ArticlePDF AvailableLiterature Review

Abstract and Figures

Modern societies are experiencing an increasing trend of reduced sleep duration, with nocturnal sleeping time below the recommended ranges for health. Epidemiological and laboratory studies have demonstrated detrimental effects of sleep deprivation on health. Sleep exerts an immune-supportive function, promoting host defense against infection and inflammatory insults. Sleep deprivation has been associated with alterations of innate and adaptive immune parameters, leading to a chronic inflammatory state and an increased risk for infectious/inflammatory pathologies, including cardiometabolic, neoplastic, autoimmune and neurodegenerative diseases. Here, we review recent advancements on the immune responses to sleep deprivation as evidenced by experimental and epidemiological studies, the pathophysiology, and the role for the sleep deprivation-induced immune changes in increasing the risk for chronic diseases. Gaps in knowledge and methodological pitfalls still remain. Further understanding of the causal relationship between sleep deprivation and immune deregulation would help to identify individuals at risk for disease and to prevent adverse health outcomes. Garbarino et al review recent experimental and epidemiological developments regarding immune responses to sleep deprivation and consider the role for the sleep deprivation induced immune changes in increasing the risk for chronic diseases.
Content may be subject to copyright.
REVIEW ARTICLE
Role of sleep deprivation in immune-related
disease risk and outcomes
Sergio Garbarino 1, Paola Lanteri2, Nicola Luigi Bragazzi3,
Nicola Magnavita4,5 & Egeria Scoditti6
Modern societies are experiencing an increasing trend of reduced sleep duration, with noc-
turnal sleeping time below the recommended ranges for health. Epidemiological and
laboratory studies have demonstrated detrimental effects of sleep deprivation on health.
Sleep exerts an immune-supportive function, promoting host defense against infection and
inammatory insults. Sleep deprivation has been associated with alterations of innate and
adaptive immune parameters, leading to a chronic inammatory state and an increased risk
for infectious/inammatory pathologies, including cardiometabolic, neoplastic, autoimmune
and neurodegenerative diseases. Here, we review recent advancements on the immune
responses to sleep deprivation as evidenced by experimental and epidemiological studies, the
pathophysiology, and the role for the sleep deprivation-induced immune changes in
increasing the risk for chronic diseases. Gaps in knowledge and methodological pitfalls still
remain. Further understanding of the causal relationship between sleep deprivation and
immune deregulation would help to identify individuals at risk for disease and to prevent
adverse health outcomes.
Sleep is an active physiological process necessary for life and normally occupying one-third
of our lives, playing a fundamental role for physical, mental, and emotional health1. Sleep
patterns and need are inuenced by a complex interplay between chronological age,
maturation stage, genetic, behavioral, environmental, and social factors26. Adults should sleep a
minimum of 7 h per night to promote optimal health7,8.
Besides medical problems including obstructive sleep apnea and insomnia, factors associated
mostly with the modern 24/7 society, such as work and social demands, smartphone addiction,
and poor diet911, contribute to cause the current phenomenon of chronic sleep deprivation, i.e.,
sleeping less than the recommended amount or, better to say, the intrinsic sleep need12.
Sleep deprivation may be categorized as acute or chronic. Acute sleep deprivation refers to no
sleep or reduction in the usual total sleep time, usually lasting 12 days, with waking time
extending beyond the typical 1618 h. Chronic sleep deprivation is dened by the Third Edition
of the International Classication of Sleep Disorders as a disorder characterized by excessive
daytime sleepiness caused by routine sleeping less than the amount required for optimal func-
tioning and health maintenance, almost every day for at least 3 months13.
Population studies reported a stably increasing prevalence of adults sleeping less than 6 h per
night over a long period12,14,15, also affecting children and adolescents16,17. Sleep duration
https://doi.org/10.1038/s42003-021-02825-4 OPEN
1Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal/Child Sciences, University of Genoa, 16132 Genoa, Italy.
2Neurophysiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy. 3Laboratory for Industrial and Applied Mathematics (LIAM),
Department of Mathematics and Statistics, York University, Toronto, ON M3J 1P3, Canada. 4Postgraduate School of Occupational Medicine, Università
Cattolica del Sacro Cuore, 00168 Rome, Italy. 5Department of Woman/Child and Public Health, Fondazione Policlinico Universitario Agostino Gemelli
IRCCS, 00168 Rome, Italy. 6National Research Council (CNR), Institute of Clinical Physiology (IFC), 73100 Lecce, Italy.
email: sgarbarino.neuro@gmail.com
COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www .nature.com/commsbio 1
1234567890():,;
decline is present not only in high-income and developed
countries18 but also in low-income or racial/ethnic minorities19,
thus representing a worldwide problem.
In addition to fatigue, excessive daytime sleepiness, and
impaired cognitive and safety-related performance, sleep depri-
vation is associated with an increased risk of adverse health
outcomes and all-cause mortality2024. Indeed, epidemiological
and experimental data support the association of sleep depriva-
tion with the risk of cardiovascular (CV) (hypertension and
coronary artery disease) and metabolic (obesity, type 2 diabetes
(T2DM)) diseases2427. In the United States, sleep deprivation has
been linked to 5 of the top 15 leading causes of death including
cardio- and cerebrovascular diseases, accidents, T2DM, and
hypertension28. Data also point to a role for sleep deprivation in
the risk of stroke, cancer, and neurodegenerative diseases
(NDDs)26,29,30. Sleep deprivation is also associated with psy-
chopathological and psychiatric disorders, including negative
mood and mood regulation, psychosis, anxiety, suicidal behavior,
and the risk for depression3136.
Both too short or too long sleep durations have been found to
be associated with adverse health outcomes and all-cause mor-
tality with an U-shaped relationship3739. Although the relation
of long sleep duration to adverse health outcomes may be con-
founded by poor health conditions occurring in older adults37,
the causal association of sleep deprivation with negative health
effects is substantiated by experimental evidence providing bio-
logical plausibility24,40,41.
Sleep profoundly affects endocrine, metabolic, and immune
pathways, whose dysfunctions play a determinant role in the
development and progression of chronic diseases4244. Speci-
cally, in many chronic diseases, a deregulated/exacerbated
immune response shifts from repair/regulation towards unre-
solved inammatory responses45.
Regular sleep is crucial for maintaining immune function
integrity and favoring a homeostatic immune defense to micro-
bial or inammatory insults46,47. Sleep deprivation may result in
deregulated immune responses with increased pro-inammatory
signaling, thus contributing to increase the risk for the onset and/
or worsening of infection, as well as inammation-related chronic
diseases.
Here we reviewed the evidence regarding the impact of sleep
deprivation on immune-related diseases by discussing the major
points as follows: (1) the immunesleep relationship; (2) the
association of sleep deprivation with the development and/or
progression of immune-related chronic diseases; and (3) the
immune consequences of sleep deprivation and their implications
for diseases. Finally, possible measures to reverse sleep
deprivation-associated immune changes were discussed.
Basic immune mechanisms of sleep regulation
The discovery of muramyl peptide, a bacterial cell wall compo-
nent that is able to activate the immune system and induce the
release of sleep-regulatory cytokines, primary regulators of the
inammatory system, provided the rst molecular link between
the immune system and sleep48. Thereafter, other microbial-
derived factors such as the endotoxin lipopolysaccharide (LPS)49,
as well as mediators of inammation, such as the cytokines
interleukin (IL)-1 and tumor necrosis factor (TNF)-α, pros-
taglandins (PGs), growth hormone-releasing hormone (GHRH),
and growth factors, were recognized as sleep-regulating factors50.
Along this line, most animal studies have consistently shown a
role in particular for IL-1, TNF-α, and PGD
2
in the physiologic,
homeostatic non-rapid eye movement (NREM) sleep regulation,
so that the inhibition of their biological action resulted in
decreased spontaneous NREM sleep, whereas their
administration enhanced NREM sleep amount and intensity, and
suppressed rapid eye movement (REM) sleep5153. Moreover, the
circulating levels of IL-1, IL-6, TNF-α, and PGD
2
are highest
during sleep54. Their effects are dose- and time-of-day-dependent
so that, for instance, low doses of IL-1 enhance NREMS, whereas
high doses inhibit sleep55. Reciprocal effects may be involved in
sleep regulation: for instance, the effects of systemic bacterial
products such as LPS may also involve TNF-α49. Links exist
between IL-1βand GHRH/growth hormone (GH) in promoting
sleep so that IL-1 induced GH release via GHRH56, and hypo-
thalamic γ-aminobutyric acid (GABA)ergic neurons (promoting
sleep) are responsive to both GHRH and IL-1β57. Instead, anti-
inammatory cytokines, including IL-4, IL-10, and IL-13, inhib-
ited NREM sleep in animal models58.
Through these substances, the immune system may signal to
the brain and interact with other factors involved in sleep reg-
ulation such as neurotransmitters (acetylcholine, dopamine,
serotonin, norepinephrine, and histamine), neuropeptides
(orexin), nucleosides (adenosine), the hormone melatonin, and
the hypothalamus-pituitary axis (HPA) axis. Signaling
mechanisms to the brain also involve vagal afferents: for
instance, vagotomy attenuates intraperitoneal TNF-α-enhanced
NREMS responses59.
Cytokines are produced by a vast array of immune cells,
including those resident in the central nervous system (CNS), and
non-immune cells, e.g., neurons, astrocytes and microglia, and
peripheral tissue cells60,61. Cytokines interact with the
brain through humoral, neural, and cellular pathways, and form a
brain cytokine network (Fig. 1) able to produce cytokines, their
receptors, and amplify cytokine signals50. Peripheral cytokines
reach the brain through different non-exclusive mechanisms,
including bloodbrain barrier (BBB) disruption62, penetration of
peripheral immune cells, and via afferent nerve bers, such as the
vagus nerve, a bundle of parasympathetic sensory bers that
conveys information from peripheral organs to the CNS63.
In the CNS, cytokines mediate a multiplicity of immunological
and nonimmunologic biological functions64, such as synaptic
scaling, synapse formation and elimination, de novo neurogen-
esis, neuronal apoptosis, brain development, cortical neuron
migration65, circuit homeostasis and plasticity66, and cortical
neuron migration65, and complex behaviors, sleep, appetite,
aging, learning and memory65, and mental health status67,68.
A common experimental nding is that after damage to any
brain area, if the animal or human survives, sleep always ensues69.
Recent evidence indicates that sleep is a self-organizing emergent
neuronal/glial network property of any viable network regardless
of size or location, whether in vivo or in vitro53,7073. Several
sleep-regulatory substances, e.g., TNF, IL-1, nitric oxide, PGs, and
adenosine are all produced within local cell circuits in response to
cell use74,75.
From this point of view, TNF-αand IL-1 are closely inter-
connected and play a similar role in the regulation of sleep7681.
IL-1βand TNF-αself-amplify and increase each others mRNA
expression in the brain82. In rats, IL-183 and TNF-α84 mRNAs
show diurnal variations in different brain areas, with the highest
concentrations recorded during increased sleep propensity and
peaks occurring at time of sleep period onset in rats and mice85.
Sleep-like states in mixed cultures of neurons and glia are
dependent in part on the IL-1 receptor accessory protein
(AcP)69,86. In the brain, there is an AcP isoform, neuron-specic
(AcPb)87, whose mRNA levels increase with sleep loss88,89. AcPb
is anti-inammatory, whereas AcP is pro-inammatory87,88.
TNF signaling promotes sleep, whereas reverse TNF-αsignal-
ing (the soluble TNF receptor) promotes waking90. The
brain production of TNF-αis neuron activity-dependent91.
Afferent activity into the somatosensory cortex enhances TNF
REVIEW ARTICLE COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4
2COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www.nature.com/commsbio
expression92, and in vitro optogenetic stimulation enhances
neuronal expression of TNF immunoreactivity93.
Peripheral immune activation following acute or chronic
infection or inammatory diseases is marketed by altered cyto-
kine concentrations and proles, and is transmitted to the CNS
initiating specic adaptive responses. Among these, a sleep
response is induced and has been hypothesized to favor recovery
from infection and inammation, supposedly via the timely
functional investment of energy into the energy-consuming
immune processes54,94. Accordingly, acute mild immune activa-
tion enhances NREM sleep and suppresses REM sleep, whereas
severe immune response with an upsurge of cytokine levels causes
sleep disturbance with the suppression of both NREM and REM
sleep49,9598. This sleep change correlates to the course of the host
immune response as observed in bacterial and Trypanosoma
infections97,99. Supportively, the increase in NREM sleep was a
favorable prognostic factor for rabbits during infectious
diseases96.
Immune regulators also mediate the complex interrelation
between sleep and the circadian systems74. Circadian rhythms in
behavior and physiology are generated by a molecular clockwork
located in the suprachiasmatic nucleus, i.e., the master circadian
pacemaker, and peripheral tissues, and involving the so-called
clock genes (Clock,Bmals,Npas2,Crys,Pers,Rors, and Rev-
erbs)100. Cytokines, including TNF-α, IL-1β101,102, and
LPS103105, suppress the peripheral and hypothalamic expression
of core clock genes and clock-controlled genes, resulting in
reduced locomotor activity accompanied by prolonged rest
time101.
Sleep deprivation and immune-related disease outcomes
In the following section, the association between sleep deprivation
and risk or outcomes of immune-related disorders, as observed in
human studies (mostly observational) and animal experimenta-
tions, will be examined. In this context, considering the
sleepimmunity relationship, research has also begun to explore
whether and how immune deregulation and inammation may
link sleep deprivation with adverse health outcomes.
Infection. A breakdown of host defense against microorganisms
has been found in sleep-deprived animals, as shown by the
increased mortality after septic insult in sleep-deprived mice
compared with control mice106, or by systemic invasion by
opportunistic microorganisms leading to increased morbidity and
lethal septicemia in sleep-deprived rats107. There is growing
evidence associating longer periods of sleep with a substantial
reduction in parasitism levels108 and reduced sleep quality with
increased risk of infection and poor infection outcome109,110.
Accordingly, patients with sleep disorders exhibited a 1.23-fold
greater risk of herpes zoster than did the comparison cohort111.
Furthermore, sleep-deprived humans, as those with habitual short
sleep (5 h) compared with 78 h sleep, are more vulnerable to
respiratory infections in cross-sectional and prospective
studies112,113, and after an experimental viral challenge109,114.
Similarly, compared with long sleep duration (around 7 h), short
sleep duration (around 6 h) is associated with an increased risk of
common illnesses, including cold, u, gastroenteritis, and other
common infectious diseases, in adolescents115.
Compared with non-sleep-deprived mice, REM-sleep-deprived
mice failed to control Plasmodium yoelii infection and, conse-
quently, presented a lower survival rate110. This was correlated to
an impaired T-cell effector activity, characterized by a reduced
differentiation of T-helper cells (Th) into Th1 phenotype and
following production of pro-inammatory cytokines, such as
interferon (IFN)-γand TNF-α, and compromised differentiation
into T-follicular helper cells (Tfh), essential to B-cell maturation,
which therefore resulted to be reduced110. Accordingly, both Maf,
a Tfh differentiation factor, and T-bet, a pro-Th1 transcription
factor, were reduced in the REM-sleep-deprived group110. The
combination of REM-sleep deprivation and P. yoelii infection
Fig. 1 Brain cytokines (CYT, orange circles) network and different suggested routes by which peripherally released inammatory signals can bypass
the bloodbrain barrier and circumventricular organs (CVOs), and activate the central nervous system: humoral, cellular, and neural pathways. CCL-2:
C-C motif chemokine ligand-2; CXCL-10: C-X-C motif chemokine ligand 10; IL-1: interleukin-1; mNTS: medial nucleus tractus solitarius; PGE
2
: prostaglandin
E2; TNF-α: tumor necrosis factor-α.
COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4 REVIEW ARTICLE
COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www .nature.com/commsbio 3
resulted in an additive effect on glucocorticoid synthesis, and
chemical inhibition of this exacerbated glucocorticoid synthesis
reduced parasitemia, death rate, and restored CD4 T-cell, Tfh,
and plasma B-cell differentiation in infected sleep-deprived
mice110, suggesting a role of HPA axis hyperactivation in
impairing host immune response under sleep deprivation.
Seep deprivation may exert detrimental effects on sepsis-
induced multi-organ damage. Sleep deprivation (3 days) after LPS
administration increased the levels of pro-inammatory cytokines
(IL-6 and TNF-α) in the plasma and organs (lung, liver, and
kidney), which could be abrogated by subdiaphragmatic vagot-
omy or splenectomy 14 days prior to LPS administration116. Gut
microbiota-vagus nerve axis and gut microbiota-spleen axis may
play essential roles in post-septic sleep deprivation-induced
aggravation of systemic inammation and multi-organ
injuries116.
Considering the association between sleep deprivation and
immune response to infections, vaccination studies allow to assess
the impact of sleep and sleep loss on ongoing immune response
and the clinical outcome. Studies in which sleep deprivation (one
or few nights) was applied to healthy humans during (mostly
after) the immunological challenge of vaccination demonstrate
that sleep deprivation reduced both the memory and effector
phases of the immune response, as indexed by suppressed
antigen-specic antibody and Th cell response compared with
undisturbed sleep117.
Congruently, habitual (and hence chronic) short sleep duration
(<6 h) compared with longer sleep duration was associated with
reduced long-term clinical protection after vaccination against
hepatitis B118. Sleep deprivation did not exert any impairing effect
on mice already immunized119. From these studies, it seems that
sleep supportsand sleep deprivation impedesthe formation of
the immunological memory. Potential mechanisms involved in
the benecial effect of normal sleep on the vaccination response
include: (i) the sleep-induced reduction in circulating immune
cells that most likely accumulate into lymphatic tissues,
increasing the probability to encounter antigens and trigger the
immune response; (ii) the sleep-associated prole of inamma-
tory activation towards Th1 cytokines (increased IL-2, IFN-γ,
etc.), which may favor macrophage activation, antigen presenta-
tion, and T-cell and B-cell activation; (iii) the effect of sleep stage
on the formation of immunological memory through specic
immune-active hormones: indeed, during slow wave sleep-rich
early sleep, the prole of immune-active hormones, characterized
by minimum concentrations of cortisol, endowed with anti-
inammatory activity, and high levels of GH, prolactin, and
aldosterone, which support Th1 cell-mediated immunity, may
facilitate the mounting of an effective adaptive immune response
to a microbial challenge54.
Cancer. Sleep deprivation has increasingly been recognized as a
risk factor for impaired anti-tumor response. Epidemiological
studies suggest, albeit not consistently120, a signicant association
between short sleep duration and the risk for several cancers,
including breast, colorectal, and prostate cancer29,121123.
Potential mechanisms underlying this association include a
shorter duration of nocturnal secretion of melatonin (putatively
due to increased light exposure at night)124, which exerts anti-
cancer properties through antimitotic, antioxidant, apoptotic,
anti-estrogenic, and anti-angiogenic mechanisms125. Melatonin
also plays immunomodulatory and anti-inammatory effects with
relevance for its anti-cancer activity, being able to inhibit the pro-
inammatory nuclear factor-κB (NF-κB)/NLRP3 inammasome
pathways, and to support T/B-cell activation and macrophage
function126. However, besides melatonin, impaired anti-tumor
immune response has been invoked in the sleep deprivation-
associated risk for cancer development. A reduced cytotoxic
activity of natural killer (NK) cells, which are immune cells with
anti-tumor effect, has been reported in 72 h sleep-deprived mice
compared with control mice, accompanied by reduced numbers
of the cytotoxic cells such as CD8 T cells and NK cells in the
tumor microenvironment after chronic sleep deprivation (for
18 h/day during 21 days) in an animal model of experimental
pulmonary metastasis127,128. In this model, the reduced anti-
tumor immunity of sleep-deprived animals was also indexed by
the reduced number of antigen-presenting cells (dendritic cells)
in the lymph nodes, as well as by the decreased effector CD4
T-cell numbers and corresponding cytokine prole (decreased
IFN-γ), resulting in lowered Th1 response of Th cells, i.e., the
most effective immune response against tumors. Therefore, an
immunosuppressive environment develops with sleep depriva-
tion, which could translate into an early onset and increased
growth rate of cancer128 or increased mortality129.
An integrated meta-analysis of transcriptomic data showed
that circadian rhythm-related genes are downregulated and
upregulated in the cortex and hypothalamus samples of mice
with sleep deprivation, respectively, with downregulated genes
associated with the immune system and upregulated genes
associated with oxidative phosphorylation, cancer, and
T2DM130. Several circadian rhythm-related genes were common
to both T2DM and cancer, and seem to associate with malignant
transformation and patient outcomes130.
Hence, although these sleep deprivation-induced immune-
mediated mechanisms in cancer warrant further conrmation in
humans, the importance of the immune function in the anti-
tumor host defense is well recognized131, thus suggesting that the
impaired immune response after sleep deprivation may represent
a plausible mediator of the associated increased risk for cancer as
described in animal models and in humans.
Neurodegenerative diseases. NDDs are aging-related diseases
that selectively target different neuron populations in the CNS,
and include Alzheimers disease, multiple sclerosis, Parkinsons
disease, Huntingtons disease, and amyotrophic lateral sclerosis.
One prevailing hypothesis is that altered sleep habits and speci-
cally sleep deprivation may be a consequence and frequently a
marker of the disease132134. However, human and animal studies
have also suggested a causative or contributing role for sleep
deprivation in the development and/or worsening of neurode-
generative processes132134.
Potential pathophysiological mechanisms involve, among
others, neuro-immune dysregulation. Indeed, a common feature
and a potential therapeutic target- of NDDs is the chronic
activation of the immune system, where aspects of peripheral
immunity and systemic inammation integrate with the brains
immune compartment, leading to neuroinammation and
neuronal damage135. Neuroinammation following sleep depri-
vation has been studied as a pathogenic mechanism potentially
mediating the association between sleep deprivation and
neurodegenerative processes. Low-grade neuroinammation as
indexed by heightened levels of pro-inammatory mediators (e.g.,
TNF-α, IL-1β, and COX-2) and activation of astrocytes and
microglia, main immune cells in the brain, was observed in the
hippocampus and piriform cortex regions of the brain of chronic
sleep-deprived rats along with neurobehavioral alterations
(anxiety, learning, and memory impairments)136. The sleep
deprivation pro-inammatory milieu was accompanied by
oxidative stress in the brain137 and BBB disruption with
consequent increased permeability to blood components138. After
acute sleep deprivation, there was a signicant increased
REVIEW ARTICLE COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4
4COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www.nature.com/commsbio
recruitment of B cells in the mouse brain, which could be
important given evidence of B cells involvement in NDDs139.
Progressive and chronic aggregations of unique proteins in the
brain and spinal cord are hallmarks of NDDs140 and trigger
inammatory responses, gradual loss of physiological functions of
the nerve cells, and cell death141. Impaired autophagy in humans,
a catabolic process of cytoplasmic components, contributes to the
aggregation and accumulation of β-amyloid (Aβ), cytoskeleton-
related protein τ, and synuclein in neuronal cells and tissues140.
Sleep plays an important role in the clearance of metabolic waste
products accumulated during wakefulness and neural activity.
Indeed, the Aβprotein is predominantly cleared from the brain
during sleep, possibly through the glymphatic pathway. Con-
gruently, acute and chronic experimental sleep deprivation in
animals142,143 and humans144 resulted in brain Aβaccumulation
and plaque formation, a typical pathological change in Alzhei-
mers disease process, the most common type of dementia.
Imaging studies have revealed that healthy humans with self-
reported short sleep were more prone to have cerebral Aβplaque
pathology145 and disruption of deep sleep (slow wave sleep)
increases Aβin human cerebrospinal uid (CSF)146. Likewise,
patients with insomnia present higher CSF levels of Aβ147.
This pathological Aβaccumulation might reect disrupted
balance of Aβproduction and clearance after sleep deprivation.
On the one hand, sleep deprivation results in reduced clearance as
suggested by clinical studies showing that Aβlevels in CSF are the
highest before sleep and the lowest after wakening, whereas Aβ
clearance from CSF was impaired by sleep deprivation148.
Impaired clearance might also derive from disrupted peripheral
Aβtransport, as suggested by the sleep deprivation-induced
downregulation of low-density lipoprotein receptor-related pro-
tein-1 (LRP-1), which promotes Aβefux from the brain to the
peripheral circulation across the BBB, and elevations of receptor of
advanced glycation end products (RAGE), which promotes on the
contrary the inux of peripheral Aβinto the brain, thus
preventing Aβclearance149. On the other hand, apart from
impairing Aβand τinterstitial uid clearance, sleep deprivation
may also have a role in increasing Aβand τexocytosis, thereby
increasing CSF Aβand τlevels150. In animals, sleep deprivation
also leads to upregulation of β-secretase 1 (BACE-1), the most
important enzyme regulating Aβgeneration in the brain142,143,149,
thus opening the hypothesis of increased Aβproduction by sleep
deprivation. Sleep deprivation-induced neuroinammatory med-
iators correlate and could lead to disturbed Aβclearance and
stimulated amyloidogenic pathway143, being pro-inammatory
cytokines able to suppress the expression of LRP-1 and to increase
RAGE151 and BACE-1 levels152. Likewise, oxidative stress induced
by sleep deprivation may also contribute to the neuroinamma-
tory burden and the increased expression of BACE-1153.
Furthermore, patients with insomnia, compared with healthy
controls, showed decreased serum levels of neurotrophins,
including brain-derived neurotrophic factor (BDNF), proteins
especially relevant in neuroplasticity, memory and sleep, and this
reduction was signicantly related to the insomnia severity154.
Sleep deprivation is associated with a rapid decline in
circulatory melatonin levels, which may be linked to rapid
consumption of melatonin as a rst-line defense against the sleep
deprivation-associated rise in oxidative stress155. Melatonin is a
potent antioxidant, interacts with BDNF156, and promotes
neurogenesis and inhibits apoptosis157. The neuroprotective
potential of melatonin can target events leading to Alzheimers
disease development including Aβpathology, τhyperphosphor-
ylation, oxidative stress, glutamate excitotoxicity, and calcium
dyshomeostasis150,158. Accordingly, melatonin treatment could
restore the autophagy ux, thereby preventing tauopathy and
cognitive decline in Alzheimers disease mice159.
Patients with Alzheimers disease have an increased incidence
of sleep-disordered breathing160. In addition, sleep-disordered
breathing is associated with an increased risk of mild cognitive
impairment or dementia and with earlier onset of Alzheimers
disease161. Sleep-disordered breathing is also associated with
altered levels of Alzheimers disease biomarkers in CSF, including
decreased levels of Aβand elevated levels of phosphorylated τ162.
Sleep-disordered breathing possibly via hypoxia, inammation,
and sleep disruption/deprivation could contribute to Alzheimers
disease processes, e.g., increase of Aβproduction and aggregation,
suppression of glymphatic clearance of Alzheimers disease
pathogenic proteins (τ,Aβ) and oxidative stress, inammation,
and synaptic damage134,163.
To summarize, the sleep deprivation-associated risk for
Alzheimers disease could be linked to the induction of
inammation in the brain and disorders of systemic innate and
adaptive immunity164. However, the relationship of sleep
deprivation to inammation in Alzheimers disease is mostly
speculative and needs to be conrmed.
Similar to Aβin Alzheimers disease, abnormal levels of α-
synuclein are common to Parkinsons disease, the second most
common NDDs165. Sleep disturbances are not only a common
comorbidity in Parkinsons disease, but often precede the onset of
classic motor symptoms166. The main pathological features of
Parkinsons disease are the reduction of dopaminergic neurons in
the extrapyramidal nigrostriatal body and the formation of Lewy
bodies formed by the aggregation of α-synuclein and its oligomers
surrounded by neurolaments. Due to the degeneration of the
dopaminergic neurons, affected people show muscle stiffness,
resting tremors, and posture instability; other pathways involved
in sleep, cognition, mental abnormalities, and other non-motor
symptoms are also affected167. Epidemiological studies also
suggest that disturbed sleep may increase the risk of Parkinsons
disease168,169. Such disease-modifying mechanisms may include
activation of inammatory and immune pathways, abnormal
proteostasis, changes in glymphatic clearance, and altered
modulation of specic sleep neural circuits that may prime
further propagation of α-synucleinopathy in the brain169.
Melatonin could reduce neurotoxin-induced α-synuclein aggre-
gation in mice. Furthermore, melatonin pretreatment reduced
neurotoxin-induced loss of axon and dendritic length in
dopaminergic neurons through suppression of autophagy acti-
vated by CDK5 and α-synuclein aggregation, thereby reducing
dyskinesia symptoms in Parkinsons disease animal models170.A
few reports have shown that melatonin exerts protective effects in
several experimental models of Parkinsons disease171.
However, although animal experimentations suggest a link
between sleep deprivation and immune dysfunction in neurode-
generative processes, no human investigations have yet conrmed
the mediating role of immune dysregulation in the association
between sleep deprivation and risk or outcomes of NDDs.
Autoimmune diseases. Sleep disturbances are frequently reported
in autoimmune diseases, and immunotherapy in patients with
autoimmune pathologies results in sleep improvement172. However,
knowledge of the immunopathology of autoimmune diseases have
disclosed new concepts on the impact of sleep deprivation on
autoimmune disease process, showing that sleep deprivation can
promote a breakdown of immunologic self-tolerance. Human
cohort studies found that non-apnea sleep disorders, including
insomnia, were associated with a higher risk of developing auto-
immune diseases such as rheumatoid arthritis, ankylosing spon-
dylitis, systemic lupus erythematosus, and systemic sclerosis
(adjusted hazard ratio: 1.47, 95% condence interval (CI)
1.411.53)173 Similarly, in relatives of systemic lupus erythematosus
COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4 REVIEW ARTICLE
COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www .nature.com/commsbio 5
patients, and hence at increased risk for systemic lupus erythema-
tosus, self-reported short sleep duration (<7 h/night) was associated
with transitioning to systemic lupus erythematosus (adjusted odds
ratio: 2.0, 95% CI 1.14.2), independent of early preclinical features
that may inuence sleep duration such as prednisone use, depres-
sion, chronic fatigue, and vitamin D deciency174.Thisroleofsleep
deprivation as a risk factor for autoimmune diseases is corroborated
by animal studies. In mice genetically predisposed to develop sys-
temic lupus erythematosus175, chronic sleep deprivation, applied at
an age when animals were yet clinically healthy, caused an early
onset of the disease, as indexed by the increased number of anti-
nuclear antibodies, without affecting disease course or severity,
according to data on proteinuria, a surrogate marker of auto-
immune nephritis, and longevity. Several mechanisms have been
postulated to explain the link between sleep deprivation and auto-
immune disease risk. Sleep deprivation can accelerate disease
development through mechanisms including sleep deprivation-
induced increased production of several pro-inammatory
cytokines44,54, as better discussed below. Indeed, cytokines are
synergistically involved in the pathogenesis of autoimmunity, such
as IL-6, whose abnormal production results in polyclonal B-cell
activation and the occurrence of autoimmune features176,andIL-17
and the related Th17-cell response177, which require IL-6 for
activation178 and can cause greater amounts of autoantibody pro-
duction and immune complex formation, or can intensify chronic
inammation by promoting angiogenesis and recruiting of
inammatory cells at inammationsitesaswellascartilageand
bone erosion179. Furthermore, experimentally sleep-deprived heal-
thy humans showed impaired suppressive activity of CD4 reg-
ulatory T cells (Treg), which normally is highest during the night
and lowest in the morning180. The suppressive function of Treg
towards excessive immune response is an important homeostatic
mechanism, whose impairment is implicated in autoimmune dis-
ease pathogenesis181. Hence, sleep deprivation may not be merely
an early symptom or a consequence of an autoimmune disease, but
may contribute directly to the pathogenesis increasing the sus-
ceptibility to develop an autoimmune disease. More studies are
warranted in this eld.
Metabolic and vascular diseases. Prospective epidemiological
evidence associate sleep deprivation (commonly <7 h/night, often
<5 h/night) with the incidence of fatal and non-fatal CV out-
comes, with a 48% higher risk of coronary heart disease25, a 15%
higher risk of stroke182, and a 12% increased risk of all-cause
mortality37, which is mainly due to CV causes, according to some
authors183. In a recent prospective cohort, a low-stable sleep
pattern (<5 h sleep/night) during the 4-year follow-up had the
highest risk of death and CV events184. Short sleep has also been
associated with increased subclinical atherosclerotic burden, the
dominant underlying cause of CV diseases185.
In addition, sleep deprivation increases the risk for obesity
(about 55% higher risk)39,186, insulin resistance, T2DM (28%
higher risk)38, and hypertension (21% higher risk)187, which are
powerful and preventable risk factors for CV diseases. Notably,
the risk for diabetes attributable to sleep deprivation is
comparable to that of other established traditional cardiometa-
bolic risk factors188, thus underscoring the clinical signicance of
targeting sleep deprivation in the prevention of cardiometabolic
diseases. In contrast with normal nocturnal sleep and in
particular NREM sleep characterized by a marked decrease in
sympathetic activity, catecholamine plasma levels, and blood
pressure, experimental sleep deprivation (acute or chronic) is
accompanied by increased sympathetic outow, with consequent
higher blood pressure and heart rate, thus providing a pathogenic
link between sleep deprivation and hypertension risk189192.
Regarding the inuence of sleep deprivation on metabolic
pathways, studies support a plausible causal link between sleep
deprivation and the risk of overweight and obesity, possibly
mediated by the effect of sleep deprivation on circulating levels of
hormones (leptin, ghrelin) controlling hunger, satiety and energy
balance, besides other factors intervening during sleep depriva-
tion, including physical inactivity and overfeeding193. Further-
more, human experimental evidence with chronic sleep
deprivation protocol demonstrate that sleep deprivation may
alter glucose metabolism194 and insulin sensitivity195, thus
increasing the risk for obesity and T2DM. The reduction in total
body insulin sensitivity observed after sleep deprivation (4.5 h per
night for 4 days) in healthy subjects was paralleled by impaired
peripheral insulin sensitivity, as demonstrated in subcutaneous fat
playing a pivotal role in energy metabolism195. Considering a
more chronic sleep deprivation, reduced insulin sensitivity was
reported in overweight adults after 14 days of experimental sleep
deprivation (5.5 h per night) compared with 8.5 h per night of
sleep196, and after habitual curtailment in sleep duration of 1.5 h
(<6 h of sleep per night) in healthy young adults with a family
history of T2DM197.
Although the mechanisms that underlie most associations
between short sleep duration and adverse cardiometabolic
outcomes are not fully understood, potential causative mechan-
isms involving immune-inammatory activation have been
postulated. It is indeed well established that the subclinical
inammatory status induced by sleep deprivation has pathogenic
implications for metabolic and CV risk factors (glucose
metabolism, diabetes, hypertension, atherogenic lipid prole,
endothelial dysfunction, and coronary calcication) and out-
comes (stroke and coronary heart disease)24. Accordingly, most
of the markers of systemic and cellular inammation (leukocyte
counts and activation state, cytokines, acute-phase proteins, and
adipose tissue-derived adipokines) found to be altered after sleep
deprivation have been epidemiologically and pathogenically
associated with insulin resistance, T2DM, and vascular
complications198. In fact, inammation is an early pathogenic
process during the development of obesity and insulin
resistance199. Many adipose tissue-released inammatory factors
with pro-atherogenic and pro-thrombotic actions have also been
regarded as a molecular link between obesity and atherosclerotic
CV diseases200. Furthermore, chronic inammatory processes are
rmly established as central to the development and clinical
complications of CV diseases, form the initiation, promotion and
progression of atherosclerotic lesions to plaque instability, and the
precipitation of thrombosis, the main underlying cause of
myocardial infarction or stroke. Most CV risk factors (adiposity,
insulin resistance, T2DM, hypertension, and dyslipidemia) act by
inducing or intensifying such underlying inammatory processes
that ultimately promote endothelial dysfunction, altered vascular
reactivity, innate and adaptive immune system activation,
leukocyte inltration into the vessel wall, and thus
atherogenesis201. Experimental sleep deprivation leads to
endothelial dysfunction, an early marker of atherosclerosis, as
indexed by impaired endothelial-dependent vasodilation or
increased levels of endothelial adhesion molecules191.
Among the inammatory markers, besides being a biomarker
of future risk for CV diseases and a predictor of clinical response
to statin therapy202, C-reactiove protein (CRP) has been shown to
be involved in the immunologic process that triggers vascular
remodeling and atherosclerotic plaque deposition202. CRP levels
lack diurnal rhythm and its liver production is stimulated by
cytokines including IL-6 and IL-17, which are upregulated by
sleep deprivation203. As such, although limited evidence have
found an elevation of circulating CRP following sleep
deprivation204, CRP is a prototypical inammatory factor with
REVIEW ARTICLE COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4
6COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www.nature.com/commsbio
the potential to mark andto some extent mediateCV risk
following sleep deprivation. Congruently, elevated and sustained
plasma levels of CRP have been observed in healthy humans after
prolonged sleep deprivation (5 or 10 nights), in concomitance
with increased heart rate190,203, lymphocyte pro-inammatory
activation, and production of cytokines (e.g., IL-1, IL-6, and IL-
17)203. Similarly, the increase in blood pressure and heart rate
observed after acute total sleep deprivation (40 h) was accom-
panied and even preceded by impaired vasodilation and by
increased levels of IL-6 and markers of endothelial dysfunction
and activation, such as cellular adhesion molecules (E-selectin,
ICAM-1, etc.)191. The sleep deprivation pro-atherogenic effect in
animal model of sleep fragmentation is mediated, at least in part,
by reduced hypothalamic release of hypocretin (i.e., orexin), a
wake-inducing neuropeptide, which limits the production of
leukocytes (monocytes and neutrophils) and atherosclerosis
development, and has been inversely associated with the risk of
myocardial infarction, heart failure, and obesity205. The activation
of the sympathetic nervous system (SNS) may be another
mechanism for the inammatory link between sleep loss and
atherosclerotic CV disease, because such activation increases the
bone marrow release of progenitor cells, the production of innate
immune cells (monocytes), and the levels of inammatory
cytokines, and triggers endothelial dysfunction, thereby leading
to systemic and vascular inammation and atherosclerosis206,207.
Playing a key role in instigating inammatory responses and
promoting atherosclerosis208, the sleep deprivation-associated
oxidative stress may also contribute to CV risk. It has also been
hypothesized a role for melatonin suppression following sleep
deprivation in the vascular impairment associated with sleep
deprivation, given that melatonin inhibits oxidative stress and
cytokine production by immune and vascular cells, and represses
atherosclerotic lesion formation in vivo209.
Therefore, a signicant and consistent association exists
between sleep deprivation and cardiometabolic risk and clinical
outcomes, with several plausible immune-mediated causative
mechanisms explaining this association.
Immune mechanisms linking sleep deprivation and diseases
As shown above, sleep deprivation has been found to alter
inammatory immune processes via multiple pathways, which
could lead to increased susceptibility to chronic inammatory
diseases (Fig. 2). Most of the current knowledge on immune
effects of sleep deprivation come from studies using controlled
experimental sleep deprivation protocols, among which chronic
partial sleep deprivation, lasting 215 days, is that mostly
resembling the human condition of chronic insufcient sleep.
Some studies have observed that sleep deprivation, compared
with regular nocturnal sleep, leads to increased circulating
numbers of total leukocytes and specic cell subsets mainly
neutrophils, monocytes, B cells, CD4 T cells, and decreased cir-
culating numbers and cytotoxic activity of NK cells203,210213.
Other studies, however, found contrasting results, including a
decrease in CD4 T cells after sleep deprivation213,214, probably
due to differences in sleep deprivation protocol, sampling meth-
odologies, and other factors. Sleep deprivation has also shown to
alter circadian rhythm of circulating leukocytes215, with higher
levels during the night and at awakening and a attened
rhythm210,212. Additional ndings are suggestive of immune
deregulation by sleep deprivation, including a decreased neu-
trophils phagocytic activity213, altered lymphocytes adhesion
molecule expression216, and reduced stimulated production of IL-
2 and IL-12, which are important for adaptive immunity211,217.
Experimental sleep deprivation has been reported to affect
systemic markers of inammation, with studies showing
increased circulating pro-inammatory molecules (IL-1, IL-6,
CRP, TNF-α, and MCP-1); this associated in some studies with a
subsequent homeostatic increase in endogenous inhibitors,
including IL-1 receptor antagonist and TNF receptors203,218220.
In agreement with experimental sleep deprivation, population
studies found a direct independent association between habitual
short sleep duration (generally < 5 or 6 h) and elevated circulating
pro-inammatory markers, e.g., acute phase proteins (CRP and
IL-6), cytokines (TNF-α, IFN-γ, IL-1, etc.), adhesion molecules,
and leukocyte counts183,221225. Furthermore, a reduced NK cell
activity226 and a decline in naive T cells227, compatible with
reduced immune competence, was reported in association with
habitual short sleep. Shortening of leukocyte telomere length, a
cellular senescence marker linked with inammation, was also
associated with shorter sleep duration228,229.
The reported elevation of systemic inammation is clinically
relevant, because it is suggested to specically mediate the
increased risk of mortality associated with short sleep23,230,231
and, as observed, the risk for chronic disease development.
Regarding cellular markers of inammation, some studies
found that the ex-vivo LPS-stimulated production of TNF-
α232,233, IL-1β, and IL-6203,232234 by human monocytes
increased during sleep deprivation but decreased during regular
nocturnal sleep54,203,232234. However, other studies reported a
decrease of TNF-αproduction by activated monocytes after sleep
deprivation compared with regular nocturnal sleep203,235. These
contrasting results need further investigations and may depend
on differences in the cytokine sensitivity to different sleep
deprivation protocols or sampling methods and time. For
instance, it seems that partial acute sleep deprivation increased
stimulated monocytic TNF-αproduction232,233, whereas more
sustained sleep deprivation decreased it203,235.
Undisturbed sleep is predominantly characterized by a Th1
polarization of Th cells (expressing IFN-γ, IL-2, and TNF-α), and
experimental sleep deprivation in humans leads to a shift from a
Th1 pattern towards a Th2 pattern (expressing IL-4, IL-5, IL-10,
and IL-13)217,236. Accordingly, conditions featured by disturbed
sleep with specicdecit in slow wave sleep, as observed in
elderly people237, alcoholic238, and insomnia239 patients, show a
cytokine shift towards Th2. The balance of Th1/Th2 immunity
and its shift during sleep deprivation may have crucial implica-
tions in anti-microbial and anti-tumor immune responses. Th2
over-activity is known to be involved in some forms of allergic
responses, and to increase the susceptibility to infection240.
Likewise, regarding the anti-tumor immune action, Th1 response
supports cytotoxic lymphocytes and tumor cells destruction with
the potential of elimination or control of tumor cell growth, so
that a type 1 adaptive immune response (increased antigen pre-
sentation, IFN-γsignaling, and T-cell receptor signaling) may be
associated with an improved survival or prognosis241,242.In
contrast, Th2 over-response is thought to contribute to tumor
development and progression, by limiting cytotoxic T lympho-
cytes proliferation and by the modulation of other inammatory
cell types241.
Several cellular and molecular signaling pathways may be
involved in mediating the inuence of sleep deprivation on
immune and inammatory functions (Fig. 3). Increased oxidative
stress markers and/or decreased antioxidant defense have been
found after sleep deprivation243245. Sleep shows an antioxidant
function, responsible for eliminating reactive oxygen species
produced during wakefulness, and contrarily sleep deprivation
may cause oxidative stress, which leads to cell senescence,
unbalanced local/systemic inammation, dysmetabolism, and
immune derangements246,247.
Effects of sleep deprivation on the immune response may
derive from the activation of the SNS with the corresponding
COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4 REVIEW ARTICLE
COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www .nature.com/commsbio 7
increase in systemic catecholamines22,248. Catecholamines signal
to immune cells via adrenergic receptors, which are primarily α-
and β-adrenergic in myeloid cells and β-adrenergic in
lymphocytes249. The immune outcome of the sympathetic sig-
naling is complex, and includes both stimulatory and inhibitory
effects depending on cell and receptor types, cell development/
activation states, and local microenvironment249,250. Some evi-
dence suggest that β-adrenergic signaling inhibits and α-
adrenergic signaling promotes excessive inammation under
endotoxemia250. Activation of α-adrenergic signaling in periph-
eral tissues induces the upregulation of pro-inammatory
cytokines250,251. Sympathetic activation also suppresses the
transcription of type I IFNs (IFN-αand IFN-β) genes and
interferon response genes, which play a key role in anti-viral
immunity252, and inhibits via β-adrenergic signaling the anti-
tumor cytotoxicity of T lymphocytes253. In vitro β-adrenergic
stimulation repressed Th1 response and stimulated Th2 response,
with varying effects found in vivo249,254. Although the specic
role of SNS activation in the immune phenotype associated with
sleep deprivation is not clearly established, data suggest a pro-
inammatory effect of SNS under sleep deprivation. Indeed,
chemical sympathectomy has been recently shown to alleviate the
inammatory response following chronic sleep deprivation in
mice255, and both α- and, to a lesser extent, β-adrenergic recep-
tors seem to contribute to the sympathetic regulation of inam-
matory responses to sleep deprivation256.
At the molecular levels, sleep deprivation led to signicant gene
expression changes in animal tissues257259 and human blood
monocytes203,233,260262, with affected genes mostly related to
immune and inammatory processes (leukocyte function, Th1/
Th2 balance, cytokine regulation, and TLR signaling), oxidative
stress, stress response, apoptosis, and circadian system, collec-
tively indicating immune activation and hyperinammation.
Sleep loss and mistimed sleep also led in the blood tran-
scriptome to alteration and reduction in the circadian rhythmicity
of gene expression261,263, which is an integral part of basic bio-
logical processes and homeostasis264266.
The activation of the pro-inammatory NF-κB/Rel family of
transcription factors by sleep deprivation, rst demonstrated in
the late 1990s in mice267, and subsequently widely
conrmed233,260,261,268272, is one of the most consistent ndings
regarding upstream transcriptional regulation. NF-κB induces the
expression of genes (e.g., cytokines/chemokines, growth factors,
receptors/transporters, enzymes, adhesion molecules) involved in
inammation, immunity, proliferation, and apoptosis273, circa-
dian clock activity274, and sleep propensity275. Potential
signals for NF-κB activation under sleep deprivation include
increased adenosine levels, oxidative stress, altered metabolism
(adiposity and decreased insulin sensitivity), brain proteins/
metabolites (e.g., Aβ), melatonin suppression276, circadian clock
proteins277, and catecholamine surge due to increased sympa-
thetic activity278. Given the role of NF-κB in the pathophysiology
of inammatory diseases273, its activation under sleep deprivation
may be a common pathway for the risk of morbidity and
mortality.
The intestinal microbiota is also affected by sleep loss279281,
showing indices of dysbiosis (increased Firmicutes:Bacteroidetes
ratio; decreased diversity and richness), which may affect the
immune system282, and are similar to those associated with car-
diometabolic diseases45.
Fig. 2 Immune consequences of sleep deprivation. Sleep deprivation, as induced experimentally or in the context of habitual short sleep, has been found to
be associated with alterations in the circulating numbers and/or activity of total leukocytes and specic cell subsets, elevation of systemic and tissue (e.g.,
brain) pro-inammatory markers including cytokines (e.g., interleukins [IL], tumor necrosis factor [TNF]-α), chemokines and acute phase proteins (such as
C reactive Protein [CRP]), altered antigen presentation (reduced dendritic cells, altered pattern of activating cytokines, etc.), lowered Th1 response, higher
Th2 response, and reduced antibody production. Furthermore, altered monocytes responsiveness to immunological challenges such as lipopolysaccharide
(LPS) may contribute to sleep deprivation-associated immune modulation. Hypothesized links between immune dysregulation by sleep deprivation and the
risk for immune-related diseases, such as infectious, cardiovascular, metabolic, and neurodegenerative and neoplastic diseases, are shown. The illustrations
were modied from Servier Medical Art (http://smart.servier.com/), licensed under a Creative Common Attribution 3.0 Generic License. APC: antigen-
presenting cells.
REVIEW ARTICLE COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4
8COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www.nature.com/commsbio
Countermeasures for sleep deprivation: effect on immune
parameters
Although the impact of strategies to improve sleep duration on
neurobehavioral performance and alertness after sleep depriva-
tion have been assessed283285, sleep deprivation countermeasures
to improve immune and inammatory parameters, and, corre-
spondingly, disease risk and outcomes have been studied to a
lesser extent.
Although extension of habitual short sleep did not show to
signicantly counterbalance the immune consequence of sleep
deprivation286288, mixed results derive from nighttime recovery
sleep following sleep deprivation (Table 1), with limited evidence
of effectiveness for specic immune parameters210,214, and mostly
after multiple consecutive nights of 8 h sleep recovery or with an
extended nocturnal sleep duration212,289.
Although daytime napping (<20 min) restores alertness, and
mental and physical performance without provoking sleep inertia
associated with longer nap290292, the effects of a short nap on
immune/inammatory parameters after sleep deprivation have
yet to be rmly established. Differently form population
studies293, laboratory studies found immune benet from
nap218,289,294,295. Regarding immune-related clinical outcomes,
controversy exists, with studies nding no association296, inverse
associations297,298 or positive association296, and a J-shaped
relationship299301 between napping and CV and metabolic dis-
eases or cancer events and mortality. Whether changes in
immune parameters could contribute to the associations between
napping and immune-related diseases remains unclear.
Among the strategies to recover sleep deprivation-induced
immune changes, cognitive behavior therapy improves sleep
outcomes in insomnia and lowers cellular and systemic inam-
matory markers302,303, and the risk score composed of CV and
metabolic risk factors304. This highlights the potential role of
targeting sleep in reducing the inammatory risk and the asso-
ciated chronic diseases.
Summary and concluding remarks
Sleep exerts immune-supportive functions and impairments of
the immune-inammatory system are a plausible mechanism
mediating the negative health effects of sleep deprivation, and in
particular, its role in the risk and outcomes of chronic diseases
such as infections, CV, metabolic and autoimmune diseases,
NDDs, and cancer. Caution should be exercised in interpreting
cellular and molecular outcomes of sleep deprivation in experi-
mental studies conducted till now as a result of an independent
effect of sleep deprivation, because other factors may play a role,
including extended wakefulness-associated processes, other fea-
tures of sleep-wakefulness, their temporal and functional segre-
gation or methodologies of sleep manipulation.
Randomized controlled trials assessing the effect of treatment
of sleep deprivation on inammatory immune dysfunction and/
or health outcomes are needed. Knowledge of inammatory and
immunological signatures in response to sleep curtailment may
inform not only on the underlying molecular links, but also
contribute to rene risk proles to be used for developing bio-
markers of sleep deprivation and sleep disturbance-related health
Fig. 3 Pro-inammatory molecular pathways induced by sleep deprivation. A schematic model of potential mechanistic pathways linking sleep
deprivation and inammatory immune activation is depicted. Sleep deprivation is associated with activation of the sympathetic nervous system and release
of norepinephrine and epinephrine into the systemic circulation, as well as to some extent with impaired hypothalamus-pituitary axis stimulation. These
neuromediators may act along with other potential stimuli accumulated following sleep deprivation including reactive oxygen species (ROS), adenosine,
metabolic waste products (e.g., β-amyloid) not cleared during normal sleep, gut microbiota dysbiosis leading to altered local and systemic pattern of
metabolic products, as well as with changes in the prole of neuro-endocrine hormones, such as prolactin, growth hormone, and altered circadian rhythm
of melatonin secretion. In immune cells located in the brain and the peripheral tissues, these stimuli may in concert trigger inammatory activation, with
release of cytokines, chemokines, acute phase protein, etc. via the recruitment of transcriptional regulators of pro-inammatory gene expression, mainly
nuclear factor (NF)-κB, and disturbing the circadian rhythmicity of gene expression of both clock genes and metabolic, immune and stress response genes
(see text for further detail). E: epinephrine; NE: norepinephrine; TLR: Toll-like receptor. Arrows indicate stimulation; lines indicate inhibition. The
illustrations were modied from Servier Medical Art (http://smart.servier.com/), licensed under a Creative Common Attribution 3.0 Generic License.
COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4 REVIEW ARTICLE
COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www .nature.com/commsbio 9
Table 1 Main human ndings on the effects of recovery sleep on sleep deprivation-induced changes in immune and inammatory parameters.
Subjects
(number and
age range
or mean)
Sleep
deprivation
protocol
Effect of sleep
deprivation on
immune parameters
compared with
baseline
Recovery sleep
protocol
Effect of recovery sleep
on immune parameters
compared with baseline
Effect of recovery sleep
on immune parameters
compared with sleep
deprivation
Reference
Healthy men and
women (n=20,
2130 yrs)
64 h TSD Granulocytes and
monocytes, NK cell
activity
1 Night (h sleep
not reported)
Granulocytes,
monocytes; =NK cell
activity
=Granulocytes;
monocytes;
NK cell activity
Dinges
et al.214
Healthy men
(n=32,
1929 yrs)
2 Nights TSD or
4 nights
of REM SD
TSD: total leukocytes,
neutrophils, CD4
T cells; REM SD: IgA
3 Nights (8 h
sleep/night)
=Total leukocytes,
neutrophils; CD4
T cells IgA
ND Ruiz
et al.308
Healthy young
men (n=10,
2129 yrs)
1 Night TSD During SD:
monocytes,
lymphocytes, NK cells.
The day after SD:
lymphocytes,
NK cells
1 Night
(8 h sleep)
=Monocytes,
lymphocytes; NK cells
ND Born
et al.210
Healthy men
(n=12, mean
age 29 yrs)
40 h TSD Plasma E-selectin;
systolic BP, heart
rate; plasma
norepinephrine;
endothelium-
dependent and
-independent
vasodilation
1 Night
(8 h sleep)
Plasma ICAM-1, IL-6,
norepinephrine
ND Sauvet
et al.191
Healthy men
(n=31,
1827 yrs)
1 Night with
2 h sleep
Total leukocytes,
neutrophils
1 Night of 8 h
sleep or 1 night
of 10 h sleep
8 h Recovery sleep:
leukocytes, neutrophils;
10 h recovery
sleep: =leukocytes,
neutrophils
8 h Recovery
sleep: =leukocytes,
neutrophils; 10 h recovery
sleep: leukocytes,
neutrophils
Faraut
et al.289
Healthy men
(n=19,
1929 yrs)
5 Nights with
4 h sleep/night
NK cells;
B cells;
plasma CRP;
IL-17, IL-1β,IL-6
(PBMC mRNA);
TNF-α(PBMC
protein)
2 Nights (8 h
sleep/night)
=NK cells; =B cells;
CRP, IL-17; =IL-1β, IL-6,
TNF-α
ND van
Leeuwen
et al.203
Healthy men
(n=9,
2227 yrs)
5 Nights with
4 h sleep/night
Total leukocytes,
monocytes,
neutrophils,
lymphocytes
7 Nights (8 h
sleep/night)
Monocytes,
lymphocytes;
neutrophils
ND Lasselin
et al.212
Healthy men and
women (n=24,
36-76 yrs)
1 Night with
4 h sleep
IL-6 and TNF-α
(PBMC protein)
1 Night
(8 h sleep)
IL-6 and TNF-αND Irwin
et al.262
Healthy men
(n=10,
2237 yrs) (Exp.
1) and healthy
88 h TSD (Exp.
1) and 10 days
with 4.2 h
Plasma CRP 3 Nights
(assessment
only in the rst
recovery day)
Plasma CRP ND Meier-
Ewert
et al.190
REVIEW ARTICLE COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4
10 COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www.nature.com/commsbio
outcomes, which may also represent potential targets of inter-
ventions. Recent metabolomic305 and transcriptomic306 studies
hold promise in biomarker discovery306.
These efforts may converge towards a new ground fostering
interactions between the sleep research and the medical com-
munity to translate scientic knowledge into the clinic, prioritize
health issues, and develop strategies and policies for subject risk
stratication, to include evidence-based sleep recommendations
in guidelines for optimal health and to address sleep hygiene at
the individual and the population levels, as a means to prevent the
negative health consequences of sleep deprivation. These actions
might also foster health literacy and empowerment of individuals
to actively better manage their own health and well-being
throughout their life course by means of lifestyle, nutritional, and
behavioral habits including sleep hygiene307.
Conclusively, in the perspective of staying healthy in this
rapidly changing society, the sleepimmunity relationship raises
relevant clinical implications for promoting sleep health and, as
evidenced here, for improving or therapeutically controlling
inammatory response by targeting sleep. This may ultimately
translate, in the era of preventive medicine, into addressing sleep
as a lifestyle approach along with diet and physical activity to
benet overall public health.
Reporting summary. Further information on research design is available in the Nature
Research Reporting Summary linked to this article.
Data availability
All data generated or analysed during this study are included in this published article.
Received: 12 January 2021; Accepted: 26 October 2021;
References
1. Luyster, F. S. et al. Sleep: a health imperative. Sleep 35, 727734 (2012).
2. Grandner, M. A. Sleep, health, and society. Sleep. Med. Clin. 12,122
(2017).
3. Ohayon, M. M., Carskadon, M. A., Guilleminault, C. & Vitiello, M. V. Meta-
analysis of quantitative sleep parameters from childhood to old age in healthy
individuals: developing normative sleep values across the human lifespan.
Sleep 27, 12551273 (2004).
4. Galland, B. C., Taylor, B. J., Elder, D. E. & Herbison, P. Normal sleep patterns
in infants and children: a systematic review of observational studies. Sleep.
Med. Rev. 16, 213222 (2012).
5. Galland, B. C. et al. Establishing normal values for pediatric nighttime sleep
measured by actigraphy: a systematic review and meta-analysis. Sleep 41,
https://doi.org/10.1093/sleep/zsy017 (2018).
6. Boulos, M. I. et al. Normal polysomnography parameters in healthy
adults: a systematic review and meta-analysis. Lancet Respir. Med. 7, 533543
(2019).
7. Consensus Conference, P. et al. Joint Consensus Statement of the American
Academy of Sleep Medicine and Sleep Research Society on the Recommended
Amount of Sleep for a Healthy Adult: methodology and discussion. J. Clin.
Sleep. Med. 11, 931952 (2015).
8. Consensus Conference, P. et al. Recommended amount of sleep for a healthy
adult: a Joint Consensus Statement of the American Academy of Sleep
Medicine and Sleep Research Society. J. Clin. Sleep. Med. 11, 591592 (2015).
9. Carskadon, M. A., Vieira, C. & Acebo, C. Association between puberty and
delayed phase preference. Sleep 16, 258262 (1993).
10. Gulia, K. K. & Kumar, V. M. Sleep disorders in the elderly: a growing
challenge. Psychogeriatrics 18, 155165 (2018).
11. Garbarino, S., Lanteri, P., Sannita, W. G., Bragazzi, N. L. & Scoditti, E.
Circadian rhythms, sleep, immunity, and fragility in the elderly: the model of
the susceptibility to infections. Front. Neurol. 11, 558417 (2020).
12. Zomers, M. L. et al. Characterizing adult sleep behavior over 20 years-the
Population-Based Doetinchem Cohort Study. Sleep 40,https://doi.org/
10.1093/sleep/zsx085 (2017).
13. American Academy of Sleep Medicine. International Classication Of Sleep
Disorders 3rd edn (American Academy of Sleep Medicine, 2014).
Table 1 (continued)
Subjects
(number and
age range
or mean)
Sleep
deprivation
protocol
Effect of sleep
deprivation on
immune parameters
compared with
baseline
Recovery sleep
protocol
Effect of recovery sleep
on immune parameters
compared with baseline
Effect of recovery sleep
on immune parameters
compared with sleep
deprivation
Reference
men and women
(n=10,
2638 yrs)
(Exp. 2)
sleep/night
(Exp. 2)
Healthy men and
women (n=21,
2539 yrs and
n=49, 60-
84 yrs)
1 night with
4 h sleep
IL-6 and TNF-α
(PBMC protein) in
younger adults
1 night
(8 h sleep)
IL-6 and TNF-αND Carroll
et al.309
Healthy men and
women (n=30,
1834 yrs)
6 Nights with
6 h sleep/night
Plasma IL-6 3 Nights (10 h
sleep/night)
=Plasma IL-6 Plasma IL-6 Pejovic
et al.310
Healthy men and
women (n=14,
1835 yrs)
5 Nights with
4 h sleep/night,
for 3 weeks
IL-6 (PBMC protein) 2 Nights (8 h
sleep/night), for
3 weeks
IL-6 (PBMC protein) ND Simpson
et al.234
BP blood pressure, exp experiment, ICAM-1 intercellular adhesion molecule-1, ND not determined, PBMC peripheral blood mononuclear cells, SD sleep deprivation, TSD total sleep deprivation, VCAM-1 vascular cell adhesion molecule-1, yrs years, signicant increase,
signicant decrease, =no signicant change.
COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4 REVIEW ARTICLE
COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www .nature.com/commsbio 11
14. Ford, E. S., Cunningham, T. J. & Croft, J. B. Trends in self-reported sleep
duration among US adults from 1985 to 2012. Sleep 38, 829832 (2015).
15. Gilmour, H. et al. Longitudinal trajectories of sleep duration in the general
population. Health Rep. 24,1420 (2013).
16. Matricciani, L., Olds, T. & Petkov, J. In search of lost sleep: secular trends in
the sleep time of school-aged children and adolescents. Sleep. Med. Rev. 16,
203211 (2012).
17. Wheaton, A. G., Jones, S. E., Cooper, A. C. & Croft, J. B. Short sleep duration
among middle school and high school students - United States, 2015. MMWR
Morb. Mortal. Wkly Rep. 67,8590 (2018).
18. Kocevska, D. et al. Sleep characteristics across the lifespan in 1.1 million
people from the Netherlands, United Kingdom and United States: a systematic
review and meta-analysis. Nat. Hum. Behav. https://doi.org/10.1038/s41562-
020-00965-x (2020).
19. Pandi-Perumal, S. R. et al. Racial/ethnic and social inequities in sleep
medicine: the tip of the iceberg? J. Natl Med. Assoc. 109, 279286 (2017).
20. Irwin, M. R. Why sleep is important for health: a psychoneuroimmunology
perspective. Annu. Rev. Psychol. 66, 143172 (2015).
21. Vgontzas, A. N., Liao, D., Bixler, E. O., Chrousos, G. P. & Vela-Bueno, A.
Insomnia with objective short sleep duration is associated with a high risk for
hypertension. Sleep 32, 491497 (2009).
22. Vgontzas, A. N., Fernandez-Mendoza, J., Liao, D. & Bixler, E. O. Insomnia
with objective short sleep duration: the most biologically severe phenotype of
the disorder. Sleep. Med. Rev. 17, 241254 (2013).
23. Smagula, S. F. et al. Actigraphy- and polysomnography-measured sleep
disturbances, inammation, and mortality among older men. Psychosom.
Med. 78, 686696 (2016).
24. Cappuccio, F. P. & Miller, M. A. Sleep and cardio-metabolic disease. Curr.
Cardiol. Rep. 19, 110 (2017).
25. Cappuccio, F. P., Cooper, D., DElia, L., Strazzullo, P. & Miller, M. A. Sleep
duration predicts cardiovascular outcomes: a systematic review and meta-
analysis of prospective studies. Eur. Heart J. 32, 14841492 (2011).
26. von Ruesten, A., Weikert, C., Fietze, I. & Boeing, H. Association of sleep
duration with chronic diseases in the European Prospective Investigation into
Cancer and Nutrition (EPIC)-Potsdam study. PLoS ONE 7, e30972 (2012).
27. Tobaldini, E. et al. Sleep, sleep deprivation, autonomic nervous system and
cardiovascular diseases. Neurosci. Biobehav. Rev. 74, 321329 (2017).
28. Kochanek, K. D., Xu, J. & Arias, E. Mortality in the United States, 2019. NCHS
Data Brief No. 395,18 (CDC, 2020).
29. Kakizaki, M. et al. Sleep duration and the risk of breast cancer: the Ohsaki
Cohort Study. Br. J. Cancer 99, 15021505 (2008).
30. Ahmadian, N., Hejazi, S., Mahmoudi, J. & Talebi, M. Tau pathology of
Alzheimer disease: possible role of sleep deprivation. Basic Clin. Neurosci. 9,
307316 (2018).
31. Baglioni, C. et al. Insomnia as a predictor of depression: a meta-analytic
evaluation of longitudinal epidemiological studies. J. Affect. Disord. 135,1019
(2011).
32. Ben Simon, E., Vallat, R., Barnes, C. M. & Walker, M. P. Sleep loss and the
socio-emotional brain. Trends Cogn. Sci. 24, 435450 (2020).
33. Bernert, R. A., Kim, J. S., Iwata, N. G. & Perlis, M. L. Sleep disturbances as an
evidence-based suicide risk factor. Curr. Psychiatry Rep. 17, 554 (2015).
34. Fredriksen, K., Rhodes, J., Reddy, R. & Way, N. Sleepless in Chicago: tracking
the effects of adolescent sleep loss during the middle school years. Child Dev.
75,8495 (2004).
35. Tomaso, C. C., Johnson, A. B. & Nelson, T. D. The effect of sleep deprivation
and restriction on mood, emotion, and emotion regulation: three meta-
analyses in one. Sleep 44,https://doi.org/10.1093/sleep/zsaa289 (2021).
36. Waters, F., Chiu, V., Atkinson, A. & Blom, J. D. Severe sleep deprivation
causes hallucinations and a gradual progression toward psychosis with
increasing time awake. Front. Psychiatry 9, 303 (2018).
37. Cappuccio, F. P., DElia, L., Strazzullo, P. & Miller, M. A. Sleep duration and
all-cause mortality: a systematic review and meta-analysis of prospective
studies. Sleep 33, 585592 (2010).
38. Cappuccio, F. P., DElia, L., Strazzullo, P. & Miller, M. A. Quantity and quality
of sleep and incidence of type 2 diabetes: a systematic review and meta-
analysis. Diabetes Care 33, 414420 (2010).
39. Bacaro, V. et al. Sleep duration and obesity in adulthood: an updated
systematic review and meta-analysis. Obes. Res. Clin. Pract. 14, 301309
(2020).
40. Bishir, M. et al. Sleep deprivation and neurological disorders. Biomed. Res. Int.
2020, 5764017 (2020).
41. Mullington, J. M., Simpson, N. S., Meier-Ewert, H. K. & Haack, M. Sleep loss
and inammation. Best. Pract. Res. Clin. Endocrinol. Metab. 24, 775784
(2010).
42. Aldabal, L. & Bahammam, A. S. Metabolic, endocrine, and immune
consequences of sleep deprivation. Open Respir. Med. J. 5,3143 (2011).
43. Dantzer, R. Neuroimmune interactions: from the brain to the immune system
and vice versa. Physiol. Rev. 98, 477504 (2018).
44. Irwin, M. R. Sleep and inammation: partners in sickness and in health. Nat.
Rev. Immunol. 19, 702715 (2019).
45. Hand, T. W., Vujkovic-Cvijin, I., Ridaura, V. K. & Belkaid, Y. Linking the
microbiota, chronic disease, and the immune system. Trends Endocrinol.
Metab. 27, 831843 (2016).
46. Irwin, M. R. & Opp, M. R. Sleep health: reciprocal regulation of sleep and
innate immunity. Neuropsychopharmacology 42, 129155 (2017).
47. Miller, M. A. & Cappuccio, F. P. Inammation, sleep, obesity and
cardiovascular disease. Curr. Vasc. Pharm. 5,93102 (2007).
48. Krueger, J. M., Pappenheimer, J. R. & Karnovsky, M. L. The composition of
sleep-promoting factor isolated from human urine. J. Biol. Chem. 257,
16641669 (1982).
49. Mullington, J. et al. Dose-dependent effects of endotoxin on human sleep. Am.
J. Physiol. Regul. Integr. Comp. Physiol. 278, R947955 (2000).
50. Zielinski, M. R. & Krueger, J. M. Sleep and innate immunity. Front. Biosci.
(Sch. Ed.) 3, 632642 (2011).
51. Opp, M. R. Cytokines and sleep. Sleep. Med. Rev. 9, 355364 (2005).
52. Urade, Y. & Hayaishi, O. Prostaglandin D2 and sleep/wake regulation. Sleep.
Med. Rev. 15, 411418 (2011).
53. Krueger, J. M., Majde, J. A. & Rector, D. M. Cytokines in immune function
and sleep regulation. Handb. Clin. Neurol. 98, 229240 (2011).
54. Besedovsky, L., Lange, T. & Haack, M. The sleep-immune crosstalk in health
and disease. Physiol. Rev. 99, 13251380 (2019).
55. Opp, M. R., Obal, F. Jr. & Krueger, J. M. Interleukin 1 alters rat sleep: temporal
and dose-related effects. Am. J. Physiol. 260, R5258 (1991).
56. Krueger, J. M. & Obal, F. Jr. Growth hormone-releasing hormone and
interleukin-1 in sleep regulation. FASEB J. 7, 645652 (1993).
57. De, A., Churchill, L., Obal, F. Jr., Simasko, S. M. & Krueger, J. M. GHRH and
IL1beta increase cytoplasmic Ca(2+) levels in cultured hypothalamic
GABAergic neurons. Brain Res. 949, 209212 (2002).
58. Kubota, T., Fang, J., Kushikata, T. & Krueger, J. M. Interleukin-13 and
transforming growth factor-beta1 inhibit spontaneous sleep in rabbits. Am. J.
Physiol. Regul. Integr. Comp. Physiol. 279, R786792 (2000).
59. Kubota, T., Fang, J., Guan, Z., Brown, R. A. & Krueger, J. M. Vagotomy
attenuates tumor necrosis factor-alpha-induced sleep and EEG delta-
activity in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 280, R12131220
(2001).
60. Curfs, J. H., Meis, J. F. & Hoogkamp-Korstanje, J. A. A primer on cytokines:
sources, receptors, effects, and inducers. Clin. Microbiol. Rev. 10, 742780
(1997).
61. Hodo, T. W., de Aquino, M. T. P., Shimamoto, A. & Shanker, A. Critical
neurotransmitters in the neuroimmune network. Front Immunol. 11, 1869
(2020).
62. Huang, X., Hussain, B. & Chang, J. Peripheral inammation and blood-brain
barrier disruption: effects and mechanisms. CNS Neurosci. Ther. 27,3647
(2021).
63. Grill, H. J. & Hayes, M. R. Hindbrain neurons as an essential hub in the
neuroanatomically distributed control of energy balance. Cell Metab. 16,
296309 (2012).
64. Kunz, N. & Kemper, C. Complement has brains-do intracellular complement
and immunometabolism cooperate in tissue homeostasis and behavior? Front.
Immunol. 12, 629986 (2021).
65. Levin, S. G. & Godukhin, O. V. Modulating effect of cytokines on mechanisms
of synaptic plasticity in the brain. Biochemistry (Mosc.) 82, 264274 (2017).
66. Wang, Y. et al. Astrocyte-secreted IL-33 mediates homeostatic synaptic
plasticity in the adult hippocampus. Proc. Natl Acad. Sci. USA 118,https://
doi.org/10.1073/pnas.2020810118 (2021).
67. Turnbull, A. V. & Rivier, C. Regulation of the HPA axis by cytokines. Brain
Behav. Immun. 9, 253275 (1995).
68. Besedovsky, H. O. & del Rey, A. The cytokine-HPA axis feed-back circuit. Z.
Rheumatol. 59(Suppl 2), II/2630 (2000).
69. Nguyen, J. T. et al. The neuron-specic interleukin-1 receptor accessory
protein alters emergent network state properties in vitro. Neurobiol. Sleep.
Circadian Rhythms 6,3543 (2019).
70. Krueger, J. M. & Obal, F. A neuronal group theory of sleep function. J. Sleep.
Res. 2,6369 (1993).
71. Rector, D. M., Topchiy, I. A., Carter, K. M. & Rojas, M. J. Local functional
state differences between rat cortical columns. Brain Res. 1047,4555
(2005).
72. Roy, S., Krueger, J. M., Rector, D. M. & Wan, Y. A network model for activity-
dependent sleep regulation. J. Theor. Biol. 253, 462468 (2008).
73. Krueger, J. M., Huang, Y. H., Rector, D. M. & Buysse, D. J. Sleep: a synchrony
of cell activity-driven small network states. Eur. J. Neurosci. 38, 21992209
(2013).
74. Krueger, J. M. Sleep and circadian rhythms: evolutionary entanglement and
local regulation. Neurobiol. Sleep. Circadian Rhythms 9, 100052 (2020).
75. Krueger, J. M., Nguyen, J. T., Dykstra-Aiello, C. J. & Taishi, P. Local sleep.
Sleep. Med. Rev. 43,1421 (2019).
REVIEW ARTICLE COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4
12 COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www.nature.com/commsbio
76. Taishi, P., Churchill, L., De, A., Obal, F. Jr. & Krueger, J. M. Cytokine mRNA
induction by interleukin-1beta or tumor necrosis factor alpha in vitro and
in vivo. Brain Res. 1226,8998 (2008).
77. Rockstrom, M. D. et al. Tumor necrosis factor alpha in sleep regulation. Sleep.
Med. Rev. 40,6978 (2018).
78. Krueger, J. M., Walter, J., Dinarello, C. A., Wolff, S. M. & Chedid, L. Sleep-
promoting effects of endogenous pyrogen (interleukin-1). Am. J. Physiol. 246,
R994999 (1984).
79. Imeri, L. & Opp, M. R. How (and why) the immune system makes us sleep.
Nat. Rev. Neurosci. 10, 199210 (2009).
80. Jewett, K. A. & Krueger, J. M. Humoral sleep regulation; interleukin-1 and
tumor necrosis factor. Vitam. Horm. 89, 241257 (2012).
81. Davis, C. J. et al. The neuron-specic interleukin-1 receptor accessory protein
is required for homeostatic sleep and sleep responses to inuenza viral
challenge in mice. Brain Behav. Immun. 47,3543 (2015).
82. Churchill, L. et al. Brain distribution of cytokine mRNA induced by systemic
administration of interleukin-1beta or tumor necrosis factor alpha. Brain Res.
1120,6473 (2006).
83. Taishi, P., Bredow, S., Guha-Thakurta, N., Obal, F. Jr. & Krueger, J. M.
Diurnal variations of interleukin-1 beta mRNA and beta-actin mRNA in rat
brain. J. Neuroimmunol. 75,6974 (1997).
84. Bredow, S., Guha-Thakurta, N., Taishi, P., Obal, F. Jr. & Krueger, J. M.
Diurnal variations of tumor necrosis factor alpha mRNA and alpha-tubulin
mRNA in rat brain. Neuroimmunomodulation 4,8490 (1997).
85. Floyd, R. A. & Krueger, J. M. Diurnal variation of TNF alpha in the rat brain.
Neuroreport 8, 915918 (1997).
86. Garlanda, C., Dinarello, C. A. & Mantovani, A. The interleukin-1 family: back
to the future. Immunity 39, 10031018 (2013).
87. Smith, D. E. et al. A central nervous system-restricted isoform of the
interleukin-1 receptor accessory protein modulates neuronal responses to
interleukin-1. Immunity 30, 817831 (2009).
88. Taishi, P. et al. Brain-specic interleukin-1 receptor accessory protein in sleep
regulation. J. Appl Physiol. (1985) 112, 10151022 (2012).
89. Oles, V. et al. Sleep- and time of day-linked RNA transcript expression in
wild-type and IL1 receptor accessory protein-null mice. J. Appl. Physiol. (1985)
128, 15061522 (2020).
90. Dykstra-Aiello, C. et al. A wake-like state in vitro induced by transmembrane
TNF/soluble TNF receptor reverse signaling. Brain Behav. Immun. 94,
245258 (2021).
91. Guan, Y. et al. Astrocytes constitute the major TNF-alpha-producing cell
population in the infarct cortex in dMCAO rats receiving intravenous MSC
infusion. Biomed. Pharmacother. 142, 111971 (2021).
92. Churchill, L. et al. Unilateral cortical application of tumor necrosis factor
alpha induces asymmetry in Fos- and interleukin-1beta-immunoreactive cells
within the corticothalamic projection. Brain Res. 1055,1524 (2005).
93. Jewett, K. A. et al. Tumor necrosis factor enhances the sleep-like state and
electrical stimulation induces a wake-like state in co-cultures of neurons and
glia. Eur. J. Neurosci. 42, 20782090 (2015).
94. Schmidt, M. H. The energy allocation function of sleep: a unifying theory of
sleep, torpor, and continuous wakefulness. Neurosci. Biobehav. Rev. 47,
122153 (2014).
95. Sharpley, A. L., Cooper, C. M., Williams, C., Godlewska, B. R. & Cowen, P. J.
Effects of typhoid vaccine on inammation and sleep in healthy participants: a
double-blind, placebo-controlled, crossover study. Psychopharmacology (Berl.)
233, 34293435 (2016).
96. Toth, L. A., Tolley, E. A. & Krueger, J. M. Sleep as a prognostic indicator
during infectious disease in rabbits. Proc. Soc. Exp. Biol. Med. 203, 179192
(1993).
97. Toth, L. A. & Krueger, J. M. Alteration of sleep in rabbits by Staphylococcus
aureus infection. Infect. Immun. 56, 17851791 (1988).
98. Seke Etet, P. F. et al. Sleep and rhythm changes at the time of Trypanosoma
brucei invasion of the brain parenchyma in the rat. Chronobiol. Int. 29,
469481 (2012).
99. Toth, L. A., Tolley, E. A., Broady, R., Blakely, B. & Krueger, J. M. Sleep during
experimental trypanosomiasis in rabbits. Proc. Soc. Exp. Biol. Med. 205,
174181 (1994).
100. Patke, A., Young, M. W. & Axelrod, S. Molecular mechanisms and
physiological importance of circadian rhythms. Nat. Rev. Mol. Cell Biol. 21,
6784 (2020).
101. Cavadini, G. et al. TNF-alpha suppresses the expression of clock genes by
interfering with E-box-mediated transcription. Proc. Natl Acad. Sci. USA 104,
1284312848 (2007).
102. Meier, D., Lopez, M., Franken, P. & Fontana, A. Twist1 is a TNF-inducible
inhibitor of clock mediated activation of period genes. PLoS ONE 10,
e0137229 (2015).
103. Marpegan, L., Bekinschtein, T. A., Costas, M. A. & Golombek, D. A. Circadian
responses to endotoxin treatment in mice. J. Neuroimmunol. 160, 102109
(2005).
104. Yamamura, Y., Yano, I., Kudo, T. & Shibata, S. Time-dependent inhibitory
effect of lipopolysaccharide injection on Per1 and Per2 gene expression in the
mouse heart and liver. Chronobiol. Int. 27, 213232 (2010).
105. Wang, Y. et al. Endotoxin disrupts circadian rhythms in macrophages via
reactive oxygen species. PLoS ONE 11, e0155075 (2016).
106. Friese, R. S., Bruns, B. & Sinton, C. M. Sleep deprivation after septic insult
increases mortality independent of age. J. Trauma 66,5054 (2009).
107. Everson, C. A. & Toth, L. A. Systemic bacterial invasion induced by sleep
deprivation. Am. J. Physiol. Regul. Integr. Comp. Physiol. 278, R905916
(2000).
108. Opp, M. R. Sleeping to fuel the immune system: mammalian sleep and
resistance to parasites. BMC Evol. Biol. 9, 8 (2009).
109. Prather, A. A., Janicki-Deverts, D., Hall, M. H. & Cohen, S. Behaviorally
assessed sleep and susceptibility to the common cold. Sleep 38, 13531359
(2015).
110. Fernandes, E. R. et al. Sleep disturbance during infection compromises Tfh
differentiation and impacts host immunity. iScience 23, 101599 (2020).
111. Chung, W. S., Lin, H. H. & Cheng, N. C. The incidence and risk of herpes
zoster in patients with sleep disorders: a population-based cohort study.
Medicine (Baltimore) 95, e2195 (2016).
112. Patel, S. R. et al. A prospective study of sleep duration and pneumonia risk in
women. Sleep 35,97101 (2012).
113. Prather, A. A. & Leung, C. W. Association of insufcient sleep with
respiratory infection among adults in the United States. JAMA Intern. Med.
176, 850852 (2016).
114. Cohen, S., Doyle, W. J., Alper, C. M., Janicki-Deverts, D. & Turner, R. B. Sleep
habits and susceptibility to the common cold. Arch. Intern. Med. 169,6267
(2009).
115. Orzech, K. M., Acebo, C., Seifer, R., Barker, D. & Carskadon, M. A. Sleep
patterns are associated with common illness in adolescents. J. Sleep. Res. 23,
133142 (2014).
116. Zhang, Y., Xie, B., Chen, X., Zhang, J. & Yuan, S. A key role of gut microbiota-
vagus nerve/spleen axis in sleep deprivation-mediated aggravation of systemic
inammation after LPS administration. Life Sci. 265, 118736 (2021).
117. Lange, T., Dimitrov, S., Bollinger, T., Diekelmann, S. & Born, J. Sleep
after vaccination boosts immunological memory. J. Immunol. 187, 283290
(2011).
118. Prather, A. A. et al. Sleep and antibody response to hepatitis B vaccination.
Sleep 35, 10631069 (2012).
119. Renegar, K. B., Floyd, R. A. & Krueger, J. M. Effects of short-term sleep
deprivation on murine immunity to inuenza virus in young adult and
senescent mice. Sleep 21, 241248 (1998).
120. Hurley, S., Goldberg, D., Bernstein, L. & Reynolds, P. Sleep duration and
cancer risk in women. Cancer Causes Control 26, 10371045 (2015).
121. Kakizaki, M. et al. Sleep duration and the risk of prostate cancer: the Ohsaki
Cohort Study. Br. J. Cancer 99, 176178 (2008).
122. Jiao, L. et al. Sleep duration and incidence of colorectal cancer in
postmenopausal women. Br. J. Cancer 108, 213221 (2013).
123. Cao, J. et al. Sleep duration and risk of breast cancer: The JACC Study. Breast
Cancer Res. Treat. 174, 219225 (2019).
124. Wu, A. H. et al. Sleep duration, melatonin and breast cancer among Chinese
women in Singapore. Carcinogenesis 29, 12441248 (2008).
125. Wehr, T. A. The durations of human melatonin secretion and sleep respond to
changes in daylength (photoperiod). J. Clin. Endocrinol. Metab. 73, 12761280
(1991).
126. Luo, J. et al. Effect of melatonin on T/B cell activation and immune regulation
in pinealectomy mice. Life Sci. 242, 117191 (2020).
127. De Lorenzo, B. H., de Oliveira Marchioro, L., Greco, C. R. & Suchecki, D.
Sleep-deprivation reduces NK cell number and function mediated by beta-
adrenergic signalling. Psychoneuroendocrinology 57, 134143 (2015).
128. De Lorenzo, B. H. P. et al. Chronic sleep restriction impairs the antitumor
immune response in mice. Neuroimmunomodulation 25,5967 (2018).
129. Maragno-Correa, J. M. et al. Sleep deprivation increases mortality in female
mice bearing Ehrlich ascitic tumor. Neuroimmunomodulation 20, 134140
(2013).
130. Barbosa Vieira, T. K. et al. Correlation between circadian rhythm related
genes, type 2 diabetes, and cancer: insights from metanalysis of
transcriptomics data. Mol. Cell Endocrinol. 526, 111214 (2021).
131. Gonzalez, H., Hagerling, C. & Werb, Z. Roles of the immune system in cancer:
from tumor initiation to metastatic progression. Genes Dev. 32, 12671284
(2018).
132. Palma, J. A., Urrestarazu, E. & Iriarte, J. Sleep loss as risk factor for neurologic
disorders: a review. Sleep. Med. 14, 229236 (2013).
133. Sabia, S. et al. Association of sleep duration in middle and old age with
incidence of dementia. Nat. Commun. 12, 2289 (2021).
134. Sadeghmousavi, S., Eskian, M., Rahmani, F. & Rezaei, N. The effect of
insomnia on development of Alzheimers disease. J. Neuroinammation 17,
289 (2020).
COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4 REVIEW ARTICLE
COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www .nature.com/commsbio 13
135. Stephenson, J., Nutma, E., van der Valk, P. & Amor, S. Inammation in CNS
neurodegenerative diseases. Immunology 154, 204219 (2018).
136. Manchanda, S., Singh, H., Kaur, T. & Kaur, G. Low-grade neuroinammation
due to chronic sleep deprivation results in anxiety and learning and memory
impairments. Mol. Cell Biochem. 449,6372 (2018).
137. Xue, R. et al. Nicotinic mitigation of neuroinammation and oxidative stress
after chronic sleep deprivation. Front. Immunol. 10, 2546 (2019).
138. Hurtado-Alvarado, G. et al. The yin/yang of inammatory status: blood-brain
barrier regulation during sleep. Brain Behav. Immun. 69, 154166 (2018).
139. Korin, B. et al. Short-term sleep deprivation in mice induces B cell migration
to the brain compartment. Sleep 43,https://doi.org/10.1093/sleep/zsz222
(2020).
140. Luo, F. et al. Melatonin and autophagy in aging-related neurodegenerative
diseases. Int. J. Mol. Sci. 21,https://doi.org/10.3390/ijms21197174 (2020).
141. Finkbeiner, E., Haindl, M., Raman, N. & Muller, S. SUMO routes ribosome
maturation. Nucleus 2, 527532 (2011).
142. Zhao, H. Y. et al. Chronic sleep restriction induces cognitive decits and
cortical beta-amyloid deposition in mice via BACE1-antisense activation. CNS
Neurosci. Ther. 23, 233240 (2017).
143. Liu, P. et al. Activation of inammation is associated with amyloid-beta
accumulation induced by chronic sleep restriction in rats. J. Alzheimers Dis.
74, 759773 (2020).
144. Shokri-Kojori, E. et al. beta-Amyloid accumulation in the human brain after
one night of sleep deprivation. Proc. Natl Acad. Sci. USA 115, 44834488
(2018).
145. Spira, A. P. et al. Self-reported sleep and beta-amyloid deposition in
community-dwelling older adults. JAMA Neurol. 70, 15371543 (2013).
146. Ju, Y. S. et al. Slow wave sleep disruption increases cerebrospinal uid
amyloid-beta levels. Brain 140, 21042111 (2017).
147. Chen, D. W., Wang, J., Zhang, L. L., Wang, Y. J. & Gao, C. Y. Cerebrospinal
uid amyloid-beta levels are increased in patients with insomnia. J. Alzheimers
Dis. 61, 645651 (2018).
148. Ooms, S. et al. Effect of 1 night of total sleep deprivation on cerebrospinal uid
beta-amyloid 42 in healthy middle-aged men: a randomized clinical trial.
JAMA Neurol. 71, 971977 (2014).
149. Zhao, B. et al. Chronic sleep restriction induces abeta accumulation by
disrupting the balance of abeta production and clearance in rats. Neurochem.
Res. 44, 859873 (2019).
150. Wu, H., Dunnett, S., Ho, Y. S. & Chang, R. C. The role of sleep deprivation
and circadian rhythm disruption as risk factors of Alzheimers disease. Front.
Neuroendocrinol. 54, 100764 (2019).
151. Jaeger, L. B. et al. Lipopolysaccharide alters the blood-brain barrier transport
of amyloid beta protein: a mechanism for inammation in the progression of
Alzheimers disease. Brain Behav. Immun. 23, 507517 (2009).
152. Sastre, M. et al. Nonsteroidal anti-inammatory drugs and peroxisome
proliferator-activated receptor-gamma agonists modulate immunostimulated
processing of amyloid precursor protein through regulation of beta-secretase.
J. Neurosci. 23, 97969804 (2003).
153. Mouton-Liger, F. et al. Oxidative stress increases BACE1 protein levels
through activation of the PKR-eIF2alpha pathway. Biochim. Biophys. Acta
1822, 885896 (2012).
154. Giese, M. et al. BDNF: an indicator of insomnia? Mol. Psychiatry 19, 151152
(2014).
155. Tan, D. X., Manchester, L. C., Terron, M. P., Flores, L. J. & Reiter, R. J. One
molecule, many derivatives: a never-ending interaction of melatonin with
reactive oxygen and nitrogen species? J. Pineal Res. 42,2842 (2007).
156. Zhang, L. et al. Melatonin ameliorates cognitive impairment induced by sleep
deprivation in rats: role of oxidative stress, BDNF and CaMKII. Behav. Brain
Res. 256,7281 (2013).
157. Fredrich, M., Hampel, M., Seidel, K., Christ, E. & Korf, H. W. Impact of
melatonin receptor-signaling on Zeitgeber time-dependent changes in cell
proliferation and apoptosis in the adult murine. Hippocampus 27, 495506
(2017).
158. Alghamdi, B. S. The neuroprotective role of melatonin in neurological
disorders. J. Neurosci. Res. 96, 11361149 (2018).
159. Luengo, E. et al. Pharmacological doses of melatonin impede cognitive decline
in tau-related Alzheimer models, once tauopathy is initiated, by restoring the
autophagic ux. J. Pineal Res. 67, e12578 (2019).
160. Emamian, F. et al. The association between obstructive sleep apnea and
Alzheimers disease: a meta-analysis perspective. Front. Aging Neurosci. 8,78
(2016).
161. Osorio, R. S. et al. Sleep-disordered breathing advances cognitive decline in the
elderly. Neurology 84, 19641971 (2015).
162. Frenkel, D. et al. Scara1 deciency impairs clearance of soluble amyloid-beta
by mononuclear phagocytes and accelerates Alzheimers-like disease
progression. Nat. Commun. 4, 2030 (2013).
163. Musiek, E. S. & Holtzman, D. M. Mechanisms linking circadian clocks, sleep,
and neurodegeneration. Science 354, 10041008 (2016).
164. Wang, J., Gu, B. J., Masters, C. L. & Wang, Y. J. A systemic view of Alzheimer
disease - insights from amyloid-beta metabolism beyond the brain. Nat. Rev.
Neurol. 13, 703 (2017).
165. Tanner, C. M. & Aston, D. A. Epidemiology of Parkinsons disease and
akinetic syndromes. Curr. Opin. Neurol. 13, 427430 (2000).
166. Barber, A. & Dashtipour, K. Sleep disturbances in Parkinsons disease with
emphasis on rapid eye movement sleep behavior disorder. Int. J. Neurosci.
122, 407412 (2012).
167. Barnett, R. Parkinsons disease. Lancet 387, 217 (2016).
168. Hsiao, Y. H. et al. Sleep disorders and an increased risk of Parkinsons disease
in individuals with non-apnea sleep disorders: a population-based cohort
study. J. Sleep. Res. 26, 623628 (2017).
169. Bohnen, N. I. & Hu, M. T. M. Sleep disturbance as potential risk and
progression factor for Parkinsons disease. J. Parkinsons Dis. 9, 603614
(2019).
170. Su, L. Y. et al. Melatonin attenuates MPTP-induced neurotoxicity via
preventing CDK5-mediated autophagy and SNCA/alpha-synuclein
aggregation. Autophagy 11, 17451759 (2015).
171. Srivastava, A. K., Roy Choudhury, S. & Karmakar, S. Melatonin/polydopamine
nanostructures for collective neuroprotection-based Parkinsons disease
therapy. Biomater. Sci. 8, 13451363 (2020).
172. Abad, V. C., Sarinas, P. S. & Guilleminault, C. Sleep and rheumatologic
disorders. Sleep. Med. Rev. 12, 211228 (2008).
173. Hsiao, Y. H. et al. Sleep disorders and increased risk of autoimmune diseases
in individuals without sleep apnea. Sleep 38, 581586 (2015).
174. Young, K. A. et al. Less than 7 hours of sleep per night is associated
with transitioning to systemic lupus erythematosus. Lupus 27, 15241531
(2018).
175. Palma, B. D., Gabriel, A. Jr., Colugnati, F. A. & Tuk, S. Effects of sleep
deprivation on the development of autoimmune disease in an experimental
model of systemic lupus erythematosus. Am. J. Physiol. Regul. Integr. Comp.
Physiol. 291, R15271532 (2006).
176. Alarcon-Riquelme, M. E., Moller, G. & Fernandez, C. Age-dependent
responsiveness to interleukin-6 in B lymphocytes from a systemic lupus
erythematosus-prone (NZB x NZW)F1 hybrid. Clin. Immunol.
Immunopathol. 62, 264269 (1992).
177. Yehuda, S., Sredni, B., Carasso, R. L. & Kenigsbuch-Sredni, D. REM sleep
deprivation in rats results in inammation and interleukin-17 elevation. J.
Interferon Cytokine Res. 29, 393398 (2009).
178. Acosta-Rodriguez, E. V., Napolitani, G., Lanzavecchia, A. & Sallusto, F.
Interleukins 1beta and 6 but not transforming growth factor-beta are essential
for the differentiation of interleukin 17-producing human T helper cells. Nat.
Immunol. 8, 942949 (2007).
179. Volin, M. V. & Shahrara, S. Role of TH-17 cells in rheumatic and other
autoimmune diseases. Rheumatology (Sunnyvale) 1,https://doi.org/10.4172/
2161-1149.1000104 (2011).
180. Bollinger, T. et al. Sleep-dependent activity of T cells and regulatory T cells.
Clin. Exp. Immunol. 155, 231238 (2009).
181. Carbone, F. et al. Regulatory T cell proliferative potential is impaired in
human autoimmune disease. Nat. Med. 20,6974 (2014).
182. Leng, Y. et al. Sleep duration and risk of fatal and nonfatal stroke: a
prospective study and meta-analysis. Neurology 84, 10721079 (2015).
183. Ferrie, J. E. et al. Associations between change in sleep duration and
inammation: ndings on C-reactive protein and interleukin 6 in the
Whitehall II Study. Am. J. Epidemiol. 178, 956961 (2013).
184. Wang, Y. H. et al. Association of longitudinal patterns of habitual sleep
duration with risk of cardiovascular events and all-cause mortality. JAMA
Netw. Open 3, e205246 (2020).
185. Dominguez, F. et al. Association of sleep duration and quality with subclinical
atherosclerosis. J. Am. Coll. Cardiol. 73, 134144 (2019).
186. Cappuccio, F. P. et al. Meta-analysis of short sleep duration and obesity in
children and adults. Sleep 31, 619626 (2008).
187. Meng, L., Zheng, Y. & Hui, R. The relationship of sleep duration and insomnia
to risk of hypertension incidence: a meta-analysis of prospective cohort
studies. Hypertens. Res. 36, 985995 (2013).
188. Anothaisintawee, T., Reutrakul, S., Van Cauter, E. & Thakkinstian, A. Sleep
disturbances compared to traditional risk factors for diabetes development:
systematic review and meta-analysis. Sleep. Med. Rev. 30,1124 (2016).
189. Zhong, X. et al. Increased sympathetic and decreased parasympathetic
cardiovascular modulation in normal humans with acute sleep deprivation. J.
Appl. Physiol. (1985) 98, 20242032 (2005).
190. Meier-Ewert, H. K. et al. Effect of sleep loss on C-reactive protein, an
inammatory marker of cardiovascular risk. J. Am. Coll. Cardiol. 43, 678683
(2004).
191. Sauvet, F. et al. Effect of acute sleep deprivation on vascular function in
healthy subjects. J. Appl. Physiol. (1985) 108,6875 (2010).
192. Dettoni, J. L. et al. Cardiovascular effects of partial sleep deprivation in healthy
volunteers. J. Appl. Physiol. (1985) 113, 232236 (2012).
REVIEW ARTICLE COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4
14 COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www.nature.com/commsbio
193. Spiegel, K., Tasali, E., Leproult, R. & Van Cauter, E. Effects of poor and short
sleep on glucose metabolism and obesity risk. Nat. Rev. Endocrinol. 5, 253261
(2009).
194. Buxton, O. M. et al. Adverse metabolic consequences in humans of prolonged
sleep restriction combined with circadian disruption. Sci. Transl. Med. 4,
129ra143 (2012).
195. Broussard, J. L., Ehrmann, D. A., Van Cauter, E., Tasali, E. & Brady, M. J.
Impaired insulin signaling in human adipocytes after experimental sleep
restriction: a randomized, crossover study. Ann. Intern. Med. 157, 549557
(2012).
196. Nedeltcheva, A. V., Kilkus, J. M., Imperial, J., Schoeller, D. A. & Penev, P. D.
Insufcient sleep undermines dietary efforts to reduce adiposity. Ann. Intern.
Med. 153, 435441 (2010).
197. Darukhanavala, A. et al. Changes in insulin secretion and action in adults with
familial risk for type 2 diabetes who curtail their sleep. Diabetes Care 34,
22592264 (2011).
198. Esser, N., Legrand-Poels, S., Piette, J., Scheen, A. J. & Paquot, N. Inammation
as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes
Res. Clin. Pract. 105, 141150 (2014).
199. Longo, M. et al. Adipose tissue dysfunction as determinant of obesity-
associated metabolic complications. Int. J. Mol. Sci. 20,https://doi.org/
10.3390/ijms20092358 (2019).
200. Freitas Lima, L. C. et al. Adipokines, diabetes and atherosclerosis: an
inammatory association. Front. Physiol. 6, 304 (2015).
201. Geovanini, G. R. & Libby, P. Atherosclerosis and inammation: overview and
updates. Clin. Sci. (Lond.) 132, 12431252 (2018).
202. Yousuf, O. et al. High-sensitivity C-reactive protein and cardiovascular
disease: a resolute belief or an elusive link? J. Am. Coll. Cardiol. 62, 397408
(2013).
203. van Leeuwen, W. M. et al. Sleep restriction increases the risk of developing
cardiovascular diseases by augmenting proinammatory responses through
IL-17 and CRP. PLoS ONE 4, e4589 (2009).
204. Irwin, M. R., Olmstead, R. & Carroll, J. E. Sleep disturbance, sleep duration,
and inammation: a systematic review and meta-analysis of cohort studies
and experimental sleep deprivation. Biol. Psychiatry 80,4052 (2016).
205. McAlpine, C. S. et al. Sleep modulates haematopoiesis and protects against
atherosclerosis. Nature 566, 383387 (2019).
206. Dutta, P. et al. Myocardial infarction accelerates atherosclerosis. Nature 487,
325329 (2012).
207. Kadoya, M. & Koyama, H. Sleep, autonomic Nnervous function and
atherosclerosis. Int. J. Mol. Sci. 20,https://doi.org/10.3390/ijms20040794
(2019).
208. Yang, X. et al. Oxidative stress-mediated atherosclerosis: mechanisms and
therapies. Front. Physiol. 8, 600 (2017).
209. Li, H. Y., Leu, Y. L., Wu, Y. C. & Wang, S. H. Melatonin inhibits in vitro
smooth muscle cell inammation and proliferation and atherosclerosis
in apolipoprotein E-decient mice. J. Agric. Food Chem. 67, 18891901
(2019).
210. Born, J., Lange, T., Hansen, K., Molle, M. & Fehm, H. L. Effects of sleep and
circadian rhythm on human circulating immune cells. J. Immunol. 158,
44544464 (1997).
211. Dimitrov, S., Lange, T., Nohroudi, K. & Born, J. Number and function of
circulating human antigen presenting cells regulated by sleep. Sleep 30,
401411 (2007).
212. Lasselin, J., Rehman, J. U., Akerstedt, T., Lekander, M. & Axelsson, J. Effect of
long-term sleep restriction and subsequent recovery sleep on the diurnal
rhythms of white blood cell subpopulations. Brain Behav. Immun. 47,9399
(2015).
213. Said, E. A. et al. Sleep deprivation alters neutrophil functions and levels of
Th1-related chemokines and CD4(+) T cells in the blood. Sleep. Breath. 23,
13311339 (2019).
214. Dinges, D. F. et al. Leukocytosis and natural killer cell function parallel
neurobehavioral fatigue induced by 64 hours of sleep deprivation. J. Clin.
Invest. 93, 19301939 (1994).
215. Scheiermann, C., Kunisaki, Y. & Frenette, P. S. Circadian control of the
immune system. Nat. Rev. Immunol. 13, 190198 (2013).
216. Redwine, L., Dang, J. & Irwin, M. Cellular adhesion molecule expression,
nocturnal sleep, and partial night sleep deprivation. Brain Behav. Immun. 18,
333340 (2004).
217. Axelsson, J. et al. Effects of sustained sleep restriction on mitogen-stimulated
cytokines, chemokines and T helper 1/ T helper 2 balance in humans. PLoS
ONE 8, e82291 (2013).
218. Shearer, W. T. et al. Soluble TNF-alpha receptor 1 and IL-6 plasma levels in
humans subjected to the sleep deprivation model of spaceight. J. Allergy Clin.
Immunol. 107, 165170 (2001).
219. Hu, J. et al. Sleep-deprived mice show altered cytokine production manifest by
perturbations in serum IL-1ra, TNFa, and IL-6 levels. Brain Behav. Immun.
17, 498504 (2003).
220. Vgontzas, A. N. et al. Adverse effects of modest sleep restriction on sleepiness,
performance, and inammatory cytokines. J. Clin. Endocrinol. Metab. 89,
21192126 (2004).
221. Miller, M. A. et al. Gender differences in the cross-sectional relationships
between sleep duration and markers of inammation: Whitehall II study. Sleep
32, 857864 (2009).
222. Patel, S. R. et al. Sleep duration and biomarkers of inammation. Sleep 32,
200204 (2009).
223. Perez de Heredia, F. et al. Self-reported sleep duration, white blood cell counts
and cytokine proles in European adolescents: the HELENA study. Sleep.
Med. 15, 12511258 (2014).
224. Bakour, C. et al. Sleep duration trajectories and systemic inammation in
young adults: results from the National Longitudinal Study of Adolescent to
Adult Health (Add Health). Sleep 40,https://doi.org/10.1093/sleep/zsx156
(2017).
225. Richardson, M. R. & Churilla, J. R. Sleep duration and C-reactive protein in
US adults. South Med. J. 110, 314317 (2017).
226. Fondell, E. et al. Short natural sleep is associated with higher T cell and lower
NK cell activities. Brain Behav. Immun. 25, 13671375 (2011).
227. Carroll, J. E. et al. Epigenetic aging and immune senescence in women with
insomnia symptoms: ndings from the Womens Health Initiative Study. Biol.
Psychiatry 81, 136144 (2017).
228. Jackowska, M. et al. Short sleep duration is associated with shorter telomere
length in healthy men: ndings from the Whitehall II cohort study. PLoS ONE
7, e47292 (2012).
229. James, S. et al. Sleep duration and telomere length in children. J. Pediatr. 187,
247252 e241 (2017).
230. Hall, M. H. et al. Association between sleep duration and mortality is
mediated by markers of inammation and health in older adults: the Health,
Aging and Body Composition Study. Sleep 38, 189195 (2015).
231. Li, Y. et al. Hs-CRP and all-cause, cardiovascular, and cancer mortality risk: a
meta-analysis. Atherosclerosis 259,7582 (2017).
232. Uthgenannt, D., Schoolmann, D., Pietrowsky, R., Fehm, H. L. & Born, J.
Effects of sleep on the production of cytokines in humans. Psychosom. Med.
57,97104 (1995).
233. Irwin, M. R., Wang, M., Campomayor, C. O., Collado-Hidalgo, A. & Cole, S.
Sleep deprivation and activation of morning levels of cellular and genomic
markers of inammation. Arch. Intern. Med. 166, 17561762 (2006).
234. Simpson, N. S. et al. Repeating patterns of sleep restriction and recovery: do
we get used to it? Brain Behav. Immun. 58, 142151 (2016).
235. Dimitrov, S., Besedovsky, L., Born, J. & Lange, T. Differential acute effects of
sleep on spontaneous and stimulated production of tumor necrosis factor in
men. Brain Behav. Immun. 47, 201210 (2015).
236. Dimitrov, S., Lange, T., Tieken, S., Fehm, H. L. & Born, J. Sleep associated
regulation of T helper 1/T helper 2 cytokine balance in humans. Brain Behav.
Immun. 18, 341348 (2004).
237. Ginaldi, L. et al. The immune system in the elderly: I. Specic humoral
immunity. Immunol. Res 20, 101108 (1999).
238. Redwine, L., Dang, J., Hall, M. & Irwin, M. Disordered sleep, nocturnal
cytokines, and immunity in alcoholics. Psychosom. Med. 65,7585 (2003).
239. Sakami, S. et al. Coemergence of insomnia and a shift in the Th1/Th2 balance
toward Th2 dominance. Neuroimmunomodulation 10, 337343 (2002).
240. Moser, E. K., Field, N. S. & Oliver, P. M. Aberrant Th2 inammation drives
dysfunction of alveolar macrophages and susceptibility to bacterial
pneumonia. Cell Mol. Immunol. 15, 480492 (2018).
241. Disis, M. L. Immune regulation of cancer. J. Clin. Oncol. 28, 45314538
(2010).
242. Lee, H. L. et al. Inammatory cytokines and change of Th1/Th2 balance as
prognostic indicators for hepatocellular carcinoma in patients treated with
transarterial chemoembolization. Sci. Rep. 9, 3260 (2019).
243. Villafuerte, G. et al. Sleep deprivation and oxidative stress in animal models: a
systematic review. Oxid. Med. Cell Longev. 2015, 234952 (2015).
244. Teixeira, K. R. C. et al. Night workers have lower levels of antioxidant defenses
and higher levels of oxidative stress damage when compared to day workers.
Sci. Rep. 9, 4455 (2019).
245. Trivedi, M. S., Holger, D., Bui, A. T., Craddock, T. J. A. & Tartar, J. L. Short-
term sleep deprivation leads to decreased systemic redox metabolites and
altered epigenetic status. PLoS ONE 12, e0181978 (2017).
246. Chen, X., Song, M., Zhang, B. & Zhang, Y. Reactive oxygen species regulate T
cell immune response in the tumor microenvironment. Oxid. Med. Cell
Longev. 2016, 1580967 (2016).
247. Liguori, I. et al. Oxidative stress, aging, and diseases. Clin. Inter. Aging 13,
757772 (2018).
248. Irwin, M., Thompson, J., Miller, C., Gillin, J. C. & Ziegler, M. Effects of sleep
and sleep deprivation on catecholamine and interleukin-2 levels in humans:
clinical implications. J. Clin. Endocrinol. Metab. 84, 19791985 (1999).
249. Nance, D. M. & Sanders, V. M. Autonomic innervation and regulation of the
immune system (1987-2007). Brain Behav. Immun. 21, 736745 (2007).
COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4 REVIEW ARTICLE
COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www .nature.com/commsbio 15
250. Barnes, M. A., Carson, M. J. & Nair, M. G. Non-traditional cytokines: How
catecholamines and adipokines inuence macrophages in immunity,
metabolism and the central nervous system. Cytokine 72, 210219 (2015).
251. Grebe, K. M. et al. Cutting edge: sympathetic nervous system increases
proinammatory cytokines and exacerbates inuenza A virus pathogenesis. J.
Immunol. 184, 540544 (2010).
252. Collado-Hidalgo, A., Sung, C. & Cole, S. Adrenergic inhibition of innate anti-
viral response: PKA blockade of Type I interferon gene transcription mediates
catecholamine support for HIV-1 replication. Brain Behav. Immun. 20,
552563 (2006).
253. Kalinichenko, V. V., Mokyr, M. B., Graf, L. H. Jr., Cohen, R. L. & Chambers,
D. A. Norepinephrine-mediated inhibition of antitumor cytotoxic T
lymphocyte generation involves a beta-adrenergic receptor mechanism and
decreased TNF-alpha gene expression. J. Immunol. 163, 24922499 (1999).
254. Irwin, M. R. & Cole, S. W. Reciprocal regulation of the neural and innate
immune systems. Nat. Rev. Immunol. 11, 625632 (2011).
255. Mishra, I. et al. Chemical sympathectomy reduces peripheral inammatory
responses to acute and chronic sleep fragmentation. Am. J. Physiol. Regul.
Integr. Comp. Physiol. 318, R781R789 (2020).
256. Wheeler, N. D., Ensminger, D. C., Rowe, M. M., Wriedt, Z. S. & Ashley, N. T.
Alpha- and beta- adrenergic receptors regulate inammatory responses to
acute and chronic sleep fragmentation in mice. PeerJ 9, e11616 (2021).
257. Cirelli, C., Gutierrez, C. M. & Tononi, G. Extensive and divergent effects of
sleep and wakefulness on brain gene expression. Neuron 41,3543 (2004).
258. Mackiewicz, M., Zimmerman, J. E., Shockley, K. R., Churchill, G. A. & Pack,
A. I. What are microarrays teaching us about sleep? Trends Mol. Med. 15,
7987 (2009).
259. Barclay, J. L. et al. Circadian desynchrony promotes metabolic disruption in a
mouse model of shiftwork. PLoS ONE 7, e37150 (2012).
260. Aho, V. et al. Partial sleep restriction activates immune response-related gene
expression pathways: experimental and epidemiological studies in humans.
PLoS ONE 8, e77184 (2013).
261. Moller-Levet, C. S. et al. Effects of insufcient sleep on circadian rhythmicity
and expression amplitude of the human blood transcriptome. Proc. Natl Acad.
Sci. USA 110, E11321141 (2013).
262. Irwin, M. R., Witarama, T., Caudill, M., Olmstead, R. & Breen, E. C. Sleep loss
activates cellular inammation and signal transducer and activator of
transcription (STAT) family proteins in humans. Brain Behav. Immun. 47,
8692 (2015).
263. Archer, S. N. et al. Mistimed sleep disrupts circadian regulation of the human
transcriptome. Proc. Natl Acad. Sci. USA 111, E682691 (2014).
264. Mure, L. S. et al. Diurnal transcriptome atlas of a primate across major neural
and peripheral tissues. Science 359,https://doi.org/10.1126/science.aao0318
(2018).
265. Sulli, G., Manoogian, E. N. C., Taub, P. R. & Panda, S. Training the circadian
clock, clocking the drugs, and drugging the clock to prevent, manage, and treat
chronic diseases. Trends Pharm. Sci. 39, 812827 (2018).
266. Christou, S. et al. Circadian regulation in human white adipose tissue revealed
by transcriptome and metabolic network analysis. Sci. Rep. 9, 2641 (2019).
267. Chen, Z., Gardi, J., Kushikata, T., Fang, J. & Krueger, J. M. Nuclear factor-
kappaB-like activity increases in murine cerebral cortex after sleep
deprivation. Am. J. Physiol. 276, R18121818 (1999).
268. Williams, J. A., Sathyanarayanan, S., Hendricks, J. C. & Sehgal, A. Interaction
between sleep and the immune response in Drosophila: a role for the
NFkappaB relish. Sleep 30, 389400 (2007).
269. Basheer, R., Rainnie, D. G., Porkka-Heiskanen, T., Ramesh, V. & McCarley,
R. W. Adenosine, prolonged wakefulness, and A1-activated NF-kappaB
DNA binding in the basal forebrain of the rat. Neuroscience 104, 731739
(2001).
270. Brandt, J. A. et al. Sleep deprivation increases the activation of nuclear factor
kappa B in lateral hypothalamic cells. Brain Res. 1004,9197 (2004).
271. Ramesh, V., Thatte, H. S., McCarley, R. W. & Basheer, R. Adenosine and sleep
deprivation promote NF-kappaB nuclear translocation in cholinergic basal
forebrain. J. Neurochem. 100, 13511363 (2007).
272. Irwin, M. R. et al. Sleep loss activates cellular inammatory signaling. Biol.
Psychiatry 64, 538540 (2008).
273. Madonna, R. & De Caterina, R. Relevance of new drug discovery to reduce
NF-kappaB activation in cardiovascular disease. Vasc. Pharm. 57,4147
(2012).
274. Bellet, M. M., Zocchi, L. & Sassone-Corsi, P. The RelB subunit of NFkappaB
acts as a negative regulator of circadian gene expression. Cell Cycle 11,
33043311 (2012).
275. Kuo, T. H., Pike, D. H., Beizaeipour, Z. & Williams, J. A. Sleep triggered by an
immune response in Drosophila is regulated by the circadian clock and
requires the NFkappaB Relish. BMC Neurosci. 11, 17 (2010).
276. Negi, G., Kumar, A. & Sharma, S. S. Melatonin modulates neuroinammation
and oxidative stress in experimental diabetic neuropathy: effects on NF-
kappaB and Nrf2 cascades. J. Pineal Res. 50, 124131 (2011).
277. Narasimamurthy, R. et al. Circadian clock protein cryptochrome regulates the
expression of proinammatory cytokines. Proc. Natl Acad. Sci. USA 109,
1266212667 (2012).
278. Kolmus, K., Tavernier, J. & Gerlo, S. beta2-Adrenergic receptors in immunity and
inammation: stressing NF-kappaB. Brain Behav. Immun. 45,297310 (2015).
279. Benedict, C. et al. Gut microbiota and glucometabolic alterations in response
to recurrent partial sleep deprivation in normal-weight young individuals.
Mol. Metab. 5, 11751186 (2016).
280. Poroyko, V. A. et al. Chronic sleep disruption alters gut mmicrobiota, induces
systemic and adipose tissue inammation and insulin resistance in mice. Sci.
Rep. 6, 35405 (2016).
281. Gao, T. et al. Role of melatonin in sleep deprivation-induced intestinal barrier
dysfunction in mice. J. Pineal Res. 67, e12574 (2019).
282. Belkaid, Y. & Hand, T. W. Role of the microbiota in immunity and
inammation. Cell 157, 121141 (2014).
283. Belenky, G. et al. Patterns of performance degradation and restoration during
sleep restriction and subsequent recovery: a sleep dose-response study. J. Sleep.
Res. 12,112 (2003).
284. Rupp, T. L., Wesensten, N. J., Bliese, P. D. & Balkin, T. J. Banking sleep:
realization of benets during subsequent sleep restriction and recovery. Sleep
32, 311321 (2009).
285. Banks, S., Van Dongen, H. P., Maislin, G. & Dinges, D. F. Neurobehavioral
dynamics following chronic sleep restriction: dose-response effects of one
night for recovery. Sleep 33, 10131026 (2010).
286. Haack, M. et al. Increasing sleep duration to lower beat-to-beat blood
pressure: a pilot study. J. Sleep. Res. 22, 295304 (2013).
287. Chennaoui, M. et al. Leukocyte expression of type 1 and type 2 purinergic
receptors and pro-inammatory cytokines during total sleep deprivation and/
or sleep extension in healthy subjects. Front Neurosci. 11, 240 (2017).
288. Swinbourne, R., Miller, J., Smart, D., Dulson, D. K. & Gill, N. The effects of
sleep extension on sleep, performance, immunity and physical stress in rugby
players. Sports (Basel) 6,https://doi.org/10.3390/sports6020042 (2018).
289. Faraut, B. et al. Benets of napping and an extended duration of recovery sleep
on alertness and immune cells after acute sleep restriction. Brain Behav.
Immun. 25,1624 (2011).
290. Takahashi, M. & Arito, H. Maintenance of alertness and performance by a
brief nap after lunch under prior sleep decit. Sleep 23, 813819 (2000).
291. Milner, C. E. & Cote, K. A. Benets of napping in healthy adults: impact of
nap length, time of day, age, and experience with napping. J. Sleep. Res. 18,
272281 (2009).
292. Keramidas, M. E., Siebenmann, C., Norrbrand, L., Gadefors, M. & Eiken, O. A
brief pre-exercise nap may alleviate physical performance impairments
induced by short-term sustained operations with partial sleep deprivation - A
eld-based study. Chronobiol. Int. 35, 14641470 (2018).
293. Leng, Y. et al. Daytime napping, sleep duration and serum C reactive protein:
a population-based cohort study. BMJ Open 4, e006071 (2014).
294. Faraut, B. et al. Napping reverses the salivary interleukin-6 and urinary
norepinephrine changes induced by sleep restriction. J. Clin. Endocrinol.
Metab. 100, E416426 (2015).
295. Vgontzas, A. N. et al. Daytime napping after a night of sleep loss decreases
sleepiness, improves performance, and causes benecial changes in cortisol
and interleukin-6 secretion. Am. J. Physiol. Endocrinol. Metab. 292, E253261
(2007).
296. Zhong, G., Wang, Y., Tao, T., Ying, J. & Zhao, Y. Daytime napping and
mortality from all causes, cardiovascular disease, and cancer: a meta-analysis
of prospective cohort studies. Sleep. Med. 16, 811819 (2015).
297. Naska, A., Oikonomou, E., Trichopoulou, A., Psaltopoulou, T. &
Trichopoulos, D. Siesta in healthy adults and coronary mortality in the general
population. Arch. Intern. Med. 167, 296301 (2007).
298. Hausler, N., Haba-Rubio, J., Heinzer, R. & Marques-Vidal, P. Association of
napping with incident cardiovascular events in a prospective cohort study.
Heart 105, 17931798 (2019).
299. Yamada, T., Hara, K., Shojima, N., Yamauchi, T. & Kadowaki, T. Daytime
napping and the risk of cardiovascular disease and all-cause mortality: a
prospective study and dose-response meta-analysis. Sleep 38, 19451953
(2015).
300. Yamada, T., Shojima, N., Yamauchi, T. & Kadowaki, T. J-curve relation
between daytime nap duration and type 2 diabetes or metabolic syndrome: A
dose-response meta-analysis. Sci. Rep. 6, 38075 (2016).
301. Mohammad, Y. Siesta and risk for ischemic stroke: results from a case-control
study. Medicina (Kaunas) 56,https://doi.org/10.3390/medicina56050222
(2020).
302. Irwin, M. R. et al. Cognitive behavioral therapy vs. Tai Chi for late life
insomnia and inammatory risk: a randomized controlled comparative
efcacy trial. Sleep 37, 15431552 (2014).
303. Irwin, M. R. et al. Cognitive behavioral therapy and tai chi reverse cellular and
genomic markers of inammation in late-life insomnia: a randomized
controlled trial. Biol. Psychiatry 78, 721729 (2015).
REVIEW ARTICLE COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4
16 COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www.nature.com/commsbio
304. Carroll, J. E. et al. Improved sleep quality in older adults with insomnia
reduces biomarkers of disease risk: pilot results from a randomized
controlled comparative efcacy trial. Psychoneuroendocrinology 55, 184192
(2015).
305. Weljie, A. M. et al. Oxalic acid and diacylglycerol 36:3 are cross-species
markers of sleep debt. Proc. Natl Acad. Sci. USA 112, 25692574 (2015).
306. Laing, E. E., Moller-Levet, C. S., Dijk, D. J. & Archer, S. N. Identifying and
validating blood mRNA biomarkers for acute and chronic insufcient sleep in
humans: a machine learning approach. Sleep 42,https://doi.org/10.1093/sleep/
zsy186 (2019).
307. Garbarino, S. & Scoditti, E. On the role of sleep hygiene in health management
during COVID-19 pandemic. Sleep. Med. 77, 74 (2020).
308. Ruiz, F. S. et al. Immune alterations after selective rapid eye movement or
total sleep deprivation in healthy male volunteers. Innate Immun. 18,4454
(2012).
309. Carroll, J. E. et al. Sleep deprivation and divergent toll-like receptor-4
activation of cellular inammation in aging. Sleep 38, 205211 (2015).
310. Pejovic, S. et al. Effects of recovery sleep after one work week of mild sleep
restriction on interleukin-6 and cortisol secretion and daytime sleepiness and
performance. Am. J. Physiol. Endocrinol. Metab. 305, E890896 (2013).
Author contributions
E.S. reviewed the literature and wrote the manuscript draft. S.G. and P.L. contributed to
writing the manuscript and revised the manuscript draft. N.L.B. and N.M. reviewed the
nal manuscript.
Competing interests
The authors declare no competing interests.
Additional information
Supplementary information The online version contains supplementary material
available at https://doi.org/10.1038/s42003-021-02825-4.
Correspondence and requests for materials should be addressed to Sergio Garbarino.
Peer review information Communications Biology thanks the anonymous reviewers for
their contribution to the peer review of this work. Primary Handling Editor: Karli
Montague-Cardoso. Peer reviewer reports are available.
Reprints and permission information is available at http://www.nature.com/reprints
Publishers note Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional afliations.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give
appropriate credit to the original author(s) and the source, provide a link to the Creative
Commons license, and indicate if changes were made. The images or other third party
material in this article are included in the articles Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the
articles Creative Commons license and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly from
the copyright holder. To view a copy of this license, visit http://creativecommons.org/
licenses/by/4.0/.
© The Author(s) 2021
COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-021-02825-4 REVIEW ARTICLE
COMMUNICATIONS BIOLOGY | (2021) 4:1304 | https://doi.org /10.1038/s42003-021-02825-4 | www .nature.com/commsbio 17
... The immune system and sleep homeostasis engage in a bidirectional regulatory relationship, where immune signals directly influence sleep architecture while sleep shapes immune competence through neuroendocrine and cellular mechanisms. This interplay is critical for maintaining physiological balance and optimizing host defense [20,21]. ...
... Hoopes et al. reported detrimental associations between irregular sleep patterns and circulating immune cells in healthy young adults [31]. The immune system and sleep are engaged in a bidirectional relationship in which the immune system regulates sleep, and sleep plays a role in immune homeostasis [20,21]. Reciprocal feedback mechanisms play roles in these relationships and involve cytokines (like tumor necrosis factor α, interferon, and interleukin 6), reactive oxygen species, neurotransmitters, and nuclear factor-κB-driven inflammation [20,21]. ...
... The immune system and sleep are engaged in a bidirectional relationship in which the immune system regulates sleep, and sleep plays a role in immune homeostasis [20,21]. Reciprocal feedback mechanisms play roles in these relationships and involve cytokines (like tumor necrosis factor α, interferon, and interleukin 6), reactive oxygen species, neurotransmitters, and nuclear factor-κB-driven inflammation [20,21]. ...
Article
Full-text available
Background/Objectives: Insomnia is associated with immune function. This study evaluated the association between insomnia and febrile neutropenia in women treated with adjuvant chemotherapy for breast cancer. Methods: This secondary analysis used data from the Canadian Cancer Trial Group MA.21 trial, which compared three chemotherapy regimens (CEF, EC/T dose-dense, or AC/T) in 2104 women with high-risk locoregional breast cancer. A total of 1731 patients completed the EORTC QLQ-C30 questionnaire. We compared “insomnia patients” with patients considered “good sleepers” based on the sleep item of this questionnaire. The primary endpoint was the occurrence of febrile neutropenia. Secondary endpoints were the occurrences of leucopenia and infection. Chemotherapy dose reduction was added as a secondary outcome in an unplanned analysis. Results: Patients with insomnia (16.3%) had a significantly higher rate of febrile neutropenia than good sleepers (12.2%; p = 0.01). After controlling for various confounders, the contribution of insomnia in explaining febrile neutropenia remained statistically significant (OR 1.45, 95% CI 1.07–1.97, p = 0.02). Chemotherapy dose reductions were significantly more frequent in patients with insomnia (30.6%) than in good sleepers (21.8%; p < 0.0001). The relationship remained significant in the multivariate analysis (OR 1.67, 95% CI 1.30–2.15, p < 0.0001). Conclusions: In the MA21 trial, insomnia was associated with febrile neutropenia. Furthermore, chemotherapy dose reductions were more common in women with insomnia. These results suggest that the role of insomnia in potential cancer outcomes needs to be confirmed in other studies, given the possible implication of dose reductions on the prognosis of women receiving chemotherapy for breast cancer.
... We show that the quality of pre-infection sleep predicts long COVID risk, independently of the severity of the acute infection, supporting and extending findings from a previous study 10 done exclusively in women. While the mechanisms behind long COVID remain unknown, sleep quality and disturbances are known to play a part in many of the candidate mechanisms under investigation, 1 such as persistent inflammation, 34 changes to the gut microbiota 35 and autoimmunity. 34 Sleep problems are additionally linked to poor mental health, 36 which has been shown to predict long COVID. ...
... While the mechanisms behind long COVID remain unknown, sleep quality and disturbances are known to play a part in many of the candidate mechanisms under investigation, 1 such as persistent inflammation, 34 changes to the gut microbiota 35 and autoimmunity. 34 Sleep problems are additionally linked to poor mental health, 36 which has been shown to predict long COVID. 37 Our sleep quality measure combined the effects of both sleep efficiency and sleep duration, but exploratory analyses suggested that low and inconsistent sleep efficiency-which can reflect a range of sleep disturbances, such as increased sleep latency and fragmented sleep-was driving the relationship observed. ...
Article
Full-text available
Background Studies into the bidirectional relationship between sleep and long COVID have been limited by retrospective pre-infection sleep data and infrequent post-infection follow-up. We therefore used prospectively collected monthly data to evaluate how pre-infection sleep characteristics affect risk of long COVID and to track changes in sleep duration during the year after SARS-CoV-2 infection. Methods COVIDENCE UK is a prospective, population-based UK study of COVID-19 in adults. We included non-hospitalised participants with evidence of SARS-CoV-2 infection and used logistic regression to estimate adjusted ORs for the association between preinfection sleep characteristics and long COVID. We assessed post-infection sleep duration using multilevel mixed models. We collected sleep data from participants using a subset of questions from the Pittsburgh Sleep Quality Index. We defined long COVID as unresolved symptoms at least 12 weeks after infection. COVIDENCE UK is registered with ClinicalTrials.gov, NCT04330599. Results We included 3994 participants in our long COVID risk analysis, of whom 327 (8.2%) reported long COVID. We found an inverse relationship between pre-infection sleep quality and risk of long COVID (medium vs good quality: OR 1.37, 95% CI 1.04 to 1.81; medium–low vs good: 1.55, 1.12 to 2.16; low vs good: 1.94, 1.11 to 3.38). Greater variability in pre-infection sleep efficiency was also associated with long COVID when adjusted for infection severity (OR per percentage-point increase 1.07, 1.02 to 1.12). We assessed post-infection sleep duration in 6860 participants, observing a 0.11 hour (95% CI 0.09 to 0.14) increase in the first month after infection compared with pre-infection, with larger increases for more severe infections. After 1 month, sleep duration largely returned to pre-infection levels, although fluctuations in duration lasted up to 6 months after infection among people reporting long COVID. Conclusions While poor-quality sleep before SARS-CoV-2 infection associates with increased risk of long COVID thereafter, changes in sleep duration after infection in these non-hospitalised cases were modest and generally quick to resolve. Trial registration number NCT04330599.
... From a pathophysiological standpoint, sleep may exert an indirect modulatory effect on suicide risk through multiple mechanisms such as neuroinflammation, emotional dysregulation, and the hypothalamicpituitary-adrenal (HPA) axis [26]. Sleep deprivation and disturbances can lead to an increase in pro-inflammatory cytokines (e.g., IL-6, TNF-α) and a decrease in antiinflammatory cytokines (e.g., IL-10) [27,28]. Elevated levels of these pro-inflammatory markers have been associated with increased depressive symptoms and suicidal ideation [29]. ...
Article
Full-text available
Objectives This study aimed to investigate the association between sleep problems and suicidal behaviors as well as healthcare utilization in Canadian adults with chronic diseases, while also examining the mediating role of mental illness. Methods Data were drawn from the 2015–16 cycle of the Canadian Community Health Survey, specifically from Ontario, Manitoba, and Saskatchewan — the provinces that included the optional sleep module. A total of 22,700 participants aged ≥ 18 years and diagnosed with at least one chronic disease were included in the analysis. Sleep problems were defined as extreme sleep durations (either < 5 or ≥ 10 h) and insomnia. Mental illness was classified as a self-reported mood or anxiety disorder. Results Participants with extreme sleep durations (compared to 7 to < 8 h) and those with insomnia (compared to no insomnia) showed a higher prevalence of suicidal ideation, suicidal plans, and increased healthcare utilization. After adjusting for multiple covariates, both extreme sleep durations and insomnia remained significantly associated with increased odds of suicidal ideation, suicidal plans, and healthcare utilization. Mediation analyses indicated that mental illness partially mediated these associations. Conclusions Both extreme sleep durations and insomnia were independently associated with higher odds of suicidal behaviors and increased healthcare utilization in adults with chronic diseases, with mental illness playing a partial mediating role in these relationships.
... Cuando hay privación del sueño, el organismo entre en un estado de activación crónica del sistema de respuesta al estrés, caracterizado por una mayor reactividad del eje HHA ante estímulos cotidianos. Esta hiperactividad se asocia con una elevación persistente de los niveles de cortisol, lo cual contribuye a alteraciones en la regulación emocional, disminución del umbral del dolor y mayor susceptibilidad a trastornos del estado de ánimo(23)(24)(25).En un mundo donde el estrés crónico y los estilos de vida poco saludables erosionan lentamente los cimientos fisiológicos de la salud humana, la alostasis se consolida como un marco teórico robusto para redefinir el cuidado de la salud. La actividad física regular, la alimentación saludable y una buena calidad del sueño no deben seguir siendo concebidos como simples recomendaciones individuales, sino como componentes estructurales de toda estrategia de promoción de la salud y prevención de enfermedades. ...
Article
El concepto de alostasis ha emergido como un marco teórico clave para comprender los procesos de adaptación del organismo frente a los desafíos del entorno. A partir de la evolución del paradigma de la homeostasis, este artículo reflexiona sobre cómo el estrés crónico, derivado de estilos de vida poco saludables, puede desencadenar una sobrecarga alostática con consecuencias fisiopatológicas como inflamación crónica, disfunción neuroendocrina y deterioro conductual, contribuyendo así al desarrollo de enfermedades no transmisibles y trastornos mentales. En este contexto, se propone la actividad física regular, una alimentación saludable y una buena calidad del sueño como tres hábitos esenciales para el cuidado de la salud. Estos comportamientos actúan como moduladores positivos de la carga alostática, favorecen procesos de recalibración fisiológica y constituyen pilares fundamentales de cualquier estrategia de promoción de la salud. Finalmente, se hace un llamado a integrar estos tres esenciales en las políticas públicas y prácticas clínicas, reconociendo su papel estructural en la sostenibilidad de la salud individual y colectiva.
... Sleep is an prime function for the rest and restoration of each organ of our body. [1] This natural and vital urge takes up nearly one-third of our lives, yet the sleep remains a mystery to us. On one hand, in supporting digestion, metabolism, nourishment and overall health. ...
Article
Full-text available
This research study entitled “Role of Pratimarsh Nasya on Sukh Nidra-Prabodham (quality of sleep)” evaluates the effects of Pratimarsha Nasya on improving overall sleep quality in individuals identified as poor sleepers based on a Pittsburgh Sleep Quality Index (PSQI) score ranging from 5 to 10. The research study, explores the effectiveness of Pratimarsh Nasya, a traditional Ayurvedic therapy, in improving sleep quality among healthy individuals. A total of 150 participants were enrolled and divided into two groups: the intervention group (n=75) received two drops of Murchhit Til Tail (processed sesame oil) intranasally each evening as Pratimarsh Nasya, while the control group (n=75) followed standard sleep hygiene practices, including consistent bedtime routines and environmental adjustments. The intervention spanned a period of two months, during which both groups demonstrated significant improvement in sleep quality. Comparative analysis revealed that the Pratimarsh Nasya group experienced more immediate and pronounced improvement in sleep quality, as indicated by higher percentage relief. However, follow-up assessments suggested that the sleep hygiene group exhibited a lower recurrence of sleep disturbances over time. These findings suggest that Pratimarsh Nasya may offer an effective short-term strategy for enhancing sleep quality, while sleep hygiene practices contribute to more sustained, long-term benefits. No treatment related adverse events were reported in the study. Further research is recommended for underlying mechanisms and long term effects of Nasya therapy on sleep health.
... Cytokines such as IL-1β and TNF-α, which are pivotal in inflammation, also serve as sleep-regulating molecules, promoting NREM sleep at physiological levels but contributing to sleep fragmentation when chronically elevated (Besedovsky et al., 2019;Zielinski and Gibbons, 2022). Conversely, sleep deprivation skews immune balance, reducing the number and function of natural killer cells (De Lorenzo et al., 2015), impairing T-cell responsiveness (Tune et al., 2021), and elevating circulating proinflammatory cytokines, thus mimicking an inflammaging-like profile even in younger individuals (Garbarino et al., 2021). ...
Article
Full-text available
The relationship between sleep and metabolism has emerged as a critical factor in aging and age-related diseases, including Alzheimer's disease and dementia. Mitochondrial oxidative phosphorylation, essential for neuronal energy production, also generates reactive oxygen species (ROS), which increase with age and contribute to oxidative stress. Sleep plays a vital role in modulating redox balance, facilitating the clearance of free radicals, and supporting mitochondrial function. Disruptions in sleep are closely linked to redox imbalances, and emerging evidence suggests that pharmacological interventions, such as dual orexin receptor antagonists and antioxidant-based therapies, may help restore redox homeostasis. Furthermore, antioxidant-rich diets and supplements have shown promise in improving both sleep quality and metabolic health in aging populations. Neurons, with their high energy demands, are particularly vulnerable to oxidative damage, making redox regulation crucial in maintaining brain integrity. This review explores the bidirectional relationship between sleep and redox metabolism through five key areas: (1) sleep's role in free radical regulation, (2) ROS as mediators of age-related sleep disturbances, (3) feedback loops between impaired sleep and brain metabolism, (4) sleep, redox, and aging in peripheral systems, and (5) therapeutic strategies to restore redox balance and improve aging outcomes. Understanding these mechanisms may provide new targets for interventions aimed at mitigating age-associated diseases.
... Sleep disturbances substantially influence patients' recovery from chronic diseases, especially in critical care settings. Disrupted sleep is associated with a variety of negative effects, including immunological dysfunction, decreased wound healing, and higher susceptibility to infections, which can impede recovery processes (Altman et al., 2017;Garbarino et al., 2021). Furthermore, sleep disturbance has an impact on the body functions that are critical to returning to a normal health state, including respiratory muscle weakness, cardiovascular effects resulting in hypertension, heart failure, and stroke, and hormonal changes resulting in insulin resistance in diabetics (Delaney et al., 2015;During & Kawai, 2017;Singh et al., 2022). ...
Article
Full-text available
Background Sleep is essential for physiologic function and mental health stability, which promotes recovery from severe illness. However, poor sleep quality is a common complaint in the critical care unit. In intensive care units (ICUs), physical aspects of patient care are often prioritized over humanized aspects of care. Objective To assess the level of sleep quality and its associated factors among adult patients admitted to critical care units in Addis Ababa. Method A cross-sectional study design was conducted in three selected public hospitals. Over 4 months of the period we consecutively collected data among ICU patients, who were oriented to time, person, and place on the day of discharge. A modified freedman self-reporting and personal characteristics questionnaire was used. The data were entered into Epi-data version 4.4.6.0 and analyzed with SPSS 25. Both bivariate and multivariate analyses were conducted. A P-value of <.05 indicated statistical significance, and an adjusted odds ratio with a 95% confidence range was used to show the strength of the association. Result Of the total 102 participants, 57.8% of ICU patients had poor sleep quality. Four variables were identified as significant associations with poor sleep quality among ICU patients. These included elderly age (adjusted odd ratio [AOR] = 3.4; 95% confidence interval [CI]: 1.42, 7.93), comorbidity (AOR = 2.5; 95% CI: 1.24, 9.03), light exposure (AOR = 2.0; 95% CI: 1.16, 5.11), and the monitor's alarm (AOR = 1.7; 95% CI: 1.04, 8.23) were identified as significant association factors for poor sleep quality. Conclusion and recommendation Poor sleep quality was a major concern for ICU patients in this study. Elderly individuals, those with comorbidity, light exposure, and monitor alarms have been associated with poor sleep quality. Reducing nighttime light and adjusting alarm settings may significantly improve sleep quality, resulting in better recovery results in critical care units.
Article
Full-text available
Introduction: Tuberculosis (TB) can cause persistent symptoms, such as sleep disturbances, even after treatment, significantly reducing the patient’s quality of life. As good sleep is vital for overall health, early identification and management of poor sleep quality are essential for improving long-term outcomes in TB survivors. Aim: To determine the sleep quality among treated TB patients and to assess its impact on health-related quality of life in a tertiary care centre. Methods: A cross-sectional study was conducted involving 60 patients with pulmonary and extrapulmonary TB who completed ATT for 6 months. Sleep quality was assessed using the Pittsburgh Sleep Quality Index (PSQI), and HRQoL was evaluated using the SF-36 Questionnaire. Statistical analyses were performed to determine the correlation between PSQI scores and various HRQoL domains. Results:The mean age was 39.7 years, and 61.7% of patients were male. No significant sex differences were observed (p = 0.254). A total of 35% had poor sleep quality (PSQI > 5), which was significantly more common in those aged > 50 years (p = 0.007). Poor sleepers had notably worse scores in pain, general health, energy/fatigue, and physical functioning (all p < 0.05), with strong negative correlations between PSQI and pain (r = -0.794), physical functioning (r = -0.669), and energy/fatigue (r = -0.635) scores. Conclusion: Sleep disturbances are common among Treated TB patients and are strongly associated with a reduced quality of life. Integrating routine sleep quality screening and psychosocial support into post-treatment care may improve long-term outcomes for TB survivors.
Article
Full-text available
Background Multiple sclerosis (MS), affected about 2.8 million people worldwide in 2020, is often associated with sleep disorders. These disturbances may arise from the disease itself—such as pain—or as side effects of symptomatic and immunological treatments. Methods The present study is cross-sectional- analytic study. The population studied consists of MS patients living in Kashan city. The sample size under investigation is comprised of 174 patients diagnosed with MS. The information was gathered through demographic information questionnaires, the hospital anxiety and depression scale(HADS), Pittsburgh Sleep Quality Index (PSQI), Epworth Sleepiness Scale (ESS), Insomnia Severity Index (ISI), and Multiple Sclerosis Impact Scale (MSIS). The data were analyzed through Chi-square tests and logistic regression using Stata version 14 software. Results : Of 174 MS patients (81.76% female, mean age 34.16 ± 7.62), Mild insomnia affected 17.05%, and 68.18% experienced frequent night awakenings. Significant predictors of sleepiness included education level (OR: 18.20; p = 0.03), insomnia severity (OR: 5.24; p = 0.04), and morning dysfunction (OR: 6.49; p = 0.04), as well as MS medication. Most patients were not taking sleep medication (86.05%). Mild anxiety and baseline depressive symptoms were prevalent (69.32% and 80.68%, respectively), and 80.11% reported low quality of life. Conclusions The final model demonstrated an effective relationship between the Sleepiness and variables of education level, drugs used in MS, Insomnia Severity, and morning dysfunction in the presence of other variables.
Chapter
Sleep is a dynamic process that is influenced by our daily behaviours and in turn impacts our waking choices. It’s important to understand that healthy sleep involves not just the duration but also the timing and architecture of sleep, which can affect disease risks and outcomes. The regulation of sleep is driven by the sleep homeostat, also known as Process S, and the circadian system, known as Process C. Sleep itself consists of Non-Rapid Eye Movement (NREM) and Rapid Eye Movement (REM) stages, each with distinct brain wave patterns and physiological functions. The circadian system, which is governed by sunlight and melatonin, synchronises our body’s clocks and regulates physiological rhythms. There is variability in individual sleep needs, which are influenced by genetics, and these needs change across the lifespan. Poor-quality sleep is linked to mental health issues, cardiovascular disease, diabetes, and other pathologies. Common sleep disorders include insomnia and obstructive sleep apnoea, with lifestyle interventions being key treatments. Good sleep health can be promoted through regular schedules, optimal bedroom environments, and managing lifestyle factors. Education and policy changes are needed to address sleep issues.
Article
Full-text available
Sleep is a recuperative process, and its dysregulation has cognitive, metabolic, and immunological effects that are largely deleterious to human health. Epidemiological and empirical studies have suggested that sleep fragmentation (SF) as result of obstructive sleep apnea (OSA) and other sleep abnormalities leads to pronounced inflammatory responses, which are influenced by the sympathetic nervous system (SNS). However, the underlying molecular mechanisms contributing to SNS regulation of SF-induced inflammation are not fully understood. To assess the effects of the SNS upon inflammatory responses to SF, C57BL/6j female mice were placed in automated SF chambers with horizontally moving bars across the bottom of each cage at specified intervals to disrupt sleep. Mice were first subjected to either control (no bar movement), acute sleep fragmentation (ASF), or chronic sleep fragmentation (CSF) on a 12:12-h light/dark schedule. ASF involved a bar sweep every 120 s for 24 h, whereas CSF involved a bar sweep every 120 s for 12 h (during 12 L; resting period) over a period of 4 weeks. After exposure to these conditions, mice received an intraperitoneal injection of either phentolamine (5 mg/kg BW; an α-adrenergic receptor blocker), propranolol (5 mg/kg BW; a β-adrenergic receptor blocker), or vehicle (saline). Serum corticosterone concentration, brain and peripheral cytokine (IL1β, TNFα, and TGFβ) mRNA expression, and body mass were assessed. ASF and CSF significantly elevated serum corticosterone concentrations as well as cytokine mRNA expression levels compared with controls, and mice subjected to CSF had decreased body mass relative to controls. Mice subjected to CSF and treated with phentolamine or propranolol had a greater propensity for a decrease in cytokine gene expression compared with ASF, but effects were tissue-specific. Taken together, these results suggest that both α- and β-adrenergic receptors contribute to the SNS mediation of inflammatory responses, and adrenergic antagonists may effectively mitigate tissue-specific SF-mediated inflammation.
Article
Full-text available
Sleep dysregulation is a feature of dementia but it remains unclear whether sleep duration prior to old age is associated with dementia incidence. Using data from 7959 participants of the Whitehall II study, we examined the association between sleep duration and incidence of dementia (521 diagnosed cases) using a 25-year follow-up. Here we report higher dementia risk associated with a sleep duration of six hours or less at age 50 and 60, compared with a normal (7 h) sleep duration, although this was imprecisely estimated for sleep duration at age 70 (hazard ratios (HR) 1.22 (95% confidence interval 1.01–1.48), 1.37 (1.10–1.72), and 1.24 (0.98–1.57), respectively). Persistent short sleep duration at age 50, 60, and 70 compared to persistent normal sleep duration was also associated with a 30% increased dementia risk independently of sociodemographic, behavioural, cardiometabolic, and mental health factors. These findings suggest that short sleep duration in midlife is associated with an increased risk of late-onset dementia. Sleep dysregulation has been linked to dementia, but it is unknown whether sleep duration earlier in life is associated with dementia risk. Here, the authors show higher dementia risk associated with short sleep duration (six hours or less) in a longitudinal study of middle and older age adults.
Article
Full-text available
The classical liver-derived and serum-effective complement system is well appreciated as a key mediator of host protection via instruction of innate and adaptive immunity. However, recent studies have discovered an intracellularly active complement system, the complosome, which has emerged as a central regulator of the core metabolic pathways fueling human immune cell activity. Induction of expression of components of the complosome, particularly complement component C3, during transmigration from the circulation into peripheral tissues is a defining characteristic of monocytes and T cells in tissues. Intracellular complement activity is required to induce metabolic reprogramming of immune cells, including increased glycolytic flux and OXPHOS, which drive the production of the pro-inflammatory cytokine IFN-γ. Consequently, reduced complosome activity translates into defects in normal monocyte activation, faulty Th1 and cytotoxic T lymphocyte responses and loss of protective tissue immunity. Intriguingly, neurological research has identified an unexpected connection between the physiological presence of innate and adaptive immune cells and certain cytokines, including IFN-γ, in and around the brain and normal brain function. In this opinion piece, we will first review the current state of research regarding complement driven metabolic reprogramming in the context of immune cell tissue entry and residency. We will then discuss how published work on the role of IFN-γ and T cells in the brain support a hypothesis that an evolutionarily conserved cooperation between the complosome, cell metabolism and IFN-γ regulates organismal behavior, as well as immunity.
Article
Full-text available
Tumor necrosis factor alpha (TNF) has sleep regulatory and brain development roles. TNF promotes sleep in vivo and in vitro while TNF inhibition diminishes sleep. Transmembrane (tm) TNF and the tmTNF receptors (Rs), are cleaved by tumor necrosis factor alpha convertase to produce soluble (s) TNF and sTNFRs. Reverse signaling occurs in cells expressing tmTNF upon sTNFR binding. sTNFR administration in vivo inhibits sleep, thus we hypothesized that a wake-like state in vitro would be induced by sTNFR-tmTNF reverse signaling. Somatosensory cortical neuron/glia co-cultures derived from male and female mice lacking both TNFRs (TNFRKO), or lacking TNF (TNFKO) and wildtype (WT) mice were plated onto six-well multi-electrode arrays. Daily one-hour electrophysiological recordings were taken on culture days 4 through 14. sTNFR1 (0.0, 0.3, 3, 30, 60, and 120 ng/µL) was administered on day 14. A final one-hour recording was taken on day 15. Four measures were characterized that are also used to define sleep in vivo: action potentials (APs), burstiness index (BI), synchronization of electrical activity (SYN), and slow wave power (SWP; 0.25-3.75 Hz). Development rates of these emergent electrophysiological properties increased in cells from mice lacking TNF or both TNFRs compared to cells from WT mice. Decreased SWP, after the three lowest doses (0.3, 3 and 30 ng/µL) of the sTNFR1, indicate a wake-like state in cells from TNFRKO mice. A wake-like state was also induced after 3 ng/µl sTNFR1 treatment in cells from TNFKO mice, which express the TNFR1 ligand, lymphotoxin alpha. Cells from WT mice showed no treatment effects. Results are consistent with prior studies demonstrating involvement of TNF in brain development, TNF reverse signaling, and sleep regulation in vivo. Further, the current demonstration of sTNFR1 induction of a wake-like state in vitro is consistent with the idea that small neuronal/glial circuits manifest sleep- and wake-like states analogous to those occurring in vivo. Finally, that sTNF forward signaling enhances sleep while sTNFR1 reverse signaling enhances a wake-like state is consistent with other sTNF/tmTNF/sTNFR1 brain actions having opposing activities.
Article
Full-text available
The blood–brain barrier (BBB) is an important physiological barrier that separates the central nervous system (CNS) from the peripheral circulation, which contains inflammatory mediators and immune cells. The BBB regulates cellular and molecular exchange between the blood vessels and brain parenchyma. Normal functioning of the BBB is crucial for the homeostasis and proper function of the brain. It has been demonstrated that peripheral inflammation can disrupt the BBB by various pathways, resulting in different CNS diseases. Recently, clinical research also showed CNS complications following SARS‐CoV‐2 infection and chimeric antigen receptor (CAR)‐T cell therapy, which both lead to a cytokine storm in the circulation. Therefore, elucidation of the mechanisms underlying the BBB disruption induced by peripheral inflammation will provide an important basis for protecting the CNS in the context of exacerbated peripheral inflammatory diseases. In the present review, we first summarize the physiological properties of the BBB that makes the CNS an immune‐privileged organ. We then discuss the relevance of peripheral inflammation‐induced BBB disruption to various CNS diseases. Finally, we elaborate various factors and mechanisms of peripheral inflammation that disrupt the BBB.
Article
Full-text available
Significance Synaptic plasticity in the hippocampus is important for learning and memory formation. In particular, homeostatic synaptic plasticity enables neurons to restore their activity levels in response to chronic neuronal activity changes. While astrocytes modulate synaptic functions via the secretion of factors, the underlying molecular mechanisms remain unclear. Here, we show that suppression of hippocampal neuronal activity increases cytokine IL-33 release from astrocytes in the CA1 region. Activation of IL-33 and its neuronal ST2 receptor complex promotes functional excitatory synapse formation. Moreover, IL-33/ST2 signaling is important for the neuronal activity blockade-induced increase of CA1 excitatory synapses in vivo and spatial memory formation. This study suggests that astrocyte-secreted IL-33 acts as a negative feedback control signal to regulate hippocampal homeostatic synaptic plasticity.
Article
Recent studies report that inhibiting TNF-α might be a novel therapeutic strategy for managing brain ischemia. Our previous study reported that mesenchymal stem cell (MSC) transplantation could suppress TNF-α level in both serum and brain. However, the cell type(s) that contribute to the production of TNF-α during ischemia following MSC transplantation has not been well studied. In the present study, we found by fluorescent immunohistochemistry, that 7.95 ± 6.17% of TNF-α⁺ cells co-expressed Iba-1 in the infarct area of dMCAO rats, a majority of which were found to be CD68⁺ (activated microglia), suggesting that resident microglial population were not the major source of TNF-α expression. 68.49 ± 5.12% of the TNF-α⁺ cells in the infarct area could be labeled by GFAP, a specific marker for astrocytes, indicating that resident GFAP⁺ astrocytes might be the major source of TNF-α expression in the infarct area. In addition to the infarct area, the GFAP⁺/TNF-α⁺ double-positive astrocytes accounted for 73.68 ± 7.48% of the TNF-α⁺ cells in striatum and corpus callosum. The infiltrating cells, including monocytes and lymphocytes, were not the main source of TNF-α either. In response to MSC transplantation, the total TNF-α⁺ cells as well as the percentage of TNF-α-expressing astrocytes were significantly reduced in the infarct area, suggesting that MSC transplantation could suppress the expression of TNF-α by astrocytes. Taken together, the results demonstrated that resident astrocytes, but not microglia, were the major source of TNF-α expression and could be suppressed by MSC infusion.
Article
Clock genes work as an auto-regulated transcription-translational loop of circadian genes that drives the circadian rhythms in each cell and they are essential to physiological requests. Since metabolism is a dynamic process, it involves several physiological variables that circadian cycling. The clock genes alterations can affect multiple systems concomitantly, because they constitute the promoter factors for relevant metabolic pathways. Considering the intertwined structure of signaling, regulatory, and metabolic processes within a cell, we employed a genome-scale biomolecular network. Accordingly, a meta-analysis of diabetic-associated transcriptomic datasets was performed, and the core information on differentially expressed genes (DEGs) was obtained by statistical analyses. In the current study, meta-analysis was performed on type 2 diabetes, circadian rhythm-related genes, and breast, bladder, liver, pancreas, colon and rectum cancer-associated transcriptome data using the integration of gene expression profiles with genome-scale biomolecular networks in diabetes samples. First, we detected downregulated and upregulated DEGs in mouse cortex and hypothalamus samples of mice with sleep deprivation. In summary, upregulated genes active genes associated with oxidative phosphorylation, cancer and diabetes, mainly in hypothalamus specimens. In cortex, we observed mainly downregulation of immune system. DEGs were combined with 214 circadian rhythm related genes to type 2 DM and cancer samples. We observed that several common genes deregulated in both diseases. Klf10, Ntkr3, Igf1, Usp2, Ezh2 were both downregulated in type 2 DM and cancer samples, while Arntl2 and Agrp were upregulated. It seems that the changes in mRNA are contributing to the phenotypic changes in type 2 DM, resulting in phenotypic changes associated with the malignant transformation. Taking those genes to perform a survival analysis, we found only Igf1, Usp2 and Arntl2 genes associated with patient outcomes. While Igf1 and Usp2 downregulation had a negative impact, Arntl2 upregulation was associated with poor survival both in BLCA and BRCA cancer samples. Our data stimulate efforts in news studies to achieve the experimental and clinical validation about these biomolecules.
Article
This report presents final 2019 U.S. mortality data on deaths and death rates by demographic and medical characteristics. These data provide information on mortality patterns among U.S. residents by variables such as sex, age, race and Hispanic origin, and cause of death. Life expectancy estimates, agespecific death rates, 10 leading causes of death, and 10 leading causes of infant death were analyzed by comparing 2019 and 2018 final data (1).
Article
Study objectives New theory and measurement approaches have facilitated nuanced investigation of how sleep loss impacts dimensions of affective functioning. To provide a quantitative summary of this literature, three conceptually related meta-analyses examined the effect of sleep restriction and sleep deprivation on mood, emotion, and emotion regulation across the lifespan (i.e., from early childhood to late adulthood). Method A total of 241 effect sizes from 64 studies were selected for inclusion, and multilevel meta-analytic techniques were used when applicable. Results There was a moderate, positive effect of sleep loss on negative mood (g = .45), which was stronger for studies with younger samples, as well as a large, negative effect of sleep loss on positive mood (g = -.93); type of sleep manipulation (i.e., restriction or deprivation) did not moderate either effect. After correcting for publication bias, a modest but significant negative effect emerged for the effect of sleep on emotion (g = .11); the valence of emotional stimuli did not change the direction of this effect, and type of sleep manipulation was also not a significant moderator. Finally, sleep restriction had a small, negative effect on adaptive emotion regulation (g = -.32), but no significant impact on maladaptive emotion regulation (g = .14); all studies on adaptive emotion regulation were conducted with youth samples. Conclusions Sleep loss compromises optimal affective functioning, though the magnitude of effects varies across components. Findings underscore the importance of sleep for healthy affective outcomes.