWS

levels, thereby ultimately making the cell more sensitive


to adenosine72. NF-KB also enhances the transcription
Glial cell
of the gene that encodes GluR1, which is a subunit of
AMPARs (A-amino-3-hydroxy-5-methyl-4-isoxazole
Postsynaptic propionic acid receptors)73. The increased expression of
Synaptic neuron
terminal P2R Inactive AMPARs renders the postsynaptic neuron more sensi-
NF-B
TNFR tive to glutamate74. The increases in receptor mRNA
TNF and +
have a much longer time course than the direct actions of
IL1 release adenosine or TNF; the subsequent production of recep-
Input
IL1R Active tor protein offers a way for the brain to keep track of
ATP ATP NF-B past neuronal network activity and translate that activity
A1AR into a greater propensity for sleep or wakefulness. The
Adenosine + differences in the time courses of action of the neuro-
transmitters (milliseconds), the conversions of ATP to
AMPAR
adenosine and the actions of adenosine (seconds), and
Glutamate +
the actions of the cytokines that are released as a result of
ATP release on gene transcription and translation (min-
utes to hours) probably form a mechanism for inducing
Figure 2 | Translation of neurotransmission into actions of sleep-regulatory
the activity-dependent oscillations that are related to
substances. During neurotransmission, ATP is released fromNature
presynaptic neurons
Reviews
67
. This
| Neuroscience
extracellular ATP has fast and slow effects on postsynaptic neurons. The rapid effects neuronal-assembly sleep (FIG. 1).
occur after breakdown of ATP into adenosine, which binds to postsynaptic purine P1 Adenosine and NO are cerebral vasodilators as
receptors (the adenosine A1 receptor, A1AR). The slower effects involve ATP-induced well as SRSs. Their half-lives are short (on the order of
release of cytokines, through activation of purine P2 receptors (P2Rs) on glial cells, as has seconds) and they are probably the immediate effector
been shown in the brain68–70 and the immune system100. This provides a possible molecules for longer-lived vasodilators and SRSs, such
mechanism by which synaptic activity could be translated into signals that act as as TNF and IL1 (REFS 33,50). The dynamics of cerebral
sleep-regulatory substances, such as tumour-necrosis factor-A (TNFA) and interleukin-1B metabolism and blood flow are related to changes in the
(IL1B). The TNFA and IL1B that are released after P2R activation by ATP subsequently act concentration of multiple SRSs. As electrical activity
on postsynaptic neurons by activating nuclear factor-KB (NF-KB), which increases the
increases in a cortical column there is the potential for
transcription of genes for receptors such as A1AR and glutamate AMPARs (A-amino-
a parallel rise in metabolism and blood flow. However,
3-hydroxy-5-methyl-4-isoxazole propionic acid receptors) to change the sensitivity of
the postsynaptic neuron over longer periods (BOX 3; FIG. 3). IL1BR, IL1B receptor; TNFR, blood flow and metabolism might reach their maxima
TNF receptor. before electrical activity does75. As a result, intracellular
ATP levels might be reduced while intracellular adenosine
levels are still increasing (although there is much more
immunoreactivity in cortical layers II–IV of the columns ATP than adenosine). In parallel with the increases in
in the somatosensory cortex that receive the enhanced electrical activity, ATP in the synaptic cleft increases
afferent input 46,60. Whisker stimulation also enhances because it is released with neurotransmission (FIG. 2).
cortical expression of GHRH receptors61. Glutamate, an The released ATP causes the release of gliotransmitters,
excitatory neurotransmitter, enhances TNF production including cytokines, by binding to and activating purine
in cultured neurons62. Adenosine and NO are produced P2 receptors68–70, so that levels of extracellular cytokines
locally in response to neuronal activity; both seem to also follow the pattern of neuronal electrical activity.
be involved in determining local-sleep intensity 63. Intracellular cytokine levels in postsynaptic neurons also
Caffeine, which is an adenosine-receptor antagonist, increase with network activity 46 (FIG. 3). The enhanced
and inhibition of nitric oxide synthase, the enzyme that intracellular cytokine levels persist beyond the changes
is involved in the production of NO, reduce EEG delta- in electrical activity, blood flow and ATP and adenosine
wave power 64,65. Collectively, these data demonstrate that levels, although to date the only point at which the levels
in the brain several SRSs can operate locally and respond have been experimentally determined is 2 hours after
to cellular activity. afferent stimulation of a cortical column46. These per-
What is it about wakefulness and neuronal activ- sistent increases in intracellular cytokine levels cause
ity that causes the enhanced SRS activity? Neuronal longer-term changes in gene expression76 and subsequent
activity manifests in pre- and postsynaptic events that cell and/or network sensitivity, through changes in the
act in both the short and the long term. Neuronal activ- production of receptors that are responsive to short-lived
ity in the presynaptic neuron results in the release of molecules such as glutamate, NO and adenosine (FIG. 1).
transmitters and ATP66,67. ATP in turn does two things It is thus the changes in cytokine levels that offer a
(FIG. 2): some of it is converted to adenosine and some plausible means by which to keep track of the past wake
acts on purine P2X7 receptors on glia to release TNF (activity) levels of a cortical column and, ultimately, of
and IL1 (REFS 68–70). After conversion from ATP, adeno- the animal.
sine interacts with neurons through the adenosine A1 Cytokines are evolutionarily old, and were probably
receptor (A1AR); this action results in hyperpolarization recruited to regulate rest–activity cycles and evolutionarily
through K+ channels33,38. The TNF that is released also early versions of sleep–wake and metabolic cycles (BOX 2).
interacts with the postsynaptic and presynaptic neurons The hypothalamus and other subcortical circuits were
to activate NF-KB33,71. NF-KB increases A1AR mRNA probably undergoing activity-dependent oscillations

914 | DECEMBER 2008 | VOLUME 9 www.nature.com/reviews/neuro


REVIEWS

Waking Sleep
Input  output (environmentally relevant) Input  output (not relevant to environment)

Delayed/long-term change
Short-term in network-finding patterns
Cellular activity Changes in neural connectivity

Change in neuron sensitivity


and receptor numbers

Increases in sleep- and


Regulation of sleep
plasticity-regulatory substances

Changes in metabolism
and ATP/adenosine levels

Figure 3 | Activity-driven changes in sleep-regulatory substances regulate sleep at the local level. During
waking, environmental input to a neuronal assembly induces environmentally relevant outputs (thatReviews
Nature is, patterns of
| Neuroscience
electrical and chemical signals from the network to other brain areas), contributing to cognitive and motor responses.
Prolonged activation of a neuronal assembly induces sleep-regulatory-substance-mediated changes in the network’s
electrical and chemical outputs, which eventually results in a state shift (that is, from awake-like to sleep-like) of the local
network. Thus, during sleep, the same environmental inputs result in a different output. As output is now not directly
relevant to the environment, there is a need to prevent the animal from behaving; in other words, there is a need for
unconsciousness.

of state even before there was a neocortex. Viewed achieved? In this section we briefly discuss a model
in this way, it is easy to envision that the biochemical that helps to explain how whole-animal sleep emerges
mechanisms of SRS-regulated subcortical oscillations from the interaction of many neuronal assemblies (for
were extended to the new cortical assemblies as they instance, cortical columns); this model is described in
appeared during evolution. Further, the new cortical full in REF. 77. We propose that synchrony of cortical-
assemblies were innervated by the subcortical networks column states is a consequence of the electrical and
that were already oscillating under activity-dependent humoral interactions between columns. Naturally
auto-control. It is also possible to see how one could occurring networks that have weakly interacting compo-
extend the application of this mechanism to animals nents, such as cortical columns, have long been known
such as octopi and fruit flies, which have a very different to display emergent properties — collective behaviours
brain organization but have all the activity-dependent that are not readily apparent from the features of the
molecules in place. Indeed, the regulation of Drosophila individual components of the network — including, in
melanogaster sleep–wake cycles shares many biochemical some cases, synchronization of the network components
mechanisms with that of mammals71. and robustness to disturbance, resulting from within-
When a cortical column goes into a sleep-like state, network interactions78,79. We have developed a plausible,
the processes discussed in the previous paragraphs are albeit abstract, nonlinear mathematical model for inter-
reversed. Thus, electrical activity, blood flow, extracellu- actions among cortical columns, and have shown that
lar ATP levels and extracellular and intracellular adeno- emergent properties in this model capture well-known
sine levels all decrease while intracellular ATP levels characteristics of sleep.
increase. Some of the consequences of these changes are The local dynamics (that is, the behaviour of each
shown in FIGS 1–3. The molecules that are responsible individual neuronal assembly) of our model have some
for changes in blood flow — adenosine, NO, TNF and commonality with the dynamics of a standard model
IL1 — are also involved in cortical-column state changes; for a single neuron or neuronal assembly known as
thus, metabolism, blood flow and state are normally the Wilson–Cowan model 80, whereas the network
integrated with each other. interactions are similar in some respects to those of
other network-regulation models. In our model77, the
Modelling local–global sleep transitions state variables change over time owing to integration
Thus far we have reviewed the evidence that different of local activity, interactions with other assemblies, and
brain regions can exhibit different intensities of sleep threshold-based transitions. In addition to these inter-
simultaneously, and that individual cortical columns nal variables for each assembly, the model comprises
oscillate between sleep and waking states. We also a network that describes the strengths of interactions
described the experimental finding that during sleep or between neuronal assemblies. Neuronal assemblies are
wakefulness most cortical columns are in the functional more quickly driven towards the sleep-like state when
state that matches that of the whole animal6. How is this connected neuronal assemblies have recently entered the

NATURE REVIEWS | NEUROSCIENCE VOLUME 9 | DECEMBER 2008 | 915


REVIEWS

Box 2 | The evolution of sleep is linked to metabolism

Evolutionary Vigilance state/ Funtions of Control


complexity regulatory signal R–A cycles system level
and sleep of organization

Single cells Environmental Metabolic Subcellular


stimuli

R–A cycles

Multiple cells Environmental Metabolic Cellular


stimuli

R–A cycles Humoral signal

Multiple cells Environmental Metabolic Multicellular


(nerve nets) stimuli

R–A cycles Humoral signal

Electrical
signal

Complex Environmental Metabolic, Neuronal


ganglia stimuli synaptic network network
stabilization
R–A cycles Sleep

Electrical Humoral

Homeotherms Environmental Metabolic, Neuronal


stimuli synaptic network network
stabilization

R–A cycles Sleep

Electrical Humoral

Body temperature

There is no evidence that sleep serves distinctly different functions in different species. Indeed, studies in fruit flies have
shown that at a biochemical level the substances that regulate fruit fly sleep are the same as those that Reviews
Nature regulate| Neuroscience
mammalian sleep — for example, adenosine, nuclear factor-KB and epidermal growth factor. The most parsimonious
course, at least for now, is to consider sleep as a universal process.
How might sleep have evolved? Temperature and light rhythms associated with the Earth’s rotation probably induced
early cells to time the production of metabolic enzymes to coincide with the period in which nutrients were most available
and most easily processed (see figure). This adaptation eventually evolved into a capacity to anticipate the earth’s orbital
mechanics with the sleep–wake cycle in animals with a brain.
The development from responding to the light–dark cycle to anticipating it would have required chemical signalling in
single cells that, later in evolution, were probably used to inform other cells of their metabolic states. As multicellular
organisms evolved, humoral signalling became more complex and was used to coordinate whole-animal physiology. The
humoral signals themselves began to be influenced by rest–activity cycles (R–A cycles) and environmental cues. Electrical
signalling resulting from humoral signals occurred in single cells, although its use in higher-order information processing
only truly came of age with the development of ganglia and nervous systems. As biochemical and neural complexity
evolved to include neuronal specialization and epigenetic plasticity, the levels of organization at which the control
systems operated also expanded. The flexible connectivity between neurons is experience-dependent4,81,88 and might
therefore have required a mechanism — that is, sleep — by which to ensure the stability of synaptic networks that encode
instinctual and learned memories4,81,89. It seems probable that sleep developed from the ‘rest’ portion of the behavioural
R–A cycle, as niche-appropriate inactivity already existed during rest. The regulatory mechanisms for sleep also probably
developed, in part, from those that already regulated rest.
Over the past few years ample evidence has been reported that supports the notion that the genes that are involved in
generating the circadian rhythm directly affect sleep17. Sleep itself directly adds to the regulatory complexity by affecting
R–A cycles, humoral signals, cellular electrical potentials, responses to environmental stimuli and every physiological
system, including metabolic regulation90. Figure modified, with permission, from REF. 91 ‘ (1995) Elsevier/North-Holland
Biomedical Press.

916 | DECEMBER 2008 | VOLUME 9 www.nature.com/reviews/neuro


REVIEWS

Hebbian plasticity sleep-like state. The strength of this interactive response restores brain function. However, at more mechanistic
The type of rapid (on the scale correlates with the strengths of the connections between levels no one has yet identified what is being restored
of minutes to hours) changes in the neuronal assemblies. Finally, if the circadian clock or whether any restoring takes place at the cellular and
neuronal connectivity that are indicates that the animal should be asleep, the activity molecular levels. This evidential deficit has led many
associated with use of the
network of which the neurons
variable effectively increases more quickly and the state theorists to posit that optimizing neuronal connectivity
are a part. The term is derived of the neuronal assembly moves more rapidly towards is the primordial function of sleep4,5,81–84.
from the work of Donald Hebb, sleep. The functional importance of sleep is illustrated by
a Canadian psychologist who With this model, in silico experiments and math- the fact that during sleep one gives up opportunities to
first posited, in 1949, that
ematical analysis demonstrate well-known characteris- reproduce, eat, drink or socialize and is subject to preda-
neurons that fire together tend
to wire together.
tics of sleep as emergent properties of interactions in the tion. Sleep could only have evolved despite these high
network; these include stability of the sleep–wake cycle evolutionary costs if it serves a crucial, primordial func-
under nominal conditions, the emergence of a synchro- tion. Maintaining adaptable flexible neural connectivity
nous sleep state, and the regional-activity dependence may be sufficiently important for the brain to allow the
of sleep. In silico results and analysis also indicate that persistence of such a periodic, disadvantaged state4,5. The
over-stimulated neuronal assemblies will enter the sleep- central idea in connectivity theories of sleep is the recog-
like state rapidly (the impact on a rat’s sleep response nition that use-dependent changes in synaptic efficacy
of repeatedly stimulating a single whisker 6 (described and connectivity would lead to dysfunction unless there
earlier in the Review) serves as an example of this) and were processes to stabilize, and thus preserve, function-
then (owing to the coupling between assemblies) induce ally optimized synaptic networks (which are prima facie
nearby neuronal assemblies to also enter the sleep-like adaptive because the organism is alive)4,81,84. One of the
state, eventually leading to whole-animal sleep. We have processes that has been posited to be responsible for
verified that the model does indeed capture the evolution this stabilization has been called ‘synaptic scaling’ in the
of a global sleep state, as we theorized above. As a further more recent literature85 (BOX 3). Synaptic scaling is a use-
step, we note that the model permits us to tie emergent dependent, slow homeostatic process that occurs over
characteristics (for instance, the amount of asynchro- hours to days74,86. A proposed mechanism for synaptic
nicity between columns during over-stimulation) to network stabilization4 involves SRS-induced changes in
network properties. local electrical properties, as described above. Indeed,
there is now direct evidence that TNF is involved in
Theoretical perspectives on sleep function synaptic scaling 74,86 (BOX 3). Kavanau81 and Tononi and
Sleep, as occurs in mammals, probably has multiple Cirelli83 proposed that intrinsic spontaneous electrical
functions, although when it first evolved it probably did activity serves this scaling function. The various pro-
so for one crucial, primordial function. At the whole- posed scaling mechanisms — for example, SRS-induced
animal behavioural level, the functions of sleep seem changes in receptor populations, transmitter release,
clear: energy is saved, performance is restored and (in ion-channel populations and so on — are not mutually
humans) affect becomes more positive. Such findings exclusive.
have led to the universal acknowledgement that sleep If we consider that sensory and internal afferent input
during waking induce an environmentally relevant neu-
ronal-network output (that is, patterns of electrical and
Box 3 | Sleep-regulatory substances are involved in synaptic scaling chemical signals from the network that project to other
brain areas and contribute to whole-animal cognitive
Sleep-regulatory substances (SRSs) seem to be involved in synaptic scaling (the up- or
and motor responses), then after prolonged activation of
downregulation of the strength of a neuron’s synapses). Synaptic scaling is a slow
homeostatic process that occurs over hours to days; it serves in part to balance, or neurons in a neuronal assembly the resulting SRS release
scale, Hebbian plasticity changes that occur rapidly as a consequence of neuronal would induce changes in the network’s electrical and
activity. Hebbian changes in both synaptic weight (the strength of the connection chemical outputs and responsiveness to inputs (FIG. 2).
between two neurons) and synaptic scaling can increase or decrease synaptic efficacy; These changes would presumably summate, perhaps
the direction depends on the nature of the prior activity. with positive feedback, and result in a state shift in the
The SRS tumour-necrosis factor-A (TNFA) promotes AMPAR (A-amino-3-hydroxy- local network. Sleep is thus initiated in neuronal assem-
5-methyl-4-isoxazole propionic acid receptor) expression and enhances cytosolic Ca2+ blies dependent on prior use. The new output, because it
levels73,92. An inhibitor of TNF inhibits AMPA-induced postsynaptic potentials93 and is qualitatively and quantitatively different from the orig-
AMPA-induced changes in cytosolic Ca2+ (REF. 91), suggesting that this action is
inal output, might not be relevant to the environmentally
physiological. If a small interfering RNA directed against the mRNA for TNF is applied
driven input. Thus, if the initial environmentally driven
to the cortex, levels of GluR1 mRNA are reduced45; GluR1 is a subunit of AMPARs, and
AMPARs influence electroencephalogram delta-wave synchronization94 and synaptic inputs induce adaptive outputs (waking), then a shift
plasticity95, and thus it seems that TNF has a role in the regulation of these processes. in input–output relationships could result in outputs (in
There is also direct evidence for the involvement of TNF in synaptic up-scaling in this case sleep) that lack the necessary network-activity
cortical layers II–IV74,86. Interleukin-1, another SRS, might also affect AMPAR expression96. patterns to induce environmentally relevant cognitive or
In cortical layer V, AMPARs are involved in down-scaling during non-rapid-eye- motor outputs (FIG. 3). This creates an adaptive need to
movement sleep (NREMS)97. The direction of scaling during sleep is probably specific to prevent the animal from behaving, and it is probably a
local brain areas and dependent on the nature of prior activity. Regardless, such data role of the traditional sleep–wake regulatory circuits to
suggest that an SRS-dependent mechanism reconfigures synaptic weights during ensure the absence of behaviour at such times. Thus, not
NREMS. This may represent the first experimentally supported reductionist function
only are the mechanisms for local sleep inseparable from
of sleep30.
the connectivity and metabolic functions of sleep, they

NATURE REVIEWS | NEUROSCIENCE VOLUME 9 | DECEMBER 2008 | 917


REVIEWS

also cause the network outputs that lead to the neces- explanations for these phenomena. For example, sleep
sity for the altered consciousness that typically pervades inertia may be a manifestation of some neuronal assem-
sleep. Previously, it was proposed5,81 that such outputs blies remaining in the sleep-like state after sufficient
during sleep serve to stabilize the networks and thereby numbers of assemblies have transitioned to the wake-
preserve them. like state to cause behavioural awakening. By extension,
the degree of sleepiness, or the speed and accuracy of
Conclusions and future directions performance, may depend on the fraction of neuronal
Our mechanistic proposal for the generation of sleep at assemblies that are in the sleep-like state rather than
the local level involves afferent-input-induced release the awake-like state. There is brain-imaging evidence
of SRSs and their actions on neuronal assemblies to which suggests that in insomnia patients some areas of
change local-network input–output relationships. This the brain show activation that is characteristic of wak-
mechanism is probably a property of neuronal assem- ing while simultaneously other areas show the reduced
blies throughout the brain, including the evolutionar- activity that is characteristic of sleep87.
ily old midbrain and lower brainstem. State changes The theory presented here that sleep is an emergent
of neuronal assemblies at the lower brain levels would property of cortical columns cannot yet address the
manifest themselves differently from those at higher question of how many assemblies need to be in the sleep-
levels, because the various levels of the brain have like state before consciousness changes. Of note, there
different functions. is a similar issue in the more traditional paradigm of
The viewpoint presented in this Review allows one to sleep regulation, which proposes a top-down imposition
frame important observations in sleep research in a dif- of sleep on the brain by regulatory circuits: it does not
ferent context from that which the currently dominant specify the areas (and how many of them) that need to
paradigm of sleep regulation allows. Currently, sleep be acted on by the regulatory circuits to produce sleep.
is typically viewed as being imposed on the brain by It will remain difficult to experimentally investigate the
sleep-regulatory circuits (BOX 1). However, that paradigm ratio of awake-like to sleep-like assemblies that defines
does not address many well-known phenomena, such global sleep, as one would have to record from thousands
as sleep inertia, restoration of peak performance during of individual assemblies. However, we anticipate that our
sleep, recovery and reorganization of sleep after brain mathematical model will, with future developments, give
lesions, the nature of sleep homeostatic mechanisms, some indication of system behaviour — for example, the
clinical conditions such as insomnia, and sleepiness fraction of assemblies that is needed for a rapid phase
and fatigue associated with multiple bodily ailments, transition. Thus, although we are far from any compre-
such as rheumatoid arthritis and most autoimmune hensive molecular or genetic understanding of sleep
disorders, and with various chemotherapies. With our or the mechanistic details of local control of sleep, the
newer view, which posits that parts of the brain can be present view provides a new evolutionary conceptual
asleep while other parts are awake, it is easier to envision structure for continued discoveries in the field.

1. Koella, W. P. The organization and regulation of sleep. 9. Rattenborg, N. C., Amlaner, C. J. & Lima, S. L. 19. Miyamoto, H., Katagiri, H. & Hensch, T. Experience-
A review of the experimental evidence and a novel Unilateral eye closure and interhemispheric EEG dependent slow-wave sleep development. Nature
integrated model of the organizing and regulating asymmetry during sleep in the pigeon Neurosci. 6, 553–554 (2003).
apparatus. Experientia 40, 309–338 (1984). (Columba livia). Brain Behav. Evol. 58, 323–332 20. Iwasaki, N., Karashima, A., Tamakawa, Y., Katayama, N.
2. Panksepp, J., Jalowiec, J. E., Zolovick, A. J., Stern, (2001). & Nakao, M. Sleep EEG dynamics in rat barrel cortex
W. C. & Morgane, P. J. Inhibition of glycolytic 10. Werth, E., Achermann, P., Dijk, D. J. & Borbely, A. A. associated with sensory deprivation. Neuroreport 15,
metabolism and sleep-waking states in cats. Pharm. Spindle frequency activity in the sleep EEG: individual 2681–2684 (2004).
Biochem. Behav. 1, 41–46 (1973). differences and topographic distribution. 21. Vyazovskiy, V., Borbely, A. A. & Tobler, I. Unilateral
3. Jouvet, M. Biogenic amines and the states of sleep. Electroencephalogr. Clin. Neurophysiol. 103, vibrissae stimulation during waking induces
Science 163, 32–41 (1969). 535–542 (1997). interhemispheric EEG asymmetry during
4. Krueger, J. M. & Obal, F. Jr. A neuronal group theory 11. Braun, A. R. et al. Regional cerebral blood flow subsequent sleep in the rat. J. Sleep Res. 9, 367–371
of sleep function. J. Sleep Res. 2, 63–69 (1993). throughout the sleep-wake cycle. An H215O PET study. (2000).
This was the first statement of the hypothesis that Brain 120, 1173–1197 (1997). 22. Cottone, L. A., Adamo, D. & Squires, N. K. The effect
sleep is a local use-dependent process. 12. Kristiansen, K. & Courtois, G. Rhythmic activity from of unilateral somatosensory stimulation on
5. Krueger, J. M. & Obal, F. Jr. Sleep function. Front. isolated cerebral cortex EEG. Clin. Neurophysiol. 1, hemispheric asymmetries during slow wave sleep.
Biosci. 8, 511–519 (2003). 265–272 (1949). Sleep 27, 63–68 (2004).
6. Rector, D. M., Topchiy, I. A., Carter, K. M. & Rojas, 13. Steriade, M. The corticothalamic system in sleep. 23. Ferrara, M., De Gennaro, L., Curcio, G., Cristiani, R. &
M. J. Local functional state differences between rat Front. Biosci. 8, d878–d899 (2003). Bertini, M. Interhemispheric asymmetry of human
cortical columns. Brain Res. 1047, 45–55 (2005). 14. Pigarev, I. N., Nothdurf, H. C. & Kastner, S. Evidence sleep EEG in response to selective slow-wave sleep
This was the first demonstration that cortical columns for asynchronous development of sleep in cortical deprivation. Behav. Neurosci. 116, 976–981
oscillate between functional states, one of which has areas. Neuroreport 8, 2557–2560 (1997). (2002).
properties similar to whole-organism sleep. 15. Koch, C. The Quest for Consciousness (Roberts and 24. Huber, R., Ghilardi, M. F., Massimini, M. & Tononi, G.
7. Mahowald, M. W. & Schenck, C. H. Insights from Company, Englewood, Colarado, 2004). Local sleep and learning. Nature 430, 78–81
studying human sleep disorders. Nature 437, 16. Walker, A. J., Topchiy, I., Kouptsov, K. & Rector, D. M. (2004).
1279–1285 (2005). ERP differences during conditioned lick response in 25. Yasuda, T., Yasuda, K., Brown, R. A. & Krueger, J. M.
These authors have recognized for over 20 years the rat. Sleep 28, A15 (2005). State-dependent effects of light-dark cycle on
that human parasomnias suggest that the brain 17. Moore, R. Y. Suprachiasmatic nucleus in sleep-wake somatosensory and visual cortex EEG in rats. Am.
can be awake and asleep simultaneously. This is a regulation. Sleep Med. 8 (Suppl. 3), 27–33 (2007). J. Physiol. 289, R1083–R1089 (2005).
comprehensive review of that work. 18. Kattler, H., Dijk, D. J. & Borbely, A. A. Effect of 26. Huber, R. et al. Arm immobilization causes cortical
8. Mukhametov, L. M., Supin, A. Y. & Polyakova, I. G. unilateral somatosensory stimulation prior to sleep on plastic changes and locally decreases sleep slow wave
Interhemispheric asymmetry of the the sleep EEG in humans. J. Sleep Res. 3, 1599–1604 activity. Nature Neurosci. 9, 1169–1176 (2006).
electroencephalographic sleep patterns in dolphins. (1994). 27. Maquet, P. The role of sleep in learning and memory.
Brain Res. 134, 581–584 (1977). The authors tested the hypothesis put forward in Science 294, 1048–1052 (2001).
This pioneering study demonstrated that dolphins reference 4 that sleep is a local activity-dependent 28. Drummond, S. P. A. et al. Sleep deprivation-induced
have high-amplitude delta-wave sleep in only one process. Much research has since extended and reduction in cortical functional response to serial
half of the brain at a time, and that they lack REMS. confirmed this pioneering study. subtraction. Neuroreport 10, 3745–3748 (1999).

918 | DECEMBER 2008 | VOLUME 9 www.nature.com/reviews/neuro


REVIEWS

29. Mascetti, G. G., Rugger, M., Vallortigara, G. & 55. McLarnon, J. G. et al. Acute actions of tumor necrosis 80. Wilson, H. R. & Cowan, J. D. Excitatory and inhibitory
Boddo, D. Monocular-unihemispheric sleep and visual factor-A on intracellular Ca2+ and K+ currents in human interactions in localized populations of model neurons.
discrimination learning in the domestic chick. Exp. microglia. Neuroscience 104, 1175–1184 (2001). Biophys. J. 12, 1–24 (1974).
Brain Res. 176, 70–84 (2007). 56. Kawada, H. et al. Tumor necrosis factor-A 81. Kavanau, J. L. Sleep and dynamic stabilization of
30. Frank, M. G., Issa, N. P. & Stryker, M. P. Sleep downregulates the voltage gated outward K+ current neural circuitry: a review and synthesis. Behav. Brain
enhances plasticity in the developing visual cortex. in cultured neonatal rat cardiomyocytes: a possible Res. 63, 111–126 (1994).
Neuron 30, 275–287 (2001). cause of electrical remodeling in diseased hearts. This is a scholarly development of the idea that
31. Marks, G. A., Shaffery, J. P., Oksenberg, A., Speciale, Circ. J. 70, 605–609 (2006). experience-driven changes in neural connectivity
S. G. & Roffwarg, H. P. A functional role for REM sleep 57. Saper, C. B., Scammell, T. E. & Lu, J. Hypothalamic lead to dysfunction unless there is a mechanism in
in brain maturation. Behav. Brain Res. 69, 1–11 regulation of sleep and circadian rhythms. Nature place to temper such activity. The author posits
(1995). 437, 1257–1263 (2005). that the intrinsic electrical activity of the brain
32. Rector, D. M., Topchiy, I. & Rojas, M. Local cortical 58. Yasuda, K. et al. Unilateral cortical application of stabilizes the activity-altered circuits and that this
column activity states and localized delta wave interleukin-1B (IL1B) induces asymmetry in Fos- and is a function of sleep.
differences. Sleep 28, A26 (2005). IL1B-immunoreactivity: implication for sleep 82. Siegel, J. M. Clues to the functions of mammalian
33. Obal, F. Jr & Krueger, J. M. Biochemical regulation of regulation. Brain Res. 1131, 44–59 (2007). sleep. Nature 437, 1264–1271 (2005).
sleep. Front. Biosci. 8, 520–550 (2003). 59. Churchill, L. et al. Unilateral cortical application of 83. Tononi, G. & Cirelli, C. Sleep function and synaptic
34. Borbély, A. A. & Tobler, I. Endogenous sleep- tumor necrosis factor A induces asymmetry in Fos- and homeostasis. Sleep Med. Rev. 10, 49–62 (2006).
promoting substances and sleep regulation. Physiol. interleukin-1B-immunoreactive cells within the 84. Crick, F. & Mitchinson, G. The function of dream sleep.
Rev. 69, 605–670 (1989). corticothalamic projection. Brain Res. 1055, 15–24 Nature 304, 111–114 (1983).
35. Jouvet, M. Neuromediateurs et facteurs hypnogenes. (2005). 85. Abbott, L. F. & Nelson, S. B. Synaptic plasticity: taming
Rev. Neurol. (Paris) 140, 389–400 (1984). 60. Krueger, J. M., Rector, D. M. & Churchill, L. Sleep the beast. Nature Neurosci. 3, 1178–1183 (2000).
36. Krueger, J. M. & Obál, F. Jr. in Sleep and Breathing and cytokines. Sleep Med. Clin. 2, 161–170 (2007). 86. Stellwagen, D. & Malenka, R. C. Synaptic scaling
(eds Saunders, N. A. & Sullivan, C. E.) 79–112 61. De, A., Fix, C., Hall, S., Churchill, L. & Krueger, J. M. mediated by glial TNF-A. Nature 440, 1054–1059
(Dekker, Inc., New York, 1994). Growth hormone releasing hormone receptive immuno- (2006).
37. Inoué, S. Biology of Sleep Substances. (CRC, Boca reactive cells increase in the barrel field in response to The authors show that TNF, an SRS, is involved in
Raton, 1989). whisker deflection in rats. Sleep 29, A9 (2006). synaptic scaling. The implications for sleep are that
38. Basheer, R., Strecker, R. E., Thakkar, M. M. & 62. De, A., Krueger, J. M. & Simasko, S. M. Glutamate sleep-regulatory mechanisms cannot be separated
McCarley, R. W. Adenosine and sleep-wake regulation. induces expression and release of tumor necrosis from a sleep-connectivity stabilization function.
Prog. Neurobiol. 73, 379–396 (2004). factor-A in cultured hypothalamic cells. Brain Res. 87. Nofzinger, E. A. et al. Regional cerebral metabolic
39. De Sarro, G., Gareri, P., Sinopoli, V. A., David, E. & 1053, 54–61 (2005). correlates of WASO during NREMS sleep in insomnia.
Rotiroti, D. Comparative, behavioural and 63. Gerashchenko, D. et al. Identification of a population J. Clin. Sleep Med. 2, 316–322 (2006).
electrocortical effects of tumor necrosis factor-A and of sleep-active cerebral cortex neurons. Proc. Natl 88. Edelman, G. H. Neural Darwinsim (Basic Books, New
interleukin-1 microinjected into the locus coeruleus of Acad. Sci. USA 105, 10227–10232 (2008). York, 1987).
rat. Life Sci. 60, 555–564 (1997). 64. Kapas, L., Fang, J. & Krueger, J. M. Inhibition of nitric 89. Panksepp, J. Affective Neuroscience (Oxford Univ.
40. Manfridi, A. et al. Interleukin-1B enhances non-rapid oxide synthesis inhibits rat sleep. Brain Res. 664, Press, New York, 1998).
eye movement sleep when microinjected into the 189–196 (1994). 90. Brandt, J. A. et al. Sleep-deprivation but not a whisker
dorsal raphe nucleus and inhibits serotonergic 65. Landolt, H. P., Dijk, D. J., Gaus, S. E. & Borbely, A. A. trim increases nerve growth factor within barrel
neurons in vitro. Eur. J. Neurosci. 18, 1041–1049 Caffeine reduces low-frequency delta activity in the cortical neurons. Brain Res. 898, 105–112 (2001).
(2003). human sleep EEG. Neuropsychopharmacology 12, This report demonstrates that the interaction of
41. De, A., Churchill, L., Obal, F. Jr, Simasko, S. & Krueger, 229–238 (1995). afferent activity and sleep alters the expression of
J. M. GHRH and IL1B increase cytoplasmic Ca2+ levels 66. Farber, K. & Kettenmann, H. Purinergic signaling and an SRS in neurons. As such it provides evidence for a
in cultured hypothalamic GABAergic neurons. Brain microglia. Pflugers Arch. 452, 615–621 (2006). mechanism by which local sleep is tied to prior activity.
Res. 949, 209–212 (2002). 67. Burnstock, G. Physiology and pathology of purinergic 91. Krueger, J. M. Obal, F. Jr, Kapas, L. & Fang, J. Brain
42. Yasuda, T., Yoshida, H., Garcia-Garcia, F., Kay, D. & neurotransmission. Physiol. Rev. 87, 659–797 organization and sleep function. Behav. Brain Res. 69,
Krueger, J. M. Interleukin-1B has a role in cerebral (2007). 177 185 (1995).
cortical state dependent electroencephalographic 68. Hide, I. et al. Extracellular ATP triggers tumor necrosis 92. De, A., Krueger, J. M. & Simasko, S. M. Tumor
slow-wave activity. Sleep 28, 177–184 (2005). factor-A release from rat microglia. J. Neurochem. 75, necrosis factor A increases cytosolic calcium response
43. Yoshida, H. et al. State-specific asymmetries in EEG 965–972 (2000). AMPA and KCl in primary cultures of rat hippocampal
slow wave activity induced by local application of 69. Suzuki, T. et al. Production and release of neurons. Brain Res. 981, 133–142 (2003).
TNFA. Brain Res. 1009, 129–136 (2004). neuroprotective tumor necrosis factor by P2X7 93. Beattie, E. C. et al. Control of synaptic strength by glial
44. Szentirmai, E. et al. Growth hormone releasing receptor-activated microglia. J. Neurosci. 24, 1–7 TNFA. Science 295, 2282–2285 (2002).
hormone: cerebral cortical EEG actions and (2004). 94. Bazhenov, M., Timofeev, I., Steriade, M. & Sejnowski,
expression. Am. J. Physiol. 293, R922–R930 (2007). 70. Bianco, F. et al. Astrocyte-derived ATP induces vesicle T. J. Model of thalamocortical slow-wave sleep
45. Taishi, P. et al. TNFA siRNA reduces brain TNF and shedding and IL-1B release from microglia. oscillations and transitions to activated states.
EEG delta wave activity in rats. Brain Res. 1156, 125– J. Immunol. 174, 7268–7277 (2005). J. Neurosci. 22, 8691–8704 (2002).
132 (2007). 71. Williams, J. A., Sathyanarayanan, S., Hendricks, J. C. 95. Malinow, R. & Malenka, R. C. AMPA receptor
46. Churchill, L. et al. Tumor necrosis factor A: activity & Sehgal, A. Interaction between sleep and the trafficking and synaptic plasticity. Ann. Rev. Neurosci.
dependent expression and promotion of cortical immune response in Drosophila: a role for the NFKB 25, 103–126 (2002).
column sleep in rats. Neuroscience 156, 71–80 Relish. Sleep 30, 389–400 (2007). 96. Lai, A. Y., Swayze, R. D., El-Husseini, A. & Song, C.
(2008). 72. Jajoo, S., Mukherjea, D., Pingle, S., Sekino, Y. & Interleukin-1B modulates AMPA receptor expression
47. Obal, F. Jr & Krueger, J. M. in The Physiological Ramkumar, V. Induction of adenosine A1 receptor and phosphorylation in hippocampal neurons.
Nature of Sleep (eds Parmeggiani, P. L. & Velluti, R.) expression by pertussis toxin via an adenosine J. Neuroimmunol. 175, 97–106 (2006).
23–44 (Imperial College Press, 2004). 5’-diphosphate ribosylation-independent pathway. 97. Czarnecki, A., Birtoli, B. & Ulrich, D. Cellular
48. Tabarean, I. V., Korn, H. & Bartfai, T. Interleukin-1B J. Pharmacol. Exp. Ther. 317, 1–10 (2006). mechanisms of burst-firing mediated long-term
induces hyperpolarization and modulates synaptic 73. Yu, Z. et al. Tumor necrosis factor A increases neuronal depression in rat neocortical pyramidal cells.
inhibition in preoptic and anterior hypothalamic vulnerability to excitotoxic necrosis by inducing J. Physiol. 578, 471–479 (2007).
neurons. Neuroscience 4, 1685–1695 (2006). expression of the AMPA–glutamate receptor subunit 98. Takahashi, S. & Krueger, J. M. Inhibition of tumor
49. Alam, M. N. et al. Interleukin-1B modulates state- GluR1 via an acid sphingomyelinase- and necrosis factor prevents warming-induced sleep
dependent discharge activity of preoptic area and NF-KB-dependent mechanism. Neurobiol. Dis. 11, responses in rabbits. Am. J. Physiol. 272,
basal forebrain neurons: role in sleep regulation. Eur. 199–213 (2002). R1325–R1329 (1997).
J. Neurosci. 20, 207–216 (2004). 74. Kaneko, M., Stellwagen, D., Malenka, R. C. & Stryker, 99. Majde, J. A. & Krueger, J. M. Links between the innate
50. Shibata, M. Hypothalamic neuronal responses to M. P. Tumor necrosis factor-A mediates one immune system and sleep. J. Allergy Clin. Immunol.
cytokines. Yale J. Biol. Med. 63, 147–156 (1990). component of competitive, experience-dependent 116, 1188–1198 (2005).
51. Schinder, A. F. & Poo, M.-M. The neurotrophin plasticity in developing visual cortex. Neuron 58, 100. Ferrari, D. et al. The P2X7 receptor: a key player in
hypothesis for synaptic plasticity. Trends Neurosci. 23, 673–680 (2008). IL-1 processing and release. J. Immunol. 176,
639–645 (2000). 75. Schei, J. L., Foust, A. J., Rojas, M. J., Navas, J. A. & 3877–3883 (2006).
52. Houzen, H., Kikuchi, S., Kanno, M., Shinpo, K. & Rector, D. M. Evoked optical responses differ between
Tashiro, K. Tumor necrosis factor enhancement of quiet sleep stages. Sleep 31, A7 (2008). Acknowledgements
transient outward potassium currents in cultured rat 76. Taishi, P., Churchill, L., De, A., Obal, F. & Krueger, This work was supported by the W.M. Keck Foundation, the
cortical neurons. J. Neurosci. Res. 50, 990–999 J. M. Cytokine mRNA induction by interleukin-1B or US National Institutes of Health (grant numbers NS 25378,
(1997). tumor necrosis factor A in vitro and in vivo. Brain Res. NS 31453 and MH 71830), the US Army Research
53. Diem, R., Meyer, R., Weishaupt, J. H. & Bahr, M. 1226, 89–98 (2008). Development and Material Command, the National Science
Reduction of potassium currents in 77. Roy, S., Krueger, J. M., Rector, D. M. & Wan, Y. Foundation, and the National Aeronautics and Space
phosphatidylinositol 3-kinase-dependent AKT Network models for activity-dependent sleep Administration.
phosphorylation by tumor necrosis factor-A rescues regulation. J. Theor. Biol. 253, 462–468 (2008).
axotomized retinal ganglion cells from retrograde cell 78. Mirollo, R. E. & Strogatz, S. H. Synchronization of
death in vivo. J. Neurosci. 21, 2058–2066 (2001). pulse-coupled biological oscillators. SIAM J. Appl. FURTHER INFORMATION
54. Koller, H., Allert, N., Oel, D., Stoll, G. & Siebler, M. Math. 50, 1645–1662 (1990). James Krueger’s homepage: http://www.vetmed.wsu.edu/
TNF alpha induces a protein kinase C-dependent 79. Asavathiratham, C., Roy S., Lesieutre, B. C. & research_vcapp/krueger/krueger.asp
reduction in astroglial K+ conductance. Neuroreport 9, Verghese, G. C. The influence model. IEEE Control Syst. ALL LINKS ARE ACTIVE IN THE ONLINE PDF
1375–1378 (1998). Mag. NY 21, 52–64 (2001).

NATURE REVIEWS | NEUROSCIENCE VOLUME 9 | DECEMBER 2008 | 919

Anda mungkin juga menyukai