Sleep and Its Disorders: Allan I. Pack Qing Yun Li Editors
Sleep and Its Disorders: Allan I. Pack Qing Yun Li Editors
Sleep and Its Disorders: Allan I. Pack Qing Yun Li Editors
Allan I. Pack
Qing Yun Li Editors
Sleep
and its
Disorders
Translational Medicine
Translational Medicine Research
Series Editors
Zhu Chen, Shanghai Jiaotong University, Shanghai, Shanghai, China
Xiaoming Shen, Shanghai Jiaotong University, Shanghai, Shanghai, China
Saijuan Chen, Shanghai Jiaotong University, Shanghai, Shanghai, China
Kerong Dai, Shanghai Jiaotong University, Shanghai, Shanghai, China
Translational medicine converts promising laboratory discoveries into clinical appli-
cations and elucidates clinical questions with the use of bench work, aiming to
facilitate the prediction, prevention, diagnosis and treatment of diseases. The devel-
opment of translational medicine will accelerate disease control and the process of
finding solutions to key health problems. It is a multidisciplinary endeavor that
integrates research from the medical sciences, basic sciences and social sciences,
with the aim of optimizing patient care and preventive measures that may extend
beyond health care services. Therefore, close and international collaboration
between all parties involved is essential to the advancement of translational
medicine.
To enhance the aforementioned international collaboration as well as to provide a
forum for communication and cross-pollenation between basic, translational and
clinical research practitioners from all relevant established and emerging disciplines,
the book series “Translational Medicine Research” features original and observa-
tional investigations in the broad fields of laboratory, clinical and public health
research, aiming to provide practical and up-to-date information on significant
research from all subspecialties of medicine and to broaden readers’ vision horizons,
from bench to bed and bed to bench.
Produced in close collaboration with National Infrastructures for Translational
Medicine (Shanghai), the largest translational medicine research center in China, the
book series offers a state-of-the-art resource for physicians and researchers alike who
are interested in the rapidly evolving field of translational medicine. Prof. Zhu Chen,
the Editor-in-Chief of the series, is a hematologist at Shanghai Jiao Tong University,
China’s former Minister of Health, and chairman of the center’s scientific advisory
board.
Allan I. Pack • Qing Yun Li
Editors
© Springer Nature B.V. and Shanghai Jiao Tong University Press 2022
This work is subject to copyright. All rights are reserved by the Publishers, whether the whole or part of
the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation,
broadcasting, reproduction on microfilms or in any other physical way, and transmission or information
storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology
now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication
does not imply, even in the absence of a specific statement, that such names are exempt from the relevant
protective laws and regulations and therefore free for general use.
The publishers, the authors, and the editors are safe to assume that the advice and information in this
book are believed to be true and accurate at the date of publication. Neither the publishers nor the
authors or the editors give a warranty, express or implied, with respect to the material contained
herein or for any errors or omissions that may have been made. The publishers remain neutral with
regard to jurisdictional claims in published maps and institutional affiliations.
This Springer imprint is published by the registered company Springer Nature B.V.
The registered company address is: Van Godewijckstraat 30, 3311 GX Dordrecht, The Netherlands
Contents
v
vi Contents
Part IV Narcolepsy
12 Narcolepsy and Orexin/Hypocretin . . . . . . . . . . . . . . . . . . . . . . . . . 229
Fu Long Xiao, Jun Zhang, and Fang Han
Allan I. Pack
Abstract There has been recent substantial progress in elucidating gene variants
that confer risk for common sleep disorders, and also for those associated with
different quantitative aspects of the sleep/circadian phenotype. The major strategy
employed has been genome-wide association studies (GWAS). Initially, the
approach was to recruit and phenotype large numbers of cases with specific sleep
disorders and controls. This was used to identify gene variants conferring risk for
Restless Legs Syndrome (RLS) and narcolepsy. This approach is, however, time-
consuming and expensive. Newer approaches have involved establishing biobanks
with large numbers of subjects with a variety of disorders and controls, all of whom
are genotyped. One of the most successful such efforts has been the UK Biobank.
Studies using data from the UK Biobank have led to successful GWAS for sleep
duration, chronotype, excessive sleepiness, and insomnia. The challenge is that the
phenotype data are quite limited. Moreover, the UK Biobank has large numbers of
relatively healthy subjects so the prevalence of any specific disorder is low. This
approach has been complemented by development of large biobanks based on
specific health systems in which patients have given blood samples for genotyping.
These biobanks have a much larger prevalence of disorders, including sleep disor-
ders. The use of these biobanks for a specific disorder requires development of
algorithms to identify individuals with a specific disorder using data in the electronic
health record (EHR). This approach is starting to be used for the study of variants
conferring risk for specific sleep disorders. Results from GWAS are being used to
develop polygenic risk scores (PRS) for specific disorders. This, when combined
with other information, can be used to predict who will develop specific disorders,
opening the opportunity for prevention.
A. I. Pack (*)
Translational Research Laboratories, Division of Sleep Medicine, Department of Medicine,
Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
e-mail: pack@pennmedicine.upenn.edu
© Springer Nature B.V. and Shanghai Jiao Tong University Press 2022 3
A. I. Pack, Q. Y. Li (eds.), Sleep and its Disorders, Translational Medicine Research,
https://doi.org/10.1007/978-94-024-2168-2_1
4 A. I. Pack
Fig. 1.1 Diagram illustrating the concept of common and rare variants. The plot shows the
frequency of the allele (x-axis) and effect size for the allele on the phenotype of interest. Common
alleles (bottom right) have, in general, a low effect size. There are examples including narcolepsy
where common variants have large effect sizes since it makes the individual susceptible to a
particular environmental challenge. Rare variants (left side) typically have large effects on the
phenotype [From Manolio et al. (2009) with permission]
There are rapidly evolving approaches and resources to identify genetic variants that
provide risk for, or protection against, common sleep disorders. Moreover, since
almost all aspects of sleep are heritable (Pack et al. 2021), there are efforts to
understand the genetic regulation of sleep in humans. In considering genetic
approaches, it is helpful to think about common variants with small effects and
rare variants with large effects (Manolio et al. 2009) (see Fig. 1.1). Common variants
occur in >5% of the population. A single common variant has a small effect with an
odds ratio typically in the range of 1.1–1.4. There are examples where common
variants have a large effect size. This typically occurs when the variant makes the
individual susceptible to an environmental challenge. Many individuals can have
this variant but not have the disorder, since they have not been exposed to the
relevant challenge. Rare variants occur typically in <1% of the population. Each rare
variant has a large effect on the relevant phenotype. While each variant is rare, there
are a lot of them. Different variants can occur in the same gene and there are
statistical approaches to allow analysis of all such variants combined. Common
and rare variants conferring risk for a particular disorder can occur in the same genes,
i.e., there are mutation hot spots. The variants being discussed here are single
nucleotide polymorphisms (SNPs). Common variants typically occur in introns or
in the regulatory region of the gene. Rare variants with large effects are typically in
1 Evolving Approaches to Identifying Genetic Risk Variants for Sleep Disorders 5
exons, i.e., in protein coding regions of the gene. Exon sequencing that is now
routine assesses the presence of these variants across the whole genome. In addition
to SNPs, copy number variants (CNVs) including insertions and deletions of geno-
mic regions (Conrad et al. 2010; Redon et al. 2006) provide additional mechanisms
for genetic underpinnings of disease.
Currently, the major approach to identifying variants of genes conferring risk for,
or protection to, sleep disorders, and that associate with various aspects of the sleep
phenotype is using a genome-wide association study (GWAS). The first GWAS was
published in 2005 for macular degeneration (Klein et al. 2005). There has been an
explosion of GWAS with the availability of online resources that catalog results of
previous GWAS (Welter et al. 2014; Visscher et al. 2012; MacArthur et al. 2017).
The first major GWAS for a sleep disorder was in 2007 for Restless Legs
Syndrome (Winkelmann et al. 2007; Stefansson et al. 2007). There were in that
year publications from Germany/Canada (Winkelmann et al. 2007) and from Iceland
(Stefansson et al. 2007). The German study was based on a detailed questionnaire
(Allen et al. 2003), while in Iceland use was made of leg actigrams to evaluate
periodic limb movements during sleep (Allen et al. 2003). The latter allowed
identification of risk variants associated with the motor component of RLS rather
than the sensory component. The two studies identified the same risk variants in
BTDP9 and MEIS1. The prevailing strategy for GWAS at the time was to assemble
large numbers of well-studied cases and controls. This had the advantage that the
phenotype was rigorously defined and in-depth phenotyping of subjects was possi-
ble. However, it was time-consuming and expensive to recruit and study the large
sample of participants that is required to protect against false-positive associations.
The findings from these GWAS in RLS have been replicated in many subsequent
studies (Kemlink et al. 2009; Yang et al. 2011; Moore et al. 2014).
GWAS has also been used to study the genetics of narcolepsy. It has long been
known that a particular HLA antigen has been implicated as conferring risk for
narcolepsy, i.e., DQB1*0602 (Juji et al. 1984). Recent data from the European
Narcolepsy Network showed a very robust association with narcolepsy cases with
cataplexy and DQB1*0602 in multiple European countries (see Table 1.1) (Tafti
et al. 2014). The odds ratio averaged across countries is 251. But the genetic variant
is common occurring in 6.93–24.32% of controls without narcolepsy in different
European countries (Tafti et al. 2014). The average occurrence in controls is 17.68%.
Thus, this is an example of a common variant with a large effect. It makes individ-
uals susceptible to an environmental challenge such as a particular infection or
particular type of influenza vaccine.
DQB1*0602 is, however, not the only genetic risk factor. Given how robust this
effect is makes it difficult to identify other genetic risk factors. Hallmayer et al.
(2009) did an innovative GWAS (see Fig. 1.2) to address this challenge. They
recruited cases with narcolepsy and cataplexy and controls but all had the
DQB1*0602 variant, i.e., in both cases and controls. They did so in multiple ethnic
groups (see Fig. 1.2). They then applied a GWAS analysis and identified variants in
the T cell receptor alpha locus as conferring risk for narcolepsy with cataplexy. The
finding was replicated in most groups but did not replicate in African Americans; this
6 A. I. Pack
Table 1.1 Association in European countries with DQB1*0602 in cases with narcolepsy and
cataplexy
Case-DQB1+N Control-DQBI+N
Country (case, control) (%) (%) OR p
DE (232, 296) 227 (97.84) 72 (24.3.2) 141.24 9.71E-26
CH (66, 473) 65 (98.48) 102 (21.56) 236.42 7.01E-8
NL (323, 469) 318 (98.45) 114 (24.31) 198.05 3.62E-30
PL (63, 197) 63 (100) 44 (22.33) 438.08 2.65E-09
SP (127, 1174) 126 (99.21) 170 (14.48) 744.14 5.25E-11
FR (341, 499) 335 (98.24) 94 (18.84) 240.56 1.18R-37
IT (66, 433) 64 (96.97) 30 (6.93) 429.87 3.21E-16
Mantel-Haenszel (meta- 1198 (98.36) 626 (17.68) 251.12 1.04E-120
analysis)
DE Germany, CH Switzerland, NL Netherlands, PL, Poland, SP Spain, FR France, IT Italy, OR
odds ratio
The frequency of DQB1*0602 in cases and controls with OR for increased risk and the p-value for
the association are shown [From Tafti et al. (2014) with permission]
Fig. 1.2 Design of a genome-wide association study to identify variants other than HLA
DQB1*0602 that confer risk for narcolepsy. To do so, all cases and controls were positive for
this variant. There was an initial discovery phase with 807 cases and 1074 controls that identified
3 SNPs in the T cell alpha locus that had a genome-wide significant association. This finding was
replicated in separate samples of White subjects and Asians but not in African Americans. The latter
had the smallest sample size [From Hallmayer et al. (2009) with permission]
may be the result of the much smaller sample size in this group. This result adds
further evidence that narcolepsy is an autoimmune disorder with a specific antigen-
presenting HLA and a specific subtype of a T cell receptor.
Thus, this initial approach to GWAS led to a significant new understanding of
genetic risk for RLS and narcolepsy/cataplexy. The expense and time-consuming
1 Evolving Approaches to Identifying Genetic Risk Variants for Sleep Disorders 7
nature of the subject recruitment has led to development of new approaches but still
using GWAS, as will now be described.
1.1 UK Biobank
An initial large-scale GWAS examined in one study genetic risks for seven common
diseases using 14,000 cases and 3000 controls (Wellcome Trust Case Control
Consortium 2007). This study was supported by the Wellcome Trust in the United
Kingdom. Subsequently, the Wellcome Trust funded the UK Biobank. The study
recruited approximately 500,000 individuals in the United Kingdom. Participants
provided samples for extraction of DNA, as well as other blood samples, filled out
questionnaires, and their medical records were available in their electronic health
records. Initially, genotyping was done using arrays to assess common SNP variants.
More recently, exome sequencing data are becoming available. Whole-genome
sequencing data will be available in the future. The remarkable aspect of this
program is that all of these data (genetic, phenotypes) are shared with the interna-
tional research community who can apply for access. The UK Biobank provides the
data in a download. They have realized that this requires large-scale computing
resources at institutions receiving the data. They are working to provide mechanisms
for investigators at institutions without such computer resources to do analyses on
the computers provided by the UK Biobank.
The questionnaires used to phenotype individuals are broad and lack depth in any
particular area. There are, however, more in-depth phenotyping in a subset of sub-
jects. Approximately 100,000 individuals were studied with an accelerometer. While
not originally intended to provide estimates of sleep duration, this can be obtained
from the data obtained (van Hees et al. 2018). Approximately the same number of
subjects have had extensive imaging studies, e.g., brain MRIs, and these data can
also be obtained by interested investigators. Currently, there are no data on objec-
tively studied sleep, an area of opportunity.
There is a similar program now being developed in the United States. This was
initially called the Precision Medicine Initiative, now the All of US Program.
The vision is to evaluate multiple chronic diseases and to use wearables to phenotype
the 1.0 million subjects who are to be recruited (Collins and Varmus 2015). Given
the rapid advances in wearables to study sleep (Chinoy et al. 2021), including
continuous remote assessment of EEG, sleep is ideally positioned to be assessed in
these developing efforts.
Despite the current lack of in-depth information on participants in the UK
Biobank, this resource has contributed greatly to elucidating the genetic basis of
sleep and circadian rhythm. There have been successful GWAS for the following:
chronotype (Jones et al. 2019a; Lane et al. 2016; Hu et al. 2016; Kalmbach et al.
2017), sleep duration based on self-report (Jones et al. 2016; Lane et al. 2017; Dashti
et al. 2019; Doherty et al. 2018), sleep duration assessed objectively from data
obtained by accelerometers (Dashti et al. 2019; Doherty et al. 2018; Jones et al.
8 A. I. Pack
2019b), excessive sleepiness (Wang et al. 2019), and insomnia (Lane et al. 2017,
2019; Jansen et al. 2019).
There have been three separate GWAS to assess variants associated with
chronotype (Jones et al. 2019a; Lane et al. 2016; Hu et al. 2016) [for review, see
Kalmbach et al. (2017)]. The studies that have been conducted involved data from
the UK Biobank and 23andMe. The latter is a commercial program that provides for
fee information on an individual’s genotype. For this program, questionnaires have
been used for participants who had provided samples and who agree to be part of
research studies. These GWAS were based on questionnaires. They were not full
chronotype questionnaires such as the Horne and Ostberg (1976) but rather individ-
uals were questioned as to whether they believed they were a morning person, night
person, or neither. All three GWAS reported associations for chronotype for SNPs in
PER2, RGS1E, FBXL13, and AK5 (Jones et al. 2019a; Lane et al. 2016; Hu et al.
2016; Kalmbach et al. 2017). Both PER2 and RGS17 are known to play a role in the
molecular clock.
GWAS assessing self-report sleep duration has consistently shown an association
with PAX8 (Jones et al. 2016; Lane et al. 2017; Dashti et al. 2019). This is a
transcription factor. The genes whose expression is altered by PAX8 that could be
involved in the regulation of sleep are currently unknown. The other variant that has
been consistently identified is VRK2. It is a kinase. Variants of this gene have been
shown to be associated with schizophrenia, major depressive Illness, and genetic
generalized epilepsy (Li and Yue 2018).
Studies with sleep duration determined by accelerometry have not surprisingly
been more fruitful. Heritability estimates of objectively measured sleep (19% [95%
CI: 18.2–19.8%]) are larger than self-report sleep duration (8.8% [95%CI:
8.5–9.0%]) (Jones et al. 2019b). GWAS of objectively measured sleep identified
variants in both studies of this type in DPYD, MEIS1, PAX8, and LOC IO1928419
(Doherty et al. 2018; Jones et al. 2019b). Other variants were identified in one of the
studies (Doherty et al. 2018; Jones et al. 2019b), including BTBD9 and ANK1. As
described earlier, both MEIS1 and BTBD9 variants have been shown to be associ-
ated with RLS (Winkelmann et al. 2007; Stefansson et al. 2007). This raises the
question as to whether there is a large number of individuals in the UK Biobank with
unrecognized RLS.
Another GWAS was conducted on excessive sleepiness (Collins and Varmus
2015). It was based on a single question—how likely are you to doze off or fall
asleep during the daytime when you do not mean to (e.g., when working or driving)?
Forty-two variants were identified to be associated with this response. (It is remark-
able how much can be learned from just one question!) Since little is known about
actual sleep disorders in the UK Biobank, some of these variants may be associated
with specific sleep disorders such as obstructive sleep apnea.
To study insomnia a different question was asked. The largest GWAS to date
included 1,331,010 subjects! This was based on 386,539 from the UK Biobank and
944,471 from 23andMe (Jansen et al. 2019). Two hundred two loci were identified to
be associated with an insomnia phenotype. Both variants in MEIS1 and BTBD9
were among these variants. This may be the result of unrecognized RLS in this
1 Evolving Approaches to Identifying Genetic Risk Variants for Sleep Disorders 9
population. Investigators in this study studied where the genes they identified were
expressed and using databases for gene expression in different brain regions, they
found that these genes were overrepresented among expressed genes in the frontal
cortex, and the anterior cingulate cortex.
Thus, much has been learned from this resource and we can anticipate more
contributions in the future. There are currently large biobanks in China, e.g., the
Kadoorie Biobank of 0.5 million subjects (Chen et al. 2011). It is unclear if they have
information about sleep/chronotype or sleep/circadian disorders. Given the scale of
this biobank, there is great potential for sleep/circadian research in China using this
resource.
Fig. 1.3 Positive (PPV) and negative (NPV) predictive values for algorithm to identify OSA from
data in the EHR. The data shown are estimates and 95% confidence intervals (CI). Data are shown
by aggregating across all sites and for individual sites—Geisinger Clinic, Kaiser Permanente
Southern California (KPSC), Mayo Clinic, Northwestern University, University of Pennsylvania
(Penn), and Vanderbilt University [From Keenan et al. (2020) with permission]
a sample of individuals who met this criteria (cases) and a random sample of controls
who had no diagnostic codes for OSA. We did this validation using random samples
from multiple institutions (see Fig. 1.3). The clinical records of these samples were
examined by trained staff to see whether they were indeed cases or controls. The
majority of cases (72.2%) had sleep study reports that confirmed the diagnosis, or
had evidence of treatment of OSA (22.3%). Only in a minority of cases did we have
to rely on clinical-based notes (5.5%). The validation showed that we had in all
institutions a high positive and negative predictive value using this algorithm.
Averaged across all institutions in our network, the positive and negative predictive
value was over 90%. At Geisinger Clinic the positive predictive value was <90%.
We, therefore, evaluated an algorithm that used 3 occasions with a diagnostic code
for OSA as being a case. This improved the positive predictive value but at the
expense of losing cases, i.e., some cases with a diagnosis on at least two occasions
did not have a third such encounter. The loss was not major at Geisinger Clinic but
was much greater at Penn. This likely reflects that Penn is very much a tertiary care
health system. Thus, investigators using this approach should seek to validate the
algorithm they plan to use in their biobank.
There has currently been active development of these algorithms for multiple
disorders. Fifty three such algorithms that use a combination of clinical variables to
define a particular phenotype are in a public repository—Phenotype Knowledge
Base (PheKB) (Kirby et al. 2016).
While many of these algorithms use CPT codes, as we have done for OSA
(Keenan et al. 2020), these codes are intended primarily for billing purposes. They
are, therefore suboptimal to define a specific disease (Hripcsak and Albers 2013).
Thus, information from CPT codes can be amplified with other clinical information
1 Evolving Approaches to Identifying Genetic Risk Variants for Sleep Disorders 11
such as results of laboratory tests. This will add specificity. The example described
for OSA could use, in addition to CPT codes, information from clinical sleep studies.
While using CPT codes has worked well for OSA (Keenan et al. 2020), algo-
rithms based solely on diagnostic codes may not work for all diagnoses. We have
worked with our colleagues at Geisinger Clinic to develop a similar algorithm for
Alzheimer’s disease (AD) (unpublished observations). We found that for those
individuals evaluated in specialized clinics an algorithm worked well. They had
extensive testing including brain imaging, neurocognitive tests, and studies of bio-
markers. However, there are a large number of individuals where the diagnosis of
AD was made by primary care physicians without such tests. It is, therefore, unclear
if these individuals have AD. Thus, for AD, an algorithm that is based not only on
CPT codes but also on data from other tests is required.
Recently, as described in an excellent review (Li et al. 2020), use is now being
made of machine learning approaches to derive specific phenotypes based on
multiple sources of data in the EHR (Banda et al. 2018). A machine learning
approach has been used to define an algorithm to identify type 2 diabetes mellitus
using EHR data (Zheng et al. 2017).
These approaches need to be replicated in EHR data from multiple institutions.
This requires efforts at standardization of data. EHR data can be mapped to common
data models, e.g., the Observational Medical Outcomes Partnership (OMOP) stan-
dardized vocabulary (Stang et al. 2010). A recent review has developed the argument
that there needs to be systematic adoption of standardized terminologies in all areas
of sleep medicine, with development of an integrated infrastructure (Mazzotti 2021).
The approach to identifying OSA in EHR data has allowed us to identify more
than 20,000 cases with OSA who have genotype data. This permits us to do a very
large GWAS. It permits us not only to do case/control analysis, but we can also
obtain quantitative data relevant to OSA from sleep study reports and actual raw
sleep study data for additional analyses. Thus, large-scale quantitative trait analyses
can also be conducted.
A similar approach to case/control analyses has already been published from the
large FinnGen program in Finland (Strausz et al. 2021). They identified OSA from
inpatient records in Finland and from death registry data. Thus, it is a somewhat
biased sample since most OSA are determined in outpatient studies. Moreover, as
pointed out in an editorial (Wang et al. 2021) that accompanied this report, there was
no ability to do quantitative analyses.
This important study in Finland identified several obesity-related genes that
conferred risk for OSA (see Fig. 1.4). By doing analyses when controlling for
BMI or not, it can be shown that some variants such as in FTO (fat mass obesity
gene) are in the obesity pathway to OSA, other variants that were identified are in the
non-obesity pathway (see Fig. 1.4) (Strausz et al. 2021). Patel et al. had previously
proposed that the way to think about genetics of OSA was in terms of obesity- and
non-obesity-related pathways (Patel 2005).
The Finnish group has developed a polygenic risk score for OSA (Strausz et al.
2021). A polygenic risk score for insomnia has also been developed based on
findings from the large GWAS described above. This is one of the goals of
12 A. I. Pack
Fig. 1.4 Manhattan plots for GWAS of OSA with 16,761 cases and 201,194 controls. The x-axis
shows the position of each variant across all the chromosomes and the y-axis the log10 p-value for
association. The dashed line on each plot indicates the log p-value for genome-wide significance,
i.e., after correcting for multiple comparisons. The top panel (a) shows the results for uncorrected
analyses while the bottom panel (b) shows the results after controlling for BMI. The latter analysis
had fewer subjects (12,759 cases and 146,972 controls). Most of the genome-wide significant
associations, including FTO (fat mass and obesity-associated protein), are no longer significant after
controlling for BMI (see panel b). One variant, i.e., RMST/NEDD1 (rhabdomyosarcoma 2 associ-
ated transcript/NEDD1 γ tubulin ring complex targeting factor), continues to be significant after
correcting for BMI [From Strausz et al. (2021) with permission]
GWAS and related genetic studies, i.e., to identify individuals with high genetic risk
for the disorder (PREDICT) and allow clinicians to move from treatment of the
disorder to PREVENTION. For OSA, prevention may involve the future use of
mandibular advancement devices in such individuals to prevent the slow progression
of the disorder (Newman et al. 2005).
But PRS are only one source of data for prediction of risk. Prediction can be
improved by using additional sources of data such as family history and relevant
environmental exposures (Li et al. 2020). The iCARE package jointly models data
from multiple sources to define relative and absolute risk and risk factor distributions
1 Evolving Approaches to Identifying Genetic Risk Variants for Sleep Disorders 13
(Pal Choudhury et al. 2020). Currently, disease risk predictions using PRS are
starting to be applied clinically, in particular, for psychiatric disorders (Kember
et al. 2021). For example, PRS have been shown to be significantly associated
with risk for schizophrenia (Zheutlin et al. 2019).
Currently, however, these polygenic risk scores both for OSA and for insomnia
only explain a small proportion of the genetic variance. There is missing heritability.
This is related to several issues. First, GWAS does not necessarily identify the
causative variant. There are other gene variants that are correlated. Current
approaches also do not address the interactive effects of different gene variants,
i.e., epistasis. There are also other sources of genetic variations. This includes rare
variants (there are as described earlier many of these) and copy number variants
(CNVs) (Conrad et al. 2010; Redon et al. 2006). There are also epigenetic modifi-
cations that alter gene transcription (Murphy and Mill 2014). For many disorders
there are ongoing efforts to address all of these issues thereby improving prediction.
Another approach that EHR data can be used for is PheWAS (Verma et al. 2019;
Bush et al. 2016). This is based on seeking an association with a specific gene variant
and all diagnoses in the EHR. This can serve to replicate previously described
associations and potentially identify pathophysiological mechanisms. For example,
a variant associated with OSA might also be associated with nasal septal deviation
indicating why this variant is a risk variant for OSA. We have conducted a PheWAS
on variants shown to be associated with OSA (Veatch et al. 2020). We did not
replicate an association with OSA for the majority of previously described associ-
ations. SNPs in LEPR, MMP-9, and GABBR1 validated for an association with a
diagnosis of OSA in European Americans from two different clinic-based biobanks
at Geisinger Clinic and Vanderbilt. LEPR encodes for a receptor for the adipocyte-
specific leptin hormone. This SNP is not associated, however, with obesity (Gozal
et al. 2016; Olza et al. 2017; Rojano-Rodriguez et al. 2016; Chavarria-Avila et al.
2015). GABBR1 encodes for a receptor for GABA, an inhibitory neurotransmitter.
For many existing biobanks, there is increasing information on other sources of
genetic variation, in particular exome sequence data. Eventually, these biobanks will
have whole-genome sequence data. Extensive exome sequence data are available in
the UK Biobank, and in the biobanks at Geisinger Clinic and the University of
Pennsylvania. One of the first biobanks to assess exome sequence data was at the
Geisinger Clinic. This clinic provides care to over 3.0 million individuals in a rural
areas of Pennsylvania. Individuals obtaining care from this clinic seldom move.
Thus, there is less than 1% loss of individuals from this system. Individuals who use
this system for their care do so from birth to death. Thus, it allows long-term follow-
up of individuals. One can examine longitudinal change in laboratory values and in
key measures such as blood pressure. This is part of what has been described as a
learning health system (Williams et al. 2018). In the first description of exome
sequence data from this biobank (Dewey et al. 2016), it was shown that 92% of
genes have a rare variant and 7% of sequenced genes are predicted to have a
homozygous loss of function variant of that gene. Although the variants are rare,
the average human carries 21 predicted loss of function rare variants. Using exome
sequencing, data can be particularly valuable to identify relevant genes when there
14 A. I. Pack
are extreme phenotypes. Extreme phenotypes of OSA have been described, e.g.,
severe OSA in individuals with a low likelihood of OSA based on age, gender, and
BMI (Rizzatti et al. 2020).
Availability of exome sequence data also allows a new strategy, i.e., callbacks.
With this approach, one can identify individuals in the biobank with a predicted loss
of function variants of genes of interest and then determine their phenotype. This
may be done using data already available from clinical studies, e.g., laboratory
values, such as sleep studies and images. They can also be recontacted for
in-depth phenotyping for specific areas of focus. For example, if a gene is thought
to be involved in sleep regulation, individuals with a predicted loss of function of
this gene could have their sleep assessed. This general approach has been used in
subjects in the Human Knock Out Project (Saleheen et al. 2017), i.e., to determine
differences in phenotype in individuals with loss of function of specific genes. This
resource is based on studies in Pakistan where there is a high level of consanguin-
eous marriage such that many individuals have homozygous loss of function variants
(Saleheen et al. 2017).
PheWAS can also be done with exome sequence data as was described above for
common variants (Verma et al. 2019; Bush et al. 2016). A recent major publication
did a genome-wide study looking at association with all rare variants and all
diagnoses in the EHR (Park et al. 2021). The study replicated known associations
such as rare variants of BRAC and breast cancer as well as new associations (Park
et al. 2021) (see Fig. 1.5).
Since each individual variant is rare, loss of function variants in a single gene or
targeted set of genes were aggregated, i.e., they conducted a gene burden PheWAS.
They identified 97 gene burdens with association with specific phenotypes at
p < 10 6. The initial analysis was using data from the biobank at Penn. They sought
to replicate the findings in other clinical biobanks, e.g., at Geisinger, and in the UK
Biobank. They found more replication with data from other health system biobanks
than with data from the UK Biobank. The UK Biobank is recognized to have a
healthy volunteer selection bias (Fry et al. 2017). There is, therefore, a lower
prevalence of specific disorders in the UK Biobank compared to biobanks developed
by health systems. For sleep disorders, there is the added complication that partic-
ipants in the UK Biobank may have undiagnosed sleep disorders such as RLS and
OSA. The prevalence of OSA in the UK Biobank is much less than the estimated
prevalence based on age, gender, and BMI (Peppard et al. 2013) (unpublished
observations).
Another key resource that has been developed is the Trans-Omics for Precision
Medicine (TOPMed) (Burgess 2021) that is supported by the National Heart, Lung
and Blood Institute. This is an innovative grant mechanism that has allowed a major
resource for genetic studies to be developed. Applicants for this program apply for
Fig. 1.5 The plot shows the landscape of gene–phenotype associations across the exome and phenotype data from the Penn Medicine Biobank. The x-axis
represents the location across the genome. The log10 of the p-value is shown for the association in the y-axis. The association for each of the 97 genome burdens
1 Evolving Approaches to Identifying Genetic Risk Variants for Sleep Disorders
with 1000 phecodes is shown. Colors are used to represent different phecode groups [From Park et al. (2021) with permission]
15
16 A. I. Pack
grants that do not provide support for the efforts of the applicant, but rather provide
resources to do analyses of samples provided by the investigators who apply. These
analyses include whole-genome sequencing (WGS), whole-genome DNA methyla-
tion, and metabolomics. Much of the effort has gone into whole-genome sequencing.
The initial results from analysis of WGS in 53,831 individuals have recently been
reported (Taliun et al. 2021). More than 400 million variants were detected. A large
number of rare variants that occur in <1% of individuals were found. Given the scale
of this effort, data from TOPMed will likely be used as the reference genome for
future genetic studies.
Currently, in TOPMed there are somewhat limited data from individuals with
sleep disorders. These largely come from sleep studies that have been performed on
individuals in population-based cohort studies (Zhang et al. 2018). Many of these
cohorts were developed to assess variables, including genetic variants that are
associated with cardiovascular risk. But some have argued that sleep apnea identified
in population-based studies is different from that found in patients who present
clinically (Arnardottir et al. 2016). The data from sleep studies obtained in these
cohorts is available from the National Sleep Research Resource (Zhang et al. 2018).
This contains data on 26,808 subjects and there are 31,166 files of sleep studies
available in the European Data Format (EDF). These data can be obtained by
interested investigators. Genetic analysis using these data with WGS generated in
TOPMed has shown that six coding and 51 noncoding variants in the gene for
GTPase-activating proteins (DLC1) are associated with average oxygen saturation
during sleep (Liang et al. 2019). Recently, new data are being added to TOPMed
from a sample of 3000 clinical patients with OSA.
1.4 Conclusion
The identification of gene variants conferring risk for specific sleep disorders is
accelerating. The approach is moving from the laborious recruitment of large
numbers of cases and controls to use of resources that have been developed in
different countries to make investigation of genetic variants more efficient. Exten-
sive use is being made of data from electronic health records as part of the
phenotyping strategy. Ultimately, however, we need to prove that the variants
identified are indeed casual. This will require functional studies not only using
assays in specific cells but also use of model systems such as zebrafish and mice.
The efforts currently underway in Europe and the USA could be duplicated in China,
thereby further accelerating progress in this area.
1 Evolving Approaches to Identifying Genetic Risk Variants for Sleep Disorders 17
References
Allen RP, Picchietti D, Hening WA, Trenkwalder C, Walters AS, Montplaisi J, et al. Restless legs
syndrome: diagnostic criteria, special considerations, and epidemiology. A report from the
restless legs syndrome diagnosis and epidemiology workshop at the National Institutes of
Health. Sleep Med. 2003;4(2):101–19.
Arnardottir ES, Bjornsdottir E, Olafsdottir KA, Benediktsdottir B, Gislason T. Obstructive sleep
apnoea in the general population: highly prevalent but minimal symptoms. Eur Respir
J. 2016;47(1):194–202.
Banda JM, Seneviratne M, Hernandez-Boussard T, Shah NH. Advances in electronic phenotyping:
from rule-based definitions to machine learning models. Annu Rev Biomed Data Sci. 2018;1:
53–68.
Burgess DJ. The TOPMed genomic resource for human health. Nat Rev Genet. 2021;22(4):200.
Bush WS, Oetjens MT, Crawford DC. Unravelling the human genome-phenome relationship using
phenome-wide association studies. Nat Rev Genet. 2016;17(3):129–45.
Chavarria-Avila E, Vazquez-Del Mercado M, Gomez-Banuelos E, Ruiz-Quezada SL, Castro-
Albarran J, Sanchez-Lopez L, et al. The impact of LEP G-2548A and LEPR Gln223Arg
polymorphisms on adiposity, leptin, and leptin-receptor serum levels in a Mexican Mestizo
population. Biomed Res Int. 2015;2015:539408.
Chen Z, Chen J, Collins R, Guo Y, Peto R, Wu F, et al. China Kadoorie Biobank of 0.5 million
people: survey methods, baseline characteristics and long-term follow-up. Int J Epidemiol.
2011;40(6):1652–66.
Chinoy ED, Cuellar JA, Huwa KE, Jameson JT, Watson CH, Bessman SC, et al. Performance of
seven consumer sleep-tracking devices compared with polysomnography. Sleep. 2021;44(5):
zsaa291.
Collins FS, Varmus H. A new initiative on precision medicine. N Engl J Med. 2015;372(9):793–5.
Conrad DF, Pinto D, Redon R, Feuk L, Gokcumen O, Zhang Y, et al. Origins and functional impact
of copy number variation in the human genome. Nature. 2010;464(7289):704–12.
Dashti HS, Jones SE, Wood AR, Lane JM, van Hees VT, Wang H, et al. Genome-wide association
study identifies genetic loci for self-reported habitual sleep duration supported by
accelerometer-derived estimates. Nat Commun. 2019;10(1):1100.
Dewey FE, Murray MF, Overton JD, Habegger L, Leader JB, Fetterolf SN, et al. Distribution and
clinical impact of functional variants in 50,726 whole-exome sequences from the DiscovEHR
study. Science. 2016;354(6319):aaf6814.
Doherty A, Smith-Byrne K, Ferreira T, Holmes MV, Holmes C, Pulit SL, et al. GWAS identifies
14 loci for device-measured physical activity and sleep duration. Nat Commun. 2018;9(1):5257.
Fry A, Littlejohns TJ, Sudlow C, Doherty N, Adamska L, Sprosen T, et al. Comparison of
sociodemographic and health-related characteristics of UK Biobank participants with those of
the general population. Am J Epidemiol. 2017;186(9):1026–34.
Gottesman O, Kuivaniemi H, Tromp G, Faucett WA, Li R, Manolio TA, et al. The electronic
medical records and genomics (eMERGE) network: past, present, and future. Genet Med.
2013;15(10):761–71.
Gozal D, Ham SA, Mokhlesi B. Sleep apnea and cancer: analysis of a nationwide population
sample. Sleep. 2016;39(8):1493–500.
Hallmayer J, Faraco J, Lin L, Hesselson S, Winkelmann J, Kawashima M, et al. Narcolepsy is
strongly associated with the T-cell receptor alpha locus. Nat Genet. 2009;41(6):708–11.
Horne JA, Ostberg O. A self-assessment questionnaire to determine morningness-eveningness in
human circadian rhythms. Int J Chronobiol. 1976;4(2):97–110.
Hripcsak G, Albers DJ. Next-generation phenotyping of electronic health records. J Am Med
Inform Assoc. 2013;20(1):117–21.
Hu Y, Shmygelska A, Tran D, Eriksson N, Tung JY, Hinds DA. GWAS of 89,283 individuals
identifies genetic variants associated with self-reporting of being a morning person. Nat
Commun. 2016;7:10448.
18 A. I. Pack
Jansen PR, Watanabe K, Stringer S, Skene N, Bryois J, Hammerschlag AR, et al. Genome-wide
analysis of insomnia in 1,331,010 individuals identifies new risk loci and functional pathways.
Nat Genet. 2019;51(3):394–403.
Jones SE, Tyrrell J, Wood AR, Beaumont RN, Ruth KS, Tuke MA, et al. Genome-wide association
analyses in 128,266 individuals identifies new morningness and sleep duration loci. PLoS
Genet. 2016;12(8):e1006125.
Jones SE, Lane JM, Wood AR, van Hees VT, Tyrrell J, Beaumont RN, et al. Genome-wide
association analyses of chronotype in 697,828 individuals provides insights into circadian
rhythms. Nat Commun. 2019a;10(1):343.
Jones SE, van Hees VT, Mazzotti DR, Marques-Vidal P, Sabia S, van der Spek A, et al. Genetic
studies of accelerometer-based sleep measures yield new insights into human sleep behaviour.
Nat Commun. 2019b;10(1):1585.
Juji T, Satake M, Honda Y, Doi Y. HLA antigens in Japanese patients with narcolepsy. All the
patients were DR2 positive. Tissue Antigens. 1984;24(5):316–9.
Kalmbach DA, Schneider LD, Cheung J, Bertrand SJ, Kariharan T, Pack AI, et al. Genetic basis of
chronotype in humans: insights from three landmark GWAS. Sleep. 2017;40(2):zsw048.
Keenan BT, Kirchner HL, Veatch OJ, Borthwick KM, Davenport VA, Feemster JC, et al. Multisite
validation of a simple electronic health record algorithm for identifying diagnosed obstructive
sleep apnea. J Clin Sleep Med. 2020;16(2):175–83.
Kember RL, Merikangas AK, Verma SS, Verma A, Judy R, Regeneron Genetics C, et al. Polygenic
Risk of Psychiatric Disorders Exhibits Cross-trait Associations in Electronic Health Record
Data From European Ancestry Individuals. Biol Psychiatry. 2021;89(3):236–45.
Kemlink D, Polo O, Frauscher B, Gschliesser V, Hogl B, Poewe W, et al. Replication of restless
legs syndrome loci in three European populations. J Med Genet. 2009;46(5):315–8.
Kirby JC, Speltz P, Rasmussen LV, Basford M, Gottesman O, Peissig PL, et al. PheKB: a catalog
and workflow for creating electronic phenotype algorithms for transportability. J Am Med
Inform Assoc. 2016;23(6):1046–52.
Klein RJ, Zeiss C, Chew EY, Tsai JY, Sackler RS, Haynes C, et al. Complement factor H
polymorphism in age-related macular degeneration. Science. 2005;308(5720):385–9.
Lane JM, Vlasac I, Anderson SG, Kyle SD, Dixon WG, Bechtold DA, et al. Genome-wide
association analysis identifies novel loci for chronotype in 100,420 individuals from the UK
Biobank. Nat Commun. 2016;7:10889.
Lane JM, Liang J, Vlasac I, Anderson SG, Bechtold DA, Bowden J, et al. Genome-wide association
analyses of sleep disturbance traits identify new loci and highlight shared genetics with
neuropsychiatric and metabolic traits. Nat Genet. 2017;49(2):274–81.
Lane JM, Jones SE, Dashti HS, Wood AR, Aragam KG, van Hees VT, et al. Biological and clinical
insights from genetics of insomnia symptoms. Nat Genet. 2019;51(3):387–93.
Li M, Yue W. VRK2, a candidate gene for psychiatric and neurological disorders. Mol Neuropsy-
chiatry. 2018;4(3):119–33.
Li R, Chen Y, Ritchie MD, Moore JH. Electronic health records and polygenic risk scores for
predicting disease risk. Nat Rev Genet. 2020;21(8):493–502.
Liang J, Cade BE, He KY, Wang H, Lee J, Sofer T, et al. Sequencing analysis at 8p23 identifies
multiple rare variants in DLC1 associated with sleep-related oxyhemoglobin saturation level.
Am J Hum Genet. 2019;105(5):1057–68.
MacArthur J, Bowler E, Cerezo M, Gil L, Hall P, Hastings E, et al. The new NHGRI-EBI Catalog of
published genome-wide association studies (GWAS Catalog). Nucleic Acids Res. 2017;45(D1):
D896–901.
Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ, et al. Finding the missing
heritability of complex diseases. Nature. 2009;461(7265):747–53.
Mazzotti DR. Landscape of biomedical informatics standards and terminologies for clinical sleep
medicine research: a systematic review. Sleep Med Rev. 2021;60:101529.
1 Evolving Approaches to Identifying Genetic Risk Variants for Sleep Disorders 19
Moore H, Winkelmann J, Lin L, Finn L, Peppard P, Mignot E. Periodic leg movements during sleep
are associated with polymorphisms in BTBD9, TOX3/BC034767, MEIS1, MAP2K5/SKOR1,
and PTPRD. Sleep. 2014;37(9):1535–42.
Murphy TM, Mill J. Epigenetics in health and disease: heralding the EWAS era. Lancet. 2014;383
(9933):1952–4.
Newman AB, Foster G, Givelber R, Nieto FJ, Redline S, Young T. Progression and regression of
sleep-disordered breathing with changes in weight: the Sleep Heart Health Study. Arch Intern
Med. 2005;165(20):2408–13.
Olza J, Ruperez AI, Gil-Campos M, Leis R, Canete R, Tojo R, et al. Leptin receptor gene variant
rs11804091 is associated with BMI and insulin resistance in Spanish female obese children: a
case-control study. Int J Mol Sci. 2017;18(8):1690.
Pack AI, Keenan BT, Gehrman PR, Justice AE. Genetics and genomic basis of sleep disorders in
humans. In: Kryger M, editor. Principles and practices of sleep medicine. Philadelphia, PA:
Elsevier; 2021.
Pal Choudhury P, Maas P, Wilcox A, Wheeler W, Brook M, Check D, et al. iCARE: an R package
to build, validate and apply absolute risk models. PLoS One. 2020;15(2):e0228198.
Park J, Lucas AM, Zhang X, Chaudhary K, Cho JH, Nadkarni G, et al. Exome-wide evaluation of
rare coding variants using electronic health records identifies new gene-phenotype associations.
Nat Med. 2021;27(1):66–72.
Patel SR. Shared genetic risk factors for obstructive sleep apnea and obesity. J Appl Physiol.
2005;99(4):1600–6.
Peppard PE, Young T, Barnet JH, Palta M, Hagen EW, Hla KM. Increased prevalence of sleep-
disordered breathing in adults. Am J Epidemiol. 2013;177(9):1006–14.
Redon R, Ishikawa S, Fitch KR, Feuk L, Perry GH, Andrews TD, et al. Global variation in copy
number in the human genome. Nature. 2006;444(7118):444–54.
Rizzatti FG, Mazzotti DR, Mindel J, Maislin G, Keenan BT, Bittencourt L, et al. Defining extreme
phenotypes of OSA across international sleep centers. Chest. 2020;158(3):1187–97.
Rojano-Rodriguez ME, Beristain-Hernandez JL, Zavaleta-Villa B, Maravilla P, Romero-
Valdovinos M, Olivo-Diaz A. Leptin receptor gene polymorphisms and morbid obesity in
Mexican patients. Hereditas. 2016;153:2.
Saleheen D, Natarajan P, Armean IM, Zhao W, Rasheed A, Khetarpal SA, et al. Human knockouts
and phenotypic analysis in a cohort with a high rate of consanguinity. Nature. 2017;544(7649):
235–9.
Stang PE, Ryan PB, Racoosin JA, Overhage JM, Hartzema AG, Reich C, et al. Advancing the
science for active surveillance: rationale and design for the Observational Medical Outcomes
Partnership. Ann Intern Med. 2010;153(9):600–6.
Stefansson H, Rye DB, Hicks A, Petursson H, Ingason A, Thorgeirsson TE, et al. A genetic risk
factor for periodic limb movements in sleep. N Engl J Med. 2007;357(7):639–47.
Strausz S, Ruotsalainen S, Ollila HM, Karjalainen J, Kiiskinen T, Reeve M, et al. Genetic analysis
of obstructive sleep apnoea discovers a strong association with cardiometabolic health. Eur
Respir J. 2021;57:2003091.
Tafti M, Hor H, Dauvilliers Y, Lammers GJ, Overeem S, Mayer G, et al. DQB1 locus alone
explains most of the risk and protection in narcolepsy with cataplexy in Europe. Sleep. 2014;37
(1):19–25.
Taliun D, Harris DN, Kessler MD, Carlson J, Szpiech ZA, Torres R, et al. Sequencing of 53,831
diverse genomes from the NHLBI TOPMed Program. Nature. 2021;590(7845):290–9.
van Hees VT, Sabia S, Jones SE, Wood AR, Anderson KN, Kivimaki M, et al. Estimating sleep
parameters using an accelerometer without sleep diary. Sci Rep. 2018;8(1):12975.
Veatch OJ, Bauer CR, Keenan BT, Josyula NS, Mazzotti DR, Bagai K, et al. Characterization of
genetic and phenotypic heterogeneity of obstructive sleep apnea using electronic health records.
BMC Med Genet. 2020;13(1):105.
Verma A, Bang L, Miller JE, Zhang Y, Lee MTM, Zhang Y, et al. Human-disease phenotype map
derived from PheWAS across 38,682 individuals. Am J Hum Genet. 2019;104(1):55–64.
20 A. I. Pack
Visscher PM, Brown MA, McCarthy MI, Yang J. Five years of GWAS discovery. Am J Hum
Genet. 2012;90(1):7–24.
Wang H, Lane JM, Jones SE, Dashti HS, Ollila HM, Wood AR, et al. Genome-wide association
analysis of self-reported daytime sleepiness identifies 42 loci that suggest biological subtypes.
Nat Commun. 2019;10(1):3503.
Wang H, Goodman MO, Sofer T, Redline S. Cutting the fat: advances and challenges in sleep
apnoea genetics. Eur Respir J. 2021;57(5):2004644.
Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of
seven common diseases and 3,000 shared controls. Nature. 2007;447(7145):661–78.
Welter D, MacArthur J, Morales J, Burdett T, Hall P, Junkins H, et al. The NHGRI GWAS Catalog,
a curated resource of SNP-trait associations. Nucleic Acids Res. 2014;42(Database issue):
D1001–6.
Williams MS, Buchanan AH, Davis FD, Faucett WA, Hallquist MLG, Leader JB, et al. Patient-
centered precision health in a learning health care system: Geisinger’s Genomic Medicine
Experience. Health Aff (Millwood). 2018;37(5):757–64.
Winkelmann J, Schormair B, Lichtner P, Ripke S, Xiong L, Jalilzadeh S, et al. Genome-wide
association study of restless legs syndrome identifies common variants in three genomic regions.
Nat Genet. 2007;39(8):1000–6.
Yang Q, Li L, Chen Q, Foldvary-Schaefer N, Ondo WG, Wang QK. Association studies of variants
in MEIS1, BTBD9, and MAP2K5/SKOR1 with restless legs syndrome in a US population.
Sleep Med. 2011;12(8):800–4.
Zhang GQ, Cui L, Mueller R, Tao S, Kim M, Rueschman M, et al. The National Sleep Research
Resource: towards a sleep data commons. J Am Med Inform Assoc. 2018;25(10):1351–8.
Zheng T, Xie W, Xu L, He X, Zhang Y, You M, et al. A machine learning-based framework to
identify type 2 diabetes through electronic health records. Int J Med Inform. 2017;97:120–7.
Zheutlin AB, Dennis J, Karlsson Linner R, Moscati A, Restrepo N, Straub P, et al. Penetrance and
pleiotropy of polygenic risk scores for schizophrenia in 106,160 patients across four health care
systems. Am J Psychiatry. 2019;176(10):846–55.
Chapter 2
Neurobiology of Sleep–Wake Control
Leszek Kubin
2.1 Introduction
Three major discoveries and the associated fundamental concepts have shaped our
current understanding of the basic neurobiology of sleep–wake states. Chronologi-
cally, they were: (a) the recognition that sleep is actively generated within a
L. Kubin (*)
Department of Biomedical Sciences 209E/VET, School of Veterinary Medicine, University of
Pennsylvania, Philadelphia, PA, USA
e-mail: lkubin@vet.upenn.edu
© Springer Nature B.V. and Shanghai Jiao Tong University Press 2022 21
A. I. Pack, Q. Y. Li (eds.), Sleep and its Disorders, Translational Medicine Research,
https://doi.org/10.1007/978-94-024-2168-2_2
22 L. Kubin
delimited part of the forebrain (von Economo 1930); (b) the determination that what
has been earlier seen as a single state of sleep, comprises two distinct entities, the
non-Rapid Eye Movement (NREM) sleep and REM sleep (Aserinsky and Kleitman
1953); and (c) introduction of a model of the relationship between the circadian and
homeostatic regulations of sleep, commonly referred to as the two-process model
(Borbély 1982).
Prior to the groundbreaking observations of von Economo (1930), sleep was
commonly regarded as a passive state resulting from a temporal suspension of the
active processes that maintain wakefulness and everything it entails, including the
ability to perceive and process external stimuli, respond to them emotionally and
physically, and create a memory of past events. Indeed, equating sleep with death
was not uncommon in pre-twentieth-century literature in both Europe and Asia (e.g.,
Dante’s Divine Comedy). The passive nature of sleep did not mean, however, that the
“refreshing” and “strength-restoring” qualities of sleep were not recognized. For
example, to account for the common observation that a sumptuous meal promotes
sleepiness, Aristotle considered a causal relationship between the occurrence of
sleep and accumulation of nutrients. Along similar lines, in ancient China, sleep
was attributed to alterations in the composition of circulating blood (Inoue et al.
1995). Hence, the modern concepts of sleep as a restorative process evolved from
ancient science and philosophy.
Analogously to the identification of the anterior and posterior parts of the
hypothalamus as the key sites for the generation of sleep and wakefulness, respec-
tively, the formal recognition of REM sleep as a distinct behavioral state (Aserinsky
and Kleitman 1953) was followed by the finding that the neuroanatomical origin of
the state is in the pontomedullary reticular formation, with the dorsomedial pontine
tegmentum being the key site (Jouvet 1962). This, and the finding that mesopontine
acetylcholine and amines (serotonin and norepinephrine) exert opposing effects on
REM sleep, has led to the formulation of the “reciprocal interaction model,” which
emphasized the mutually inhibitory interaction between pontine aminergic neurons
(especially those containing serotonin and noradrenergic neurons of the locus
coeruleus, a.k.a. the catecholaminergic A6 group) and those containing acetylcho-
line (specifically, the Ch5 and Ch6 groups) (McCarley and Hobson 1975). Since its
original publication, the model has been modified and expanded, with some of its
core assumptions being questioned (Lu et al. 2006; Luppi et al. 2012; Reiner 1995).
In addition, following the discovery of the excitatory peptides, orexins
(a.k.a. hypocretins), for which the posterior hypothalamus is the only source in the
brain (de Lecea et al. 1998) and whose absence results in narcolepsy-cataplexy, a
disorder of REM sleep (Chemelli et al. 1999), attention has considerably shifted
from the brainstem to the hypothalamus and its role in the control of REM sleep.
Nevertheless, the reciprocal interaction model, as originally proposed, stimulated
studies of cellular behaviors in relation to the stages of sleep and wakefulness that
continue to this day and yield novel findings [for reviews, see Hassani et al. (2009a)
and Hobson et al. (1986)].
The basic principle of the interaction between the circadian and homeostatic
regulations of sleep illustrated in Fig. 2.1 was widely adapted in sleep medicine
2 Neurobiology of Sleep–Wake Control 23
Fig. 2.1 In its simplest form, the interaction between the drive for sleep and the central circadian
clock can be visualized as a circadian gate that prevents entry into sleep (promotes wakefulness)
over a pre-set portion of the 24 h circadian cycle. In diurnal species, the circadian system actively
supports wakefulness during the day, thereby effectively disallowing sleep despite the progressive
accumulation of sleep drive with time spent awake. During the night, when sleep can actually occur,
the homeostatic drive for sleep is gradually dissipated
Fig. 2.2 The basic rest–activity cycle (BRAC) (Kleitman 1963, 1982) is, after circadian rhythm,
the second major endogenous rhythm that determines the timing and pattern of sleep during the
night and waxing and waning of attention during the day. While the neurobiological basis of this
ultradian rhythm remains to be identified, its presence is strongly manifest across the entire
circadian cycle. During the active phase of the circadian cycle, BRAC determines the periodic
variation in attention; during the rest/sleep phase, BRAC is responsible for rhythmic cycling
between successive substages of NREM sleep (NREM 1–4) and REM sleep
26 L. Kubin
influence the timing of transitions from NREM sleep to REM sleep and also the
durations and “intensities” of individual episodes of wakefulness, NREM and REM
sleep (Lavie 1991; McPartland and Kupfer 1978). If BRAC is the clock for the
wake-NREM-REM sleep cycle, it is plausible that the sleep–wake cycle is generated
by a three-, rather than two-, phase oscillator. If so, this would be analogous to the
central pattern generator for breathing. As with the sleep–wake rhythm, the respira-
tory generator was originally seen as a bistable oscillator that alternated between
inspiration and expiration. However, it then became recognized that the original
expiratory phase consisted of two functionally distinct sub-phases, the post-
inspiratory phase, and active expiratory phase [for reviews, see Feldman et al.
(2003) and Richter and Spyer (2001)]. Accordingly, it may be productive to see
the sleep–wake cycle as a three-phase cycle (wake–NREM sleep–REM sleep), rather
than a process resulting from an interaction between two relatively independent
bistable and reciprocally organized oscillators, one for NREM sleep and wakeful-
ness, and the other for REM sleep and wakefulness (or REM and NREM sleep).
Although recent models include connections between such two bistable oscillators
(Saper et al. 2010) (see also Sect. 2.4.2), a fully integrated three-phase model of the
sleep–wake cycle remains to be developed. It is also of note that, in humans, four
phases of NREM sleep are often distinguished and referred to as NREM 1 through
NREM 4, of these NREM 3–4 can be also collectively named slow–wake sleep
(SWS); in animals, the entire period of NREM sleep is referred to as SWS.
Thus, the baseline sleep–wake behavior is shaped by two rhythmic processes, one
derived from the endogenous circadian clocks and the other from BRAC. Figure 2.3
shows one nearly full circadian cycle of natural sleep–wake behavior in the rat
which, together with mice, is the most commonly used vertebrate model for studies
on the regulation of sleep. Both the day–night difference in the amounts of sleep and
wake and the recurrent sequential occurrence of wake, SWS and REM sleep are well
expressed in this typical record.
Identification of the hypothalamus as a major center for the maintenance of sleep and
vigilance and the pons as the site for the generation of REM sleep by means of brain
lesion and transection experiments enabled and focused the search for specific
groups and types of neurons responsible for the generation of sleep–wake behavior.
These studies, conducted mainly by means of electrophysiological recordings from
single cells and to some extent using c-Fos immunohistochemistry as indirect means
of assessing cell activity (Basheer et al. 1997; Boissard et al. 2002; Dentico et al.
2 Neurobiology of Sleep–Wake Control
Fig. 2.3 Polygraphic record of natural sleep–wake behavior during a period of 23 h in a freely behaving rat. The example illustrates how the circadian,
ultradian, and homeostatic processes converge and collectively control the rest–activity and sleep–wake rhythms. Successive bouts of slow-wave sleep (SWS)
are marked by periods of high-amplitude signal in cortical electroencephalogram (EEG) (top trace). The second trace from the top shows EEG power in delta (Δ)
range (<4 Hz), which characteristically dominates during SWS. The third trace shows the ratio of EEG powers in β2 range (12–14 Hz) and Δ2 range (0.5–2 Hz),
which in rodents greatly increases during each transition from SWS to rapid eye movement sleep (REMS). The fourth trace shows integrated electromyogram
(EMG) of dorsal neck muscles. It is high during wakefulness (W) and lowest during REMS. The bottom trace shows the hypnogram for this recording session.
The following characteristic features are noteworthy: (1) the circadian difference in the amount of SWS between the rest period and active period (marked by the
bar below the record); (2) the semi-rhythmic occurrence of the SWS-REMS-W cycle, reflecting the modulation of the sleep–wake behavior by BRAC;
(3) diminished probability of sleep and an extended period of W with motor activity near the end of the active period (arrow above the nuchal EMG trace) which
reflects the circadian reinforcement of W against the accumulating drive for sleep; and (4) increased delta power during the initial sleep episodes after the end of
the lights-off/active period (arrow above the delta power trace), reflecting the increased pressure for sleep that accumulated during the active period.
(Unpublished data from L. Kubin, K. Herr & G.L. Mann at the University of Pennsylvania, Philadelphia, PA)
27
28 L. Kubin
2009; Gvilia et al. 2006; Leger et al. 2009; Maloney et al. 2000; Modirrousta et al.
2005; Sherin et al. 1996) indicated that groups of cells exhibiting different activity
patterns relative to the distinct states of sleep and wake aggregate together and
belong to different neurochemical phenotypes (Figs. 2.4 and 2.5). At the very
basic level, cells located in the anterior hypothalamic median and ventrolateral
preoptic nuclei (MnPO and VLPO) were identified as containing gamma-amino
butyric acid (GABA), an inhibitory transmitter, and the peptide galanin and have
increased discharge in association with NREM sleep. These cells have widespread
axonal projections to the forebrain, midbrain, and hindbrain where they target
different cell groups whose activity is associated with wakefulness (Uschakov
et al. 2007). More recently, another group of SWS-active and GABAergic cells
was localized in the reticular formation near the pontomedullary junction medial to
the fibers of the facial nerve (Anaclet et al. 2012, 2014). Thus, the main executive
units responsible for the generation of SWS are located in two brain regions and all
appear to be GABAergic. Notably, the magnitude of their activation during SWS
increases with the increasing need for sleep (Gvilia et al. 2006, 2011; Alam et al.
2014). As such, the anterior hypothalamic sleep-active neurons may accumulate and
then discharge the drive for sleep, thereby mediating a component of the signal
responsible for the homeostatic regulation of sleep.
Whereas the occurrence of SWS appears to be achieved by activation of two
groups of GABAergic neurons, wake-active neurons are distributed more broadly
and belong to at least six different neurochemical phenotypes. The main distinct
groups contain the following neuromodulators, neurotransmitters, and peptides:
norepinephrine (NE), serotonin (5HT), histamine (HI), acetylcholine (ACh), dopa-
mine (DA), and orexins (ORX). These neurons are located in the posterior hypo-
thalamus (HI and ORX), the basal forebrain or rostral pons/caudal midbrain (ACh),
and in the midbrain, pons, and medulla (NE, 5HT, DA). Like the sleep-active
neurons, the wake-active groups have widespread axonal projections throughout
the brain (including the cortex and thalamus), as well as the spinal cord. Through
their direct and indirect connections, when active, they suppress activity of NREM-
active neurons. Hence, there is a mutually reciprocal inhibitory interaction between
sleep- and wake-active neurons. Common to all wake-active neuronal groups is that
activation of any one of them alone is sufficient to terminate sleep and elicit
wakefulness, which may be taken to suggest a high degree of redundancy within
the wake-active network. However, studies also indicate specialization and comple-
mentary roles of different groups in the generation of various aspects of wakefulness.
For example, basal forebrain ACh neurons and NE neurons of the locus coeruleus
(LC) are distinctly activated in association with cortical activation and support
attention to, and processing of, external stimuli. DA, medullary 5HT, and
pontomedullary NE neurons other than LC are particularly activated in connection
with motor activation. ORX cells appear to reinforce wake-related activation of other
wake-active groups. Furthermore, activation of NE, 5HT, and ORX neurons actively
opposes the occurrence of REM sleep and, as such, they are important components
of most models explaining the generation of this state (next section).
2 Neurobiology of Sleep–Wake Control 29
Fig. 2.4 Network of key connections among sleep-active neurons of the anterior hypothalamus and
wake-active neurons of the forebrain and hindbrain responsible for the generation of sleep–wake
states. The anterior hypothalamic cells of the ventrolateral preoptic nucleus (VLPO) synthesize the
inhibitory gamma-aminobutyric acid (GABA) and the inhibitory neuropeptide, galanin (blue). They
are maximally active during NREM sleep and have extensive descending projections that primarily
target different groups of wake-active neurons. As such, they represent the key component of the
sleep-promoting and maintaining part of the network. The wake-active counterpart of the network
includes cholinergic (ACh) neurons of the basal forebrain (BF), histaminergic (HIS) neurons of the
tuberomammillary region (TMN), orexin (ORX, a.k.a. hypocretin)-containing neurons of the
posterior lateral hypothalamus, pontine noradrenergic (NE) neurons symbolized here by the largest
member of this group, the locus coeruleus (LC), dopaminergic (DA) neurons of the A10 and A11
groups which contribute to motor activation during wakefulness, and serotonin (5HT)-containing
neurons of the dorsal and caudal raphé nuclei (DR and CR). Included in the scheme is adenosine
which is one of the established metabolites that accumulate in extracellular space during periods of
sustained cellular activation (e.g., during wakefulness) and provide negative feedback that limit
further activation. As such, adenosine is one of the biochemical substrates of the drive for sleep
(Sect. 2.5). Also, embedded within the network of NE, 5HT, and ACh neuronal groups of the caudal
midbrain and rostral pons is a sub-network of GABA- and glutamate (Glu)-containing neurons that
are responsible for the generation and maintenance of REM sleep. The scheme is adapted with
permission from Fig. 1B in Mignot et al. (2002). Additional contributors to the generation of sleep–
wake states have been discovered and characterized, including distinct subpopulations of hypotha-
lamic and brainstem GABAergic neurons that play important roles in switching between NREM
and REM sleep (Luppi et al. 2017)
30 L. Kubin
Fig. 2.5 At least 17 groups of neurons important for the generation and maintenance of sleep–wake
states can be distinguished based on the combination of three criteria: anatomical location, main
transmitter, or peptide that they use for communication with other neurons, and pattern of their
activity across sleep–wake states. The data set included here distinguishes between active and quiet
wakefulness and, where available, provides information about cell activity during cataplexy.
Cataplexy is a dissociated state that occurs in the disorder of REM sleep called narcolepsy-
cataplexy. During cataplectic attacks, patients experience muscle weakness, or a fully developed
postural atonia like that during REM sleep, but are awake and aware of the external environment.
Information about differences, or lack thereof, in cell firing between active and quiet wakefulness,
on the one hand, and between REM sleep and cataplexy, on the other hand, helps define the specific
roles of different groups of cells in different aspects of behavioral state control. For example, the
histaminergic wake-active cells of the posterior hypothalamus maintain a high level of activity
during cataplectic episodes when subjects are awake but have no postural muscle tone, whereas
noradrenergic and serotonergic wake-active cells are silent or profoundly depressed during cata-
plexy. This distinction suggests a major role for histaminergic cells in the maintenance of wake-
related cortical activation and an important contribution of noradrenergic and serotonergic neurons
to the wake-related maintenance of muscle tone. Abbreviations unique for the Figure: d-l dorsolat-
eral, LPGi lateral paragigantoccellular region, PAG periaqueductal gray, v-l ventrolateral
In contrast to NE, 5HT, and ORX neurons, activation of HI cells is important for
the maintenance of consciousness (and cortical activation) but appears to have a
weak relationship to motor activation or the absence thereof. Supporting this dis-
tinction are single-cell recordings obtained during cataplectic episodes from dogs
selectively bred as genetic models of narcolepsy-cataplexy. Cataplectic attacks are
dissociated states in which motor activity is suppressed through activation of a
subset of the same neural mechanisms that are being activated during natural REM
sleep but, in contrast to REM sleep, awareness of the external environment is
preserved. Under these conditions, HI cells maintain high levels of activity whereas
2 Neurobiology of Sleep–Wake Control 31
NE and 5HT cell activity is abolished, or at least profoundly suppressed, like during
natural REM sleep (John et al. 2004; Wu et al. 1999, 2004). Recordings from
different sleep- and wake-active neurons during dissociated states such as cataplexy
can provide more information about specific roles of different state-dependent
neuronal groups in the generation of different external characteristics of distinct
states of sleep and vigilance. To date, only a limited number of different neuronal
groups have been investigated (Fig. 2.5). The availability of genetic mouse models
of narcolepsy-cataplexy (Chemelli et al. 1999; Zhang et al. 2007) may help to
expand such studies (Thankachan et al. 2009).
The scheme in Fig. 2.4 is an example of the basic network responsible for the
generation of sleep–wake states [adapted from Mignot et al. (2002)]. It emphasizes a
major role of the anterior hypothalamic GABAergic and NREM sleep-active neu-
rons in the generation of sleep and a major role of posterior hypothalamic orexin
neurons in the generation and maintenance of wakefulness. The scheme includes
selected brainstem components of the sleep–wake network but they are given a
relatively subordinate role, and the local mesopontine elements and connections
responsible for the generation and maintenance of REM sleep are not detailed. Other
published variants of the basic sleep–wake network include the components shown
in Fig. 2.4 but differ in the level of detail and relative importance ascribed to
neurochemically different cell groups and their interconnections (Saper et al. 2010;
Fort et al. 2009; Jones 2005; Siegel 2009).
A notable development in the studies of the neuronal network responsible for the
generation of sleep–wake states is the growing evidence for only a limited corre-
spondence between neurochemically different cell groups and different states of
vigilance that they support. Such a diversity occurs in the case of pontine cholinergic
neurons, of which some are distinctly active during all states associated with cortical
activation (during both wakefulness and REM sleep), some have relatively selective
activity increases during REM sleep, and still some are distinctly activated during
eye movements and/or other phasic events occurring during this state (Boucetta et al.
2014; el Mansari et al. 1989; Steriade et al. 1990). A similar functional diversity
involves GABAergic neurons (Maloney et al. 2000). In particular, data indicate that
the caudal midbrain and rostral pons contain different populations of GABAergic
neurons of which some are wake-active and some are REM sleep-active. These
neurons locally interact with excitatory glutamatergic neurons, and probably also
with a separate population of ventromedial medullary REM sleep-active GABAergic
neurons, to generate REM sleep, as well as to terminate this state (Boucetta et al.
2014; Lai et al. 1993, 1999; Luppi et al. 2017; Weber et al. 2015). Furthermore, a
subpopulation of GABAergic neurons that are intermixed among cholinergic neu-
rons within the basal forebrain is wake active (Xu et al. 2015), whereas another
population of posterior hypothalamic GABAergic neurons intermixed among wake-
active orexin-containing neurons are SWS active (Hassani et al. 2010). Additionally,
the melanin-concentrating hormone (MCH) containing posterior hypothalamic neu-
rons are preferentially activated during REM sleep and are also GABAergic
(Hassani et al. 2009b; Peyron et al. 2009). Finally, there is a population of GABA-
and galanin-containing neurons in the region of “extended” VLPO that is
32 L. Kubin
preferentially activated during REM sleep (Lu et al. 2000). Thus, GABAergic
transmission is important, if not essential, for the generation and maintenance of
all three major behavioral states.
Figure 2.5 provides a summary of activity patterns among neurochemically
different cell groups that support the generation of sleep–wake states. This land-
scape, albeit still evolving, provides the basis for the design of different network
models of the generation and maintenance of the states of NREM sleep, REM sleep
and wake, as discussed in the next section.
Fig. 2.6 Models of the interactions among neurochemically and neuroanatomically distinct groups
of neurons that contribute to the generation of sleep–wake states. (a): A model designed to generate
stable states of wakefulness and sleep in which GABA- and galanin (GAL)-containing cells located
in the ventrolateral preoptic nucleus (VLPO) and the extended (e) VLPO of the anterior hypothal-
amus play an active role generating NREM sleep and enabling the emergence of REM sleep from
the former. These cells have extensive efferent connections through which they inhibit multiple cell
groups responsible for the maintenance of wakefulness, such as histaminergic (HIST) cells of the
posterior hypothalamic tuberomammillary region (TMN), noradrenergic (NE) cells of the pontine
locus coeruleus (LC), midbrain serotonergic (5HT) cells of the dorsal raphé nucleus (DR), and
mesopontine acetylcholine-containing cells of the pedunculopontine and laterodorsal tegmental
nuclei (PPT and LDT). The latter are shown in a position to promote both wakefulness and REM
sleep. When active, all wake-related cell groups inhibit sleep-promoting neurons of the VLPO. In
this model, a special role in stabilization of behavioral states is proposed for the wake-related cells
of the posterior lateral hypothalamus that contain the excitatory peptides orexins (ORX). These
cells, when active, reinforce activation of all other wake-active groups. They are themselves
inhibited by sleep-active neurons of the VLPO. The model emphasizes the reciprocal interactions
between sleep- and wake-promoting brain regions in a manner that results in alternative generation
of distinct and stable states and secures rapid and reliable state switching. To emphasize the latter
aspect, the model is referred to as a “flip-flop” switch model. Reproduced with permission from
Saper et al. (2001). (b): A model of interactions among neurochemically different groups of dorsal
pontomesencephalic neurons important for the generation and maintenance of REM sleep. Similar
to the original reciprocal interaction model (McCarley and Hobson 1975), this model also includes a
reciprocal arrangement between serotonergic and noradrenergic neurons (DRN-LC), on the one
hand, and mesopontine cholinergic neurons (PPT-LDT), on the other hand. However, in contrast to
the original model, the interaction is indirect, whereas a key role is ascribed to mutually inhibitory
interaction between inhibitory (GABAergic) REM sleep-active (REM-on) neurons of the pontine
sublaterodorsal region (SLD) and another group of inhibitory neurons located in the ventrolateral
periaqueductal gray region (vlPAG) and in the lateral pontine tegmentum (LPT) that are silent
during REM sleep (REM-off). Additionally, the REM-on part of the circuit includes excitatory
neurons of the caudal pontine (PC) reticular formation whose proposed function is to generate
external manifestations of REM sleep, such as cortical activation and postural atonia. This core
reciprocally organized oscillator is externally enabled by inhibitory REM sleep-related inputs that
descend onto its REM-off component from the melanin-concentrating hormone (MCH) cells of the
posterior lateral hypothalamus and eVLPO cells of the anterior hypothalamus (both GABAergic).
Conversely, external activation of the REM-off component is mediated by excitatory projections
from hypothalamic ORX neurons. This model of flip-flop switching between REM sleep and the
other two major states of vigilance (W and SWS) accounts for two distinct roles of inhibition
mediated by GABA, one to prevent, and the other to promote, REM sleep. Reproduced with
permission from Lu et al. (2006)
34 L. Kubin
neuronal groups that have state-dependent activity patterns (see Fig. 2.5) and
propose specific connections among these groups. As such, they offer an attractive
framework on which to build specific experiments testing different aspects of these
models. One obstacle toward this goal is the increasing recognition that sleep–wake
circuits are often physically intertwined with networks controlling other vital func-
tions, such as mood or metabolism, in a way that makes neuroanatomical and
neurochemical signatures of studied cells of limited use for their unique identifica-
tion as neurons specifically involved in the generation of one or another state of sleep
or wakefulness (e.g., GABAergic cells in the posterior hypothalamus, or
GABAergic cells of the dorsal mesopontine tegmentum). Genetic models and the
selectivity offered by optogenetic activation or inhibition of genetically unique cell
groups should help overcome the methodical problems with unique identification of
different members of the network generating sleep–wake states. Once the basic
models that can generate a tri-phasic sleep–wake rhythm are validated, there will
be a need to better define and model their efferent pathways responsible for the
production and shaping of different external manifestations of behavioral states,
such as electrocortical changes, sleep effects on motor activity, or distinct regula-
tions of memory and cognitive functions during sleep and wake. Also, while the
current “flip-flop” models emphasize the need for stable maintenance of distinct
behavioral states, the cellular and subcellular mechanisms that allow, trigger, and
shape the process of transitions among different states (actual state switching) have
not been well elaborated. This needs further study.
adequately supports ventilation (Orem 1980; Orem and Kubin 2005). On the other
hand, activity of many accessory respiratory muscles that in some individuals need
to be active in order to keep the upper airway open for breathing is profoundly
diminished during REM sleep (or both NREM and REM sleep) which enables the
disorder known as the obstructive sleep apnea syndrome (White and Younes 2012).
Moreover, among the different trunk, proximal limb, and distant limb muscles,
motor suppression is least prominent in the latter, which explains the occurrence
of twitches in the foot and hand muscles. Data also indicate that a gradual deterio-
ration of motor suppression during REM sleep precedes the occurrence of overt
clinical signs of aging-related degenerative disorders (Howell and Schenck 2015;
Iranzo et al. 2013; Postuma et al. 2009). Hence, there are substantial quantitative
differences in the REM sleep-related control of different pools of motor neurons,
both cranial and spinal, and monitoring of the magnitude and pattern of motor
activation, or the absence thereof, during REM sleep may be of diagnostic value.
There is now evidence that three neuroanatomically and neurochemically distinct
mechanisms may contribute to the suppression of motor activity during REM sleep.
They may utilize the following processes: (a) REM sleep-related withdrawal of
motoneuronal activation that is exerted in other states by norepinephrine, serotonin,
and associated neuropeptides (thyrotropin-releasing hormone, substance P, orexins);
(b) active inhibition of motoneurons by cholinergic pathways that are activated
during REM sleep; and (c) active, postsynaptic inhibition of motoneurons by
REM sleep-specific pathways that terminate on motoneurons and release inhibitory
amino acids, such as glycine or GABA. The last of the three potential mechanisms
have been by far most widely considered because intracellular recordings from
lumbar motoneurons during REM sleep revealed the presence of inhibitory postsyn-
aptic potentials (IPSPs) of which some had uniquely large amplitudes (Glenn and
Dement 1981; Morales and Chase 1982; Morales et al. 1987). Similar potentials
were also detected in trigeminal and hypoglossal motoneurons (Chandler et al. 1980;
Fung and Chase 2015; Pedroarena et al. 1994; Yamuy et al. 1999). Furthermore, in
lumbar motoneurons, these IPSPs were abolished by strychnine, an antagonist of
chloride-dependent inhibition mediated by glycine, when the drug was administered
onto individual motoneurons (Chase et al. 1989).
Collectively, the presence of strychnine-sensitive IPSPs in motoneurons supports
the concept that the atonia of REM sleep is caused by an active, postsynaptic
inhibition of motoneurons (Chase and Morales 1990). However, all studies
attempting to block the REM sleep-related suppression of activity in trigeminal or
hypoglossal motoneurons (or their target muscles) by local delivery of antagonists of
the inhibitory glycine or GABA receptors indicated that active inhibition does not
cause the atonia because these well-established antagonists could not abolish, or
even substantially diminish, the atonia (Brooks and Peever 2008; Fenik et al. 2005a;
Kubin et al. 1993; Morrison et al. 2003; Soja et al. 1987). Indeed, all these studies
pointed to a small, if any, role of glycine or GABA in causing the REM sleep-related
suppression of motoneuronal activity. Consequently, one had to conclude that, at
least in hypoglossal and trigeminal motoneurons, REM sleep-specific IPSPs occur
but they are not the main cause of depression of motoneuronal activity during this
36 L. Kubin
state (Kubin 2008). It must, however, be noted that experiments designed similar to
those with hypoglossal and trigeminal motoneurons are yet to be conducted with
postural motoneurons of the spinal cord. Until then, one has to be open to the
possibility that REM sleep-specific active inhibition has a more prominent role in
the spinal motor circuits than in cranial motoneurons that innervate orofacial
muscles.
Withdrawal of aminergic activation of motoneurons (disfacilitation) that must
occur during REM sleep when noradrenergic and serotonergic neurons cease firing
(Fig. 2.5) has been tested in hypoglossal and trigeminal motoneurons as a mecha-
nism of the atonia of REM sleep alternative to the concept of active amino acid-
mediated inhibition. Consistent with the disfacilitation hypothesis, studies revealed
that pharmacological antagonism of appropriate serotonin and/or norepinephrine
receptors located within the regions of the trigeminal or hypoglossal motor nuclei
resulted in decrements of motoneuronal activity comparable in magnitude to those
observed during REM sleep (Chan et al. 2006; Fenik et al. 2005b; Kubin et al. 1992;
Veasey et al. 1996). As the next step in the exploration of this concept, experiments
were designed to test whether a combined withdrawal from motoneurons of excita-
tions mediated by norepinephrine and serotonin causes a depression of motoneuro-
nal activity that is functionally equivalent to the depression of motoneuronal activity
during REM sleep (Fenik et al. 2004, 2005a, b). These experiments, conducted in a
rat pharmacological model of REM sleep, used combined antagonism of appropriate
serotonergic and adrenergic receptors to reduce hypoglossal nerve activity. Impor-
tantly, when episodes of REM sleep-like state were elicited while the endogenous
aminergic effects were fully blocked, there was no further reduction of XII nerve
activity. This indicated that, in hypoglossal motoneurons, a combined withdrawal
from motoneurons of the activations mediated by just two modulators, norepineph-
rine and serotonin, were functionally equivalent to the endogenous neurochemical
processes causing the atonia of REM sleep (Fenik et al. 2005b).
The third mechanism that is proposed to be a potentially important contributor to
the atonia of REM sleep is based on active inhibition mediated by cholinergic
pathways to motoneurons that are specifically activated during REM sleep. The
concept is based on data indicating that there is a subset of brainstem cholinergic
neurons that have axonal projections within the brainstem and to the spinal cord
(Rukhadze and Kubin 2007; Weng et al. 2014), and are selectively activated during
REM sleep (see Fig. 2.5). In support of the cholinergic mechanism, tongue muscle
activity was significantly increased when a broad spectrum muscarinic cholinergic
receptor antagonist, scopolamine, was delivered to the hypoglossal nucleus region
by means of reverse microdialysis during REM sleep (Grace et al. 2013, 2014).
Although activity was also increased during wakefulness and NREM sleep, the
prominent increases obtained during REM sleep were quite remarkable when com-
pared to weak effects reported in earlier studies with antagonists of inhibitory
glycine or GABA receptors. Thus, while the state-specificity of the cholinergic
effect remains to be further investigated, the data show that it is possible to
pharmacologically overcome the depression of activity of hypoglossal motoneurons
during REM sleep.
2 Neurobiology of Sleep–Wake Control 37
Thus, the current landscape of the field of studies on the mechanisms of motor
depression during REM sleep shows that a combination of several distinct neuro-
transmitters and pathways may be involved and that their relative contributions may
vary among different pools of motoneurons [for reviews, see Kubin (2008), Arrigoni
et al. (2016), and Kubin (2016)].
accumulation and removal of adenosine (Bjorness et al. 2016; Frank 2013; Halassa
et al. 2009).
A number of endogenous compounds associated with fever and inflammation
have NREM sleep-promoting properties but data indicate that their role is not limited
to the modulation of sleep during infection and other challenges. Several cytokines
typically generated as a part of immune response facilitate sleep. This includes
interleukin-1β (IL-1β) and tumor necrosis factor α (TNFα) (Krueger 2008; Krueger
et al. 2011). The effects of IL-1β are mediated by the growth hormone-releasing
hormone (GHRH) (Obal and Krueger 2004; Obal et al. 1995), and IL-1β effects
depend on adenosine generated by both neurons and glia (Ingiosi et al. 2015; Nadjar
et al. 2013; Opp and Krueger 2015).
Local cell activation and the use-/activity-dependent generation of ATP and nitric
oxide synthase (NOS) contribute to local modulation of cellular excitability in a
homeostatic manner (Krueger et al. 2013). These and other local cellular and
molecular processes promote local rest/sleep in cortical networks following a period
of intense activation (Krueger et al. 2013; Krueger and Tononi 2011; Vyazovskiy
et al. 2009). Together with a host of other activity-dependent biochemical and
molecular changes, they act as intermediaries for the use-dependent structural
plasticity, such as synaptic scaling and generation or retraction of dendritic spines,
that are necessary for processing and consolidation of memory (Havekes et al. 2016;
Huber et al. 2004; Naidoo et al. 2012; Nelson et al. 2004).
Thus, the different mechanisms put in motion during alternating periods of
increased use and disuse of selected networks and neuronal connections interact
and complement each other in a manner that ensures long-term homeostasis at both
local and organismal levels and supports adaptation to the changing environment.
2.6 Conclusion
A major goal of sleep research and sleep medicine is to identify the mechanisms
responsible for the generation and regulation of natural sleep–wake states and
develop remedies for sleep disorders in humans. To achieve this, a major research
effort is directed toward the exploration of animal models. The underlying premise is
that most fundamental mechanisms are common to all mammals and may even have
their origin in simpler vertebrate and non-vertebrate species. Accordingly, the state
of knowledge discussed in this chapter is extensively informed by experimental
findings derived from animals. Over the last 20 years, rodents, in particular, have
become models of choice for sleep research due to their suitable size, good avail-
ability across a broad and ever-increasing range of genetic modifications, and
sophisticated infrastructure of research tools, such as gene and protein data bases,
well-characterized drugs and antibodies, brain atlases, and data collection equip-
ment. Information derived from this research effort has repeatedly proven to be
applicable to sleep–wake control in humans. Still, beyond fundamental principles,
major species differences exist and need to be considered in translational medicine.
40 L. Kubin
Most rodents, for example, are nocturnal and tend to have more robust circadian
regulation than humans. The metabolic rate of rodents is faster than humans’ which
is probably the reason for the relatively high frequency of most of their rhythmic
processes, including sleep–wake cycling, respiratory rate, and heart rate (Lo et al.
2004). These interspecies differences, albeit important for the practice of medicine,
are beyond consideration in this overview of fundamental principles of sleep–wake
regulation that are common across many species.
Under normal, physiologic conditions, sleep and wake rhythmically alternate as a
result of external gating and pacing imposed on a distributed network of neurons
responsible for the generation and maintenance of each of the three distinct behav-
ioral states, wakefulness, NREM sleep, and REM sleep. The system is robust but
also vulnerable to external and internal perturbations. Major sleep fragmentation,
such as the obstructive sleep apnea syndrome, neuropsychiatric disorders, such as
anxiety disorders or post-traumatic stress disorder (PTSD), shift work, metabolic
derangements, degenerative disorders of aging, and pain are among the factors and
conditions associated with severe sleep disruptions. They occur and exert their
detrimental effects on the development, health status, and quality of life of individ-
uals by altering the properties and functions of the neural networks and cellular
processes discussed in this chapter. The pathophysiology of these changes and
various established and emerging therapies are discussed in other sections of this
volume.
Acknowledgments Our research discussed in this review has been supported by the following
grants from the National Institutes of Health (USA): HL042236, HL047600, HL060287,
HL071097, and HL074385.
References
Alam MA, Kumar S, McGinty D, Alam MN, Szymusiak R. Neuronal activity in the preoptic
hypothalamus during sleep deprivation and recovery sleep. J Neurophysiol. 2014;111(2):
287–99.
Albrecht U. Timing to perfection: the biology of central and peripheral circadian clocks. Neuron.
2012;74(2):246–60.
Anaclet C, Lin JS, Vetrivelan R, Krenzer M, Vong L, Fuller PM, et al. Identification and
characterization of a sleep-active cell group in the rostral medullary brainstem. J Neurosci.
2012;32(50):17970–6.
Anaclet C, Ferrari L, Arrigoni E, Bass CE, Saper CB, Lu J, et al. The GABAergic parafacial zone is
a medullary slow wave sleep-promoting center. Nat Neurosci. 2014;17(9):1217–24.
Arrigoni E, Chen MC, Fuller PM. The anatomical, cellular and synaptic basis of motor atonia
during rapid eye movement sleep. J Physiol. 2016;594(19):5391–414.
Aserinsky E, Kleitman N. Regularly occurring periods of eye motility, and concomitant phenom-
ena, during sleep. Science. 1953;118(3062):273–4.
Basheer R, Sherin JE, Saper CB, Morgan JI, McCarley RW, Shiromani PJ. Effects of sleep on
wake-induced c-fos expression. J Neurosci. 1997;17(24):9746–50.
Bass J, Takahashi JS. Circadian integration of metabolism and energetics. Science. 2010;330
(6009):1349–54.
2 Neurobiology of Sleep–Wake Control 41
Benington JH, Heller HC. Does the function of REM sleep concern non-REM sleep or waking?
Prog Neurobiol. 1994;44(5):433–49.
Benington JH, Kodali SK, Heller HC. Stimulation of A1 adenosine receptors mimics the electro-
encephalographic effects of sleep deprivation. Brain Res. 1995;692(1–2):79–85.
Bjorness TE, Dale N, Mettlach G, Sonneborn A, Sahin B, Fienberg AA, et al. An adenosine-
mediated glial-neuronal circuit for homeostatic sleep. J Neurosci. 2016;36(13):3709–21.
Blanco-Centurion C, Xu M, Murillo-Rodriguez E, Gerashchenko D, Shiromani AM, Salin-Pascual
RJ, et al. Adenosine and sleep homeostasis in the Basal forebrain. J Neurosci. 2006;26(31):
8092–100.
Boissard R, Gervasoni D, Schmidt MH, Barbagli B, Fort P, Luppi PH. The rat ponto-medullary
network responsible for paradoxical sleep onset and maintenance: a combined microinjection
and functional neuroanatomical study. Eur J Neurosci. 2002;16(10):1959–73.
Borbély AA. A two process model of sleep regulation. Hum Neurobiol. 1982;1(3):195–204.
Boucetta S, Cisse Y, Mainville L, Morales M, Jones BE. Discharge profiles across the sleep-waking
cycle of identified cholinergic, GABAergic, and glutamatergic neurons in the
pontomesencephalic tegmentum of the rat. J Neurosci. 2014;34(13):4708–27.
Brooks PL, Peever JH. Glycinergic and GABAA-mediated inhibition of somatic motoneurons does
not mediate rapid eye movement sleep motor atonia. J Neurosci. 2008;28(14):3535–45.
Brown EN, Lydic R, Schiff ND. General anesthesia, sleep, and coma. N Engl J Med. 2010;363(27):
2638–50.
Buckley TM, Schatzberg AF. On the interactions of the hypothalamic-pituitary-adrenal (HPA) axis
and sleep: normal HPA axis activity and circadian rhythm, exemplary sleep disorders. J Clin
Endocrinol Metab. 2005;90(5):3106–14.
Chan E, Steenland HW, Liu H, Horner RL. Endogenous excitatory drive modulating respiratory
muscle activity across sleep-wake states. Am J Respir Crit Care Med. 2006;174(11):1264–73.
Chandler SH, Chase MH, Nakamura Y. Intracellular analysis of synaptic mechanisms controlling
trigeminal motoneuron activity during sleep and wakefulness. J Neurophysiol. 1980;44(2):
359–71.
Chase MH, Morales FR. The atonia and myoclonia of active (REM) sleep. Annu Rev Psychol.
1990;41:557–84.
Chase MH, Soja PJ, Morales FR. Evidence that glycine mediates the postsynaptic potentials that
inhibit lumbar motoneurons during the atonia of active sleep. J Neurosci. 1989;9(3):743–51.
Chemelli RM, Willie JT, Sinton CM, Elmquist JK, Scammell T, Lee C, et al. Narcolepsy in orexin
knockout mice: molecular genetics of sleep regulation. Cell. 1999;98(4):437–51.
D’Olimpio F, Renzi P. Ultradian rhythms in young and adult mice: further support for the basic rest-
activity cycle. Physiol Behav. 1998;64(5):697–701.
de Lecea L, Kilduff TS, Peyron C, Gao X, Foye PE, Danielson PE, et al. The hypocretins:
hypothalamus-specific peptides with neuroexcitatory activity. Proc Natl Acad Sci U S
A. 1998;95(1):322–7.
Dentico D, Amici R, Baracchi F, Cerri M, Del Sindaco E, Luppi M, et al. c-Fos expression in
preoptic nuclei as a marker of sleep rebound in the rat. Eur J Neurosci. 2009;30(4):651–61.
Dworak M, McCarley RW, Kim T, Kalinchuk AV, Basheer R. Sleep and brain energy levels: ATP
changes during sleep. J Neurosci. 2010;30(26):9007–16.
el Mansari M, Sakai K, Jouvet M. Unitary characteristics of presumptive cholinergic tegmental
neurons during the sleep-waking cycle in freely moving cats. Exp Brain Res. 1989;76(3):
519–29.
Emens JS, Burgess HJ. Effect of light and melatonin and other melatonin receptor agonists on
human circadian physiology. Sleep Med Clin. 2015;10(4):435–53.
Feldman JL, Mitchell GS, Nattie EE. Breathing: rhythmicity, plasticity, chemosensitivity. Annu
Rev Neurosci. 2003;26:239–66.
Fenik VB, Kubin L. Differential localization of carbachol- and bicuculline-sensitive pontine sites
for eliciting REM sleep-like effects in anesthetized rats. J Sleep Res. 2009;18(1):99–112.
42 L. Kubin
Fenik V, Davies RO, Kubin L. Combined antagonism of aminergic excitatory and amino acid
inhibitory receptors in the XII nucleus abolishes REM sleep-like depression of hypoglossal
motoneuronal activity. Arch Ital Biol. 2004;142(3):237–49.
Fenik VB, Davies RO, Kubin L. Noradrenergic, serotonergic and GABAergic antagonists injected
together into the XII nucleus abolish the REM sleep-like depression of hypoglossal motoneu-
ronal activity. J Sleep Res. 2005a;14(4):419–29.
Fenik VB, Davies RO, Kubin L. REM sleep-like atonia of hypoglossal (XII) motoneurons is caused
by loss of noradrenergic and serotonergic inputs. Am J Respir Crit Care Med. 2005b;172(10):
1322–30.
Fort P, Bassetti CL, Luppi PH. Alternating vigilance states: new insights regarding neuronal
networks and mechanisms. Eur J Neurosci. 2009;29(9):1741–53.
Frank MG. Astroglial regulation of sleep homeostasis. Curr Opin Neurobiol. 2013;23(5):812–8.
Franken P, Chollet D, Tafti M. The homeostatic regulation of sleep need is under genetic control. J
Neurosci. 2001;21(8):2610–21.
Fung SJ, Chase MH. Postsynaptic inhibition of hypoglossal motoneurons produces atonia of the
genioglossal muscle during rapid eye movement sleep. Sleep. 2015;38(1):139–46.
Glenn LL, Dement WC. Membrane potential, synaptic activity, and excitability of hindlimb
motoneurons during wakefulness and sleep. J Neurophysiol. 1981;46(4):839–54.
Grace KP, Hughes SW, Horner RL. Identification of the mechanism mediating genioglossus muscle
suppression in REM sleep. Am J Respir Crit Care Med. 2013;187(3):311–9.
Grace KP, Hughes SW, Horner RL. Identification of a pharmacological target for genioglossus
reactivation throughout sleep. Sleep. 2014;37(1):41–50.
Gvilia I, Xu F, McGinty D, Szymusiak R. Homeostatic regulation of sleep: a role for preoptic area
neurons. J Neurosci. 2006;26(37):9426–33.
Gvilia I, Suntsova N, Angara B, McGinty D, Szymusiak R. Maturation of sleep homeostasis in
developing rats: a role for preoptic area neurons. Am J Physiol Regul Integr Comp Physiol.
2011;300(4):R885–94.
Halassa MM, Florian C, Fellin T, Munoz JR, Lee SY, Abel T, et al. Astrocytic modulation of sleep
homeostasis and cognitive consequences of sleep loss. Neuron. 2009;61(2):213–9.
Hassani OK, Lee MG, Henny P, Jones BE. Discharge profiles of identified GABAergic in
comparison to cholinergic and putative glutamatergic basal forebrain neurons across the
sleep-wake cycle. J Neurosci. 2009a;29(38):11828–40.
Hassani OK, Lee MG, Jones BE. Melanin-concentrating hormone neurons discharge in a reciprocal
manner to orexin neurons across the sleep-wake cycle. Proc Natl Acad Sci U S A. 2009b;106(7):
2418–22.
Hassani OK, Henny P, Lee MG, Jones BE. GABAergic neurons intermingled with orexin and MCH
neurons in the lateral hypothalamus discharge maximally during sleep. Eur J Neurosci. 2010;32
(3):448–57.
Havekes R, Park AJ, Tudor JC, Luczak VG, Hansen RT, Ferri SL, et al. Sleep deprivation causes
memory deficits by negatively impacting neuronal connectivity in hippocampal area CA1.
eLife. 2016;5:e13424.
Hendricks JC, Finn SM, Panckeri KA, Chavkin J, Williams JA, Sehgal A, et al. Rest in Drosophila
is a sleep-like state. Neuron. 2000;25(1):129–38.
Hobson JA, Lydic R, Baghdoyan HA. Evolving concepts of sleep cycle generation: from brain
centers to neuronal populations. Behav Brain Sci. 1986;9:371–448.
Horne JA. REM sleep—by default? Neurosci Biobehav Rev. 2000;24(8):777–97.
Howell MJ, Schenck CH. Rapid eye movement sleep behavior disorder and neurodegenerative
disease. JAMA Neurol. 2015;72(6):707–12.
Huber R, Ghilardi MF, Massimini M, Tononi G. Local sleep and learning. Nature. 2004;430(6995):
78–81.
Ikeda M, Ikeda-Sagara M, Okada T, Clement P, Urade Y, Nagai T, et al. Brain oxidation is an initial
process in sleep induction. Neuroscience. 2005;130(4):1029–40.
2 Neurobiology of Sleep–Wake Control 43
Ingiosi AM, Raymond RM Jr, Pavlova MN, Opp MR. Selective contributions of neuronal and
astroglial interleukin-1 receptor 1 to the regulation of sleep. Brain Behav Immun. 2015;48:244–
57.
Inoue S, Honda K, Komoda Y. Sleep as neuronal detoxification and restitution. Behav Brain Res.
1995;69(1–2):91–6.
Iranzo A, Tolosa E, Gelpi E, Molinuevo JL, Valldeoriola F, Serradell M, et al. Neurodegenerative
disease status and post-mortem pathology in idiopathic rapid-eye-movement sleep behaviour
disorder: an observational cohort study. Lancet Neurol. 2013;12(5):443–53.
John J, Wu MF, Boehmer LN, Siegel JM. Cataplexy-active neurons in the hypothalamus: implica-
tions for the role of histamine in sleep and waking behavior. Neuron. 2004;42(4):619–34.
Jones BE. Paradoxical sleep and its chemical/structural substrates in the brain. Neuroscience.
1991;40(3):637–56.
Jones BE. From waking to sleeping: neuronal and chemical substrates. Trends Pharmacol Sci.
2005;26(11):578–86.
Jouvet M. [Research on the neural structures and responsible mechanisms in different phases of
physiological sleep]. Arch Ital Biol 1962;100(2):125–206.
Kalsbeek A, Yi CX, la Fleur SE, Buijs RM, Fliers E. Suprachiasmatic nucleus and autonomic
nervous system influences on awakening from sleep. Int Rev Neurobiol. 2010;93:91–107.
Kim Y, Laposky AD, Bergmann BM, Turek FW. Repeated sleep restriction in rats leads to
homeostatic and allostatic responses during recovery sleep. Proc Natl Acad Sci U S
A. 2007;104(25):10697–702.
Kleitman N. Biological rhythms and cycles. Physiol Rev. 1949;29(1):1–30.
Kleitman N. Sleep and wakefulness. Chicago: University of Chicago Press; 1963.
Kleitman N. Basic rest-activity cycle—22 years later. Sleep. 1982;5(4):311–7.
Krueger JM. The role of cytokines in sleep regulation. Curr Pharm Des. 2008;14(32):3408–16.
Krueger JM, Tononi G. Local use-dependent sleep; synthesis of the new paradigm. Curr Top Med
Chem. 2011;11(19):2490–2.
Krueger JM, Clinton JM, Winters BD, Zielinski MR, Taishi P, Jewett KA, et al. Involvement of
cytokines in slow wave sleep. Prog Brain Res. 2011;193:39–47.
Krueger JM, Huang YH, Rector DM, Buysse DJ. Sleep: a synchrony of cell activity-driven small
network states. Eur J Neurosci. 2013;38(2):2199–209.
Kubin L. Carbachol models of REM sleep: recent developments and new directions. Arch Ital Biol.
2001;139(1–2):147–68.
Kubin L. Adventures and tribulations in the search for the mechanisms of the atonia of REM sleep.
Sleep. 2008;31(11):1473–6.
Kubin L. Neural control of the upper airway: respiratory and state-dependent mechanisms. Compr
Physiol. 2016;6(4):1801–50.
Kubin L, Tojima H, Davies RO, Pack AI. Serotonergic excitatory drive to hypoglossal motoneurons
in the decerebrate cat. Neurosci Lett. 1992;139(2):243–8.
Kubin L, Kimura H, Tojima H, Davies RO, Pack AI. Suppression of hypoglossal motoneurons
during the carbachol-induced atonia of REM sleep is not caused by fast synaptic inhibition.
Brain Res. 1993;611(2):300–12.
Lai YY, Clements JR, Siegel JM. Glutamatergic and cholinergic projections to the pontine
inhibitory area identified with horseradish peroxidase retrograde transport and immunohisto-
chemistry. J Comp Neurol. 1993;336(3):321–30.
Lai YY, Clements JR, Wu XY, Shalita T, Wu JP, Kuo JS, et al. Brainstem projections to the
ventromedial medulla in cat: retrograde transport horseradish peroxidase and immunohisto-
chemical studies. J Comp Neurol. 1999;408(3):419–36.
Landolt HP. Sleep homeostasis: a role for adenosine in humans? Biochem Pharmacol. 2008;75(11):
2070–9.
Lavie P. REM periodicity under ultrashort sleep/wake cycle in narcoleptic patients. Can J Psychol.
1991;45(2):185–93.
44 L. Kubin
Leger L, Goutagny R, Sapin E, Salvert D, Fort P, Luppi PH. Noradrenergic neurons expressing Fos
during waking and paradoxical sleep deprivation in the rat. J Chem Neuroanat. 2009;37(3):
149–57.
Lin L, Faraco J, Li R, Kadotani H, Rogers W, Lin X, et al. The sleep disorder canine narcolepsy is
caused by a mutation in the hypocretin (orexin) receptor 2 gene. Cell. 1999;98(3):365–76.
Liu M, Thankachan S, Kaur S, Begum S, Blanco-Centurion C, Sakurai T, et al. Orexin (hypocretin)
gene transfer diminishes narcoleptic sleep behavior in mice. Eur J Neurosci. 2008;28(7):
1382–93.
Lo CC, Chou T, Penzel T, Scammell TE, Strecker RE, Stanley HE, et al. Common scale-invariant
patterns of sleep-wake transitions across mammalian species. Proc Natl Acad Sci U S
A. 2004;101(50):17545–8.
Lu J, Greco MA, Shiromani P, Saper CB. Effect of lesions of the ventrolateral preoptic nucleus on
NREM and REM sleep. J Neurosci. 2000;20(10):3830–42.
Lu J, Sherman D, Devor M, Saper CB. A putative flip-flop switch for control of REM sleep. Nature.
2006;441(7093):589–94.
Luppi PH, Clement O, Sapin E, Peyron C, Gervasoni D, Leger L, et al. Brainstem mechanisms of
paradoxical (REM) sleep generation. Pflugers Arch. 2012;463(1):43–52.
Luppi PH, Peyron C, Fort P. Not a single but multiple populations of GABAergic neurons control
sleep. Sleep Med Rev. 2017;32:85–94.
Lydic R, Baghdoyan HA. Sleep, anesthesiology, and the neurobiology of arousal state control.
Anesthesiology. 2005;103(6):1268–95.
Mackiewicz M, Nikonova EV, Zimmerman JE, Galante RJ, Zhang L, Cater JR, et al. Enzymes of
adenosine metabolism in the brain: diurnal rhythm and the effect of sleep deprivation. J
Neurochem. 2003;85(2):348–57.
Maloney KJ, Mainville L, Jones BE. c-Fos expression in GABAergic, serotonergic, and other
neurons of the pontomedullary reticular formation and raphe after paradoxical sleep deprivation
and recovery. J Neurosci. 2000;20(12):4669–79.
Matsuki T, Takasu M, Hirose Y, Murakoshi N, Sinton CM, Motoike T, et al. GABAA receptor-
mediated input change on orexin neurons following sleep deprivation in mice. Neuroscience.
2015;284:217–24.
McCarley RW, Hobson JA. Neuronal excitability modulation over the sleep cycle: a structural and
mathematical model. Science. 1975;189(4196):58–60.
McPartland RJ, Kupfer DJ. Rapid eye movement sleep cycle, clock time and sleep onset.
Electroencephalogr Clin Neurophysiol. 1978;45(2):178–85.
Mignot E, Taheri S, Nishino S. Sleeping with the hypothalamus: emerging therapeutic targets for
sleep disorders. Nat Neurosci. 2002;5(Suppl):1071–5.
Mistlberger RE. Circadian regulation of sleep in mammals: role of the suprachiasmatic nucleus.
Brain Res Brain Res Rev. 2005;49(3):429–54.
Modirrousta M, Mainville L, Jones BE. Orexin and MCH neurons express c-Fos differently after
sleep deprivation vs. recovery and bear different adrenergic receptors. Eur J Neurosci. 2005;21
(10):2807–16.
Modirrousta M, Mainville L, Jones BE. Dynamic changes in GABAA receptors on basal forebrain
cholinergic neurons following sleep deprivation and recovery. BMC Neurosci. 2007;8:15.
Morales FR, Chase MH. Repetitive synaptic potentials responsible for inhibition of spinal cord
motoneurons during active sleep. Exp Neurol. 1982;78(2):471–6.
Morales FR, Boxer P, Chase MH. Behavioral state-specific inhibitory postsynaptic potentials
impinge on cat lumbar motoneurons during active sleep. Exp Neurol. 1987;98(2):418–35.
Morrison JL, Sood S, Liu H, Park E, Liu X, Nolan P, et al. Role of inhibitory amino acids in control
of hypoglossal motor outflow to genioglossus muscle in naturally sleeping rats. J Physiol.
2003;552(Pt 3):975–91.
Nadjar A, Blutstein T, Aubert A, Laye S, Haydon PG. Astrocyte-derived adenosine modulates
increased sleep pressure during inflammatory response. Glia. 2013;61(5):724–31.
2 Neurobiology of Sleep–Wake Control 45
Naidoo N, Ferber M, Galante RJ, McShane B, Hu JH, Zimmerman J, et al. Role of Homer proteins
in the maintenance of sleep-wake states. PLoS One. 2012;7(4):e35174.
Nelson SB, Sjostrom PJ, Turrigiano GG. Rate and timing in cortical synaptic plasticity. Philos
Trans R Soc Lond Ser B Biol Sci. 2002;357(1428):1851–7.
Nelson SE, Duricka DL, Campbell K, Churchill L, Krueger JM. Homer1a and 1bc levels in the rat
somatosensory cortex vary with the time of day and sleep loss. Neurosci Lett. 2004;367(1):
105–8.
Nykamp K, Rosenthal L, Folkerts M, Roehrs T, Guido P, Roth T. The effects of REM sleep
deprivation on the level of sleepiness/alertness. Sleep. 1998;21(6):609–14.
Obal F Jr, Krueger JM. GHRH and sleep. Sleep Med Rev. 2004;8(5):367–77.
Obal F Jr, Fang J, Payne LC, Krueger JM. Growth-hormone-releasing hormone mediates the sleep-
promoting activity of interleukin-1 in rats. Neuroendocrinology. 1995;61(5):559–65.
Ocampo-Garces A, Vivaldi EA. Short-term homeostasis of REM sleep assessed in an intermittent
REM sleep deprivation protocol in the rat. J Sleep Res. 2002;11(1):81–9.
Opp MR, Krueger JM. Sleep and immunity: a growing field with clinical impact. Brain Behav
Immun. 2015;47:1–3.
Orem J. Neuronal mechanisms of respiration in REM sleep. Sleep. 1980;3(3–4):251–67.
Orem J, Kubin L. Respiratory physiology: central neural control. In: Kryger MH, Roth T, Dement
WC, editors. Principles and practice of sleep medicine. 4th ed. Philadelphia, PA: Elsevier-
Saunders; 2005. p. 213–23.
Paul KN, Losee-Olson S, Pinckney L, Turek FW. The ability of stress to alter sleep in mice is
sensitive to reproductive hormones. Brain Res. 2009;1305:74–85.
Pedroarena C, Castillo P, Chase MH, Morales FR. The control of jaw-opener motoneurons during
active sleep. Brain Res. 1994;653(1–2):31–8.
Petit JM, Burlet-Godinot S, Magistretti PJ, Allaman I. Glycogen metabolism and the homeostatic
regulation of sleep. Metab Brain Dis. 2015;30(1):263–79.
Peyron C, Sapin E, Leger L, Luppi PH, Fort P. Role of the melanin-concentrating hormone
neuropeptide in sleep regulation. Peptides. 2009;30(11):2052–9.
Pollock MS, Mistlberger RE. Rapid eye movement sleep induction by microinjection of the GABA-
A antagonist bicuculline into the dorsal subcoeruleus area of the rat. Brain Res. 2003;962(1–2):
68–77.
Porkka-Heiskanen T, Strecker RE, McCarley RW. Brain site-specificity of extracellular adenosine
concentration changes during sleep deprivation and spontaneous sleep: an in vivo microdialysis
study. Neuroscience. 2000;99(3):507–17.
Postuma RB, Gagnon JF, Vendette M, Montplaisir JY. Markers of neurodegeneration in idiopathic
rapid eye movement sleep behaviour disorder and Parkinson’s disease. Brain. 2009;132(Pt 12):
3298–307.
Qiu MH, Chen MC, Lu J. Cortical neuronal activity does not regulate sleep homeostasis. Neuro-
science. 2015;297:211–8.
Radulovacki M. Role of adenosine in sleep in rats. Rev Clin Basic Pharm. 1985;5(3–4):327–39.
Rai S, Kumar S, Alam MA, Szymusiak R, McGinty D, Alam MN. A1 receptor mediated
adenosinergic regulation of perifornical-lateral hypothalamic area neurons in freely behaving
rats. Neuroscience. 2010;167(1):40–8.
Rector DM, Schei JL, Van Dongen HP, Belenky G, Krueger JM. Physiological markers of local
sleep. Eur J Neurosci. 2009;29(9):1771–8.
Reid KJ, Abbott SM. Jet lag and shift work disorder. Sleep Med Clin. 2015;10(4):523–35.
Reiner PB. Are mesopontine cholinergic neurons either necessary or sufficient components of the
ascending reticular activating system? Semin Neurosci. 1995;7(5):355–9.
Richter DW, Spyer KM. Studying rhythmogenesis of breathing: comparison of in vivo and in vitro
models. Trends Neurosci. 2001;24(8):464–72.
Rolls A, Pang WW, Ibarra I, Colas D, Bonnavion P, Korin B, et al. Sleep disruption impairs
haematopoietic stem cell transplantation in mice. Nat Commun. 2015;6:8516.
46 L. Kubin
McCarley RW, Sinton CM. Neurobiology of sleep and wakefulness. Scholarpedia. 2008;3:3313.
http://www.scholarpedia.org/article/Neurobiology_of_sleep_and_wakefulness. Accessed
Nov 2021.
National Institute of Neurological Disorders and Stroke. Brain basics: understanding sleep. https://
www.ninds.nih.gov/Disorders/Patient-Caregiver-Education/Understanding-Sleep. Accessed
Nov 2021.
Wikipedia. Neuroscience of sleep. https://en.wikipedia.org/wiki/Neuroscience_of_sleep. Accessed
Nov 2021.
Chapter 3
Prostaglandins, Adenosine,
and Histaminergic System in the Regulation
of Sleep and Wakefulness
Abstract This chapter provides an overview of the current knowledge about the
roles of prostaglandins, adenosine, and the histaminergic system in sleep–wake
regulation, focusing on prostaglandins, adenosine, and histamine in the central
nervous system, their level regulation, their receptors, and pharmacological and
molecular biological manipulations of the adenosine and histaminergic systems.
Prostaglandin (PG) D2 is an endogenous somnogen that can increase the extracellu-
lar adenosine under the subarachnoid space of the basal forebrain, thereby induce
physiological sleep. Adenosine is found neither stored nor released as a classical
neurotransmitter, which is formed inside cells or on their surface and derived from
adenine nucleotide breakdown. Prolonged wakefulness increases extracellular aden-
osine concentration in the cortex and basal forebrain and the concentration will go
back during the sleep recovery period. Therefore, adenosine has been thought of as a
homeostatic regulator of sleep and a link between the humoral and neural mecha-
nisms of sleep–wake regulation. Both the adenosine A1 receptor (A1R) and the A2AR
are involved in sleep induction. The somnogenic effects of PGD2 are predominantly
dependent on A2AR. In addition, it is proved that the A2AR is necessary for the
arousal effect of caffeine by using gene-manipulated mice. In contrast, the role of the
A1R is more complicated. Although stimulation of A1R in wake-promoting brain
areas increases sleep, activation of A1R in the lateral preoptic area induces wakeful-
ness, indicating that the A1R acts in a site-dependent manner in sleep–wake regula-
tion. The histaminergic system also plays an essential role in sleep–wake regulation
and is indispensable for the sleep/wakefulness-promoting effects induced by the A1R
and A2AR. A brief discussion about the potential therapeutic applications of agonists
and antagonists of these receptors in sleep disorders is also included at the end of this
chapter.
© Springer Nature B.V. and Shanghai Jiao Tong University Press 2022 49
A. I. Pack, Q. Y. Li (eds.), Sleep and its Disorders, Translational Medicine Research,
https://doi.org/10.1007/978-94-024-2168-2_3
50 Z.-L. Huang et al.
Abbreviations
3.1 Introduction
Sleep propensity rises with the prolonged wakefulness time and dissipates with the
progression of sleep. The waxing and waning of the sleep drive are presumed to be
regulated by endogenous sleep factors acting on specific neurons in the brain
(Porkka-Heiskanen et al. 1999). One of these sleep factors is adenosine, a purine
nucleoside naturally occurring in all cells (Fredholm 2007), the other one is
3 Prostaglandins, Adenosine, and Histaminergic System in the Regulation. . . 51
prostaglandin (PG) D2, an eicosanoid acting as tissue or local hormone (Huang et al.
2007). Both adenosine and PGD2 are released as neuromodulators in the brain. Here,
we are discussing some aspects of high general complexity: (a) adenosine is a key
signaling molecule for PGD2-induced sleep; (b) the metabolism of adenosine, which
is ubiquitously present in all cells; (c) key roles of the A2A receptor (A2AR) in sleep
induction; (d) adenosine A1R plays a role in sleep–wake regulation in a site-
dependent manner.
The discovery that adenosine acts as a mediator of PGD2-induced sleep led to the
identification of the mechanism by which endogenous PGD2 promotes sleep
(Fig. 3.1) (Satoh et al. 1996). PGD2 was found to be one of the most potent
somnogens and also the most abundant prostaglandin in the brains of rats (Narumiya
et al. 1982) and other mammals including humans (Ogorochi et al. 1984) involved in
physiological sleep (Urade and Hayaishi 2010) The PGD2 concentration in the
cerebrospinal fluid (CSF) of rats fluctuates along with the sleep–wake rhythms
(Pandey et al. 1995) and elevates with an increase of sleep propensity during sleep
deprivation (Ram et al. 1997), implying that PGD2 may have some important role in
the CNS. The sleep-inducing activity of PGD2 was discovered while the scientists
were trying to investigate its neural function (Ueno et al. 1982). Both non-rapid eye
Fig. 3.1 Molecular mechanism of sleep–wake regulation. The endogenous somnogen PGD2
(green), produced by L-PGDS, circulates within the CSF, stimulates DP1R (light blue) on the
ventral surface from the basal forebrain to the hypothalamus, and increases the level of extracellular
adenosine. Adenosine (purple) diffuses into the brain parenchyma as the secondary somnogen,
inhibits arousal neurons in the basal forebrain (orange area) and TMN (red area) via A1R (blue
lines), and activates sleep–active VLPO neurons (blue area) via A2AR (red arrows) to induce sleep.
The flip-flop switch of sleep–wakefulness regulation between the VLPO and TMN is stabilized by
their OX/Hcrt-mediated activation and adenosine A1R-mediated suppression. Ach Acetylcholine,
EP4 Prostaglandin E2 receptor, subtype EP4, H1R Histamine H1 receptor, Hcrt Hypocretin,
L-PGDS Lipocalin-type prostaglandin D synthase, OX Orexin, TMN Tuberomammillary nucleus,
VLPO Ventrolateral preoptic area. Adapted and modified from Urade and Hayaishi (2010) with
permission
52 Z.-L. Huang et al.
movement (non-REM, NREM) and REM sleep increased in the rat brain just by
microinjection of nanomolar quantities of PGD2 (Ueno et al. 1983). The same sleep-
inducing effect was found in the rhesus monkey by an i.c.v. infusion of PGD2 (Onoe
et al. 1988). The PGD2 can significantly induce sleep with as small as picomolar
quantities per minute, which is almost identical to physiological sleep as judged by
several electrophysiological and behavioral criteria.
PGD2 can be produced by two distinct types of PGD synthase (PGDS) in
the CNS: one is lipocalin-type PGDS (L-PGDS), substantially locates in the arach-
noid membrane, choroid plexus, and oligodendrocytes (Mizoguchi et al. 2001;
Urade et al. 1993); and the other, hematopoietic PGDS (H-PGDS), expresses in
microglia (Mohri et al. 2003). PGD2 that are produced by either of these two
enzymes will be secreted into the CSF, and the level of PGD2 there shows a circadian
change in freely moving rats (Pandey et al. 1995). L-PGDS is the same as β-trace, a
major protein in human CSF. Also, the serum L-PGDS/β-trace concentration
exhibits a circadian alteration with a nocturnal increase, which is suppressed by
total sleep deprivation but not influenced during REM sleep deprivation in humans
(Jordan et al. 2004). In patients with narcolepsy and idiopathic hypersomnia,
L-PGDS levels were reported to be increased (Wang et al. 2021).
Furthermore, sleep is inhibited in a time- and dose-dependent fashion and is
reversible when rats are administered the specific and reversible inhibitors of PGDS,
inorganic tetravalent selenium (Se4+) compounds such as selenium tetrachloride
(SeCl4) (Islam et al. 1991; Matsumura et al. 1991; Takahata et al. 1993). These
results suggest that PGD2/PGDS plays a key role in sleep induction.
To investigate the mechanism of PGD2 in regulating physiological sleep, we
tested the effect of inhibition of PGD2 synthesis by SeCl4 on the sleep in wild-type
(WT) mice as well as PGDS and PGD2 receptor (DP1R) knockout (KO) mice, and
the effect of a DP1R antagonist, ONO-4127Na, on sleep in rats. The PGD2 concen-
tration in the brain of WT mice decreased by the i.p. injection of SeCl4 without
affecting the amounts of PGE2 and PGF2α. The injection dose-dependently
suppressed sleep and induced almost complete insomnia after the administration
during the light phase when mice normally sleep. The SeCl4-induced insomnia was
observed in H-PGDS KO mice but not in L-PGDS KO, H- and L-PGDS double KO,
or DP1R KO mice. Furthermore, the amount of sleep decreased when the DP1R
antagonist ONO-4127Na was infused into the subarachnoid space under the rostral
basal forebrain in rats (Qu et al. 2006). These findings indicate that the L-PGDS/
PGD2/DP1R system is critical for physiological sleep regulation.
The extracellular adenosine concentration was increased dose dependently while
PGD2 was infused into the subarachnoid space of the basal forebrain of WT mice
(Mizoguchi et al. 2001), in which DP1Rs are remarkably abundant, whereas the
mechanism linking PGD2 and adenosine accumulation is still unclear. The increase
of extracellular adenosine induced by PGD2 was only observed in WT mice, but not
in DP1R KO mice (Fig. 3.2) (Mizoguchi et al. 2001). Moreover, the sleep-promoting
effect of PGD2 can be eliminated by i.p. injection of the A2AR-selective antagonist
KF 17837. These results suggest that the adenosine increase was dependent on the
activation of DP1R and that endogenous adenosine acting at A2ARs may mediate the
3 Prostaglandins, Adenosine, and Histaminergic System in the Regulation. . . 53
Fig. 3.2 Effect of PGD2 perfusion on the extracellular adenosine level in the subarachnoid space
below the rostral basal forebrain of WT and DP/ mice. Vehicle or PGD2 was perfused for 2 h
(black bar) into the subarachnoid space below the rostral basal forebrain of WT (a) and DP/ (b)
mice under anesthesia. The basal level of adenosine for 1 h before the PGD2 perfusion was
0.46 0.04 pmol/20 μl in WT mice and 0.24 0.04 pmol/20 μl in DP/ mice. The data are
expressed as a percentage of the baseline value (mean SEM, n ¼ 5–8). *,{P < 0.05; **,{{P < 0.01,
compared with the vehicle group. Adapted from Mizoguchi et al. (2001) with modification and
permission
PGD2-induced sleep (Fig. 3.1). In addition, it was found that PGD2-induced sleep
attenuated in animals with adenosine A2AR deficiency (Oishi et al. 2017), suggesting
that adenosine A2AR is necessary for this process.
Fig. 3.3 Intracellular and extracellular pathways for the formation and metabolism of adenosine.
Inside the cell, adenosine is formed from ATP, cAMP, or SAH, while outside the cell it arises from
equilibrating nucleoside transporter-mediated release or metabolism from ATP or cAMP. Adapted
from Sawynok and Liu (2003) with modification and permission
mediate adenosine reuptake, and the concentration gradient on both sides of the
membrane determines the direction of the transport (Gu et al. 1995). These equili-
brating transporters are abundant in most cells, including astrocytes (Peng et al.
2005).
Extracellular adenosine derives from two major mechanisms: (1) the release of
adenosine caused by an increase in the intracellular levels of adenosine through
nucleoside transporters (Geiger and Fyda 1991); and (2) the extracellular formation
of adenosine through the ectonucleotidase pathway on the release of adenine nucle-
otides, especially ATP.
Adenosine can be removed by nucleoside transporters and three kinds of
enzymes, i.e., adenosine kinase (AK), adenosine deaminase (ADA), and S-
adenosylhomocysteine hydrolase (SAHH). The clearance of extracellular adenosine
mostly occurs through the non-concentrating nucleoside transporters, and ecto-ADA
deaminates adenosine to inosine (Fredholm et al. 2005). The major intracellular
metabolic pathways of adenosine are the formation of AMP through AK and the
irreversible breakdown of inosine via ADA. AK is found enriched in neurons,
whereas ADA is more abundant in astrocytes (Fredholm et al. 2005) and histamin-
ergic neurons in the tuberomammillary nuclei (TMN) (Nagy et al. 1984; Oishi et al.
2008). A third enzyme, SAHH, which can convert adenosine to S-
adenosylhomocysteine in cardiomyocytes, might be of less importance in the CNS
(Fredholm et al. 2005).
There are four adenosine receptor subtypes and all of them are G-protein-coupled
receptors (GPCR): A1 and A3 are primarily coupled to the Gi family of G proteins,
whereas A2A and A2B are mostly coupled to Gs, or Golf protein (Peng et al. 2005).
Stimulation of A1Rs inhibits adenylate cyclase through activation of Gi proteins,
activates phospholipase C (PLC), opens several types of K+ channels, and inacti-
vates Q-, P-, and N-type Ca2+ channels (Fredholm et al. 2005; Jacobson and Gao
2006). On the other hand, activation of the A2AR subtype increases adenylate
cyclase activity through activation of Gs or Golf (in the striatum) proteins, induces
the formation of inositol phosphates, and activates protein kinase C (Fredholm et al.
2005; Jacobson and Gao 2006). It has been demonstrated that both A1 and A2AR
participate in sleep induction, whereas A2AR is more important in adenosine-
induced sleep. Although A2BR is expressed broadly, it is generally at very low
levels; in contrast, the A3R is expressed at intermediate levels in the hippocampus
and cerebellum (Yaar et al. 2005). However, the functional significance of A2BR and
A3R in sleep regulation is still hardly known. The main properties of adenosine
receptors are shown in Table 3.1.
Accumulated evidence indicates that the A2AR is critical for the effects of
adenosine-induced sleep. The concentrations of A2ARs are high in the CNS, mainly
in the striatum (Yuan et al. 2017), nucleus accumbens (Oishi et al. 2017), and
olfactory bulb (Wang et al. 2017; Fredholm et al. 2001). In rats, selective A2AR
agonists such as CG-S21680 administered to the subarachnoid space adjacent to the
basal forebrain and lateral preoptic area reliably induce NREM sleep, whereas
infusion of A1R agonists produces weak and variable effects (Mohri et al. 2003;
Methippara et al. 2005; Satoh et al. 1998; Scammell et al. 2001). When infused into
the medial pontine reticular formation in rats, CGS-21680 was tenfold more potent
than the A1R agonist N6-cyclohexyl-adenosine, in inducing REM sleep (Marks et al.
2003). Sleep induced by the A2AR agonist CGS-21680 is followed by a strong
rebound of wakefulness after the cessation of CGS-21680 infusion (Gerashchenko
et al. 2000). The major region that mediates A2AR-induced sleep is thought to be
located in or near the rostral basal forebrain, which is proved by the sleep-promoting
effect and c-Fos expression after local infusion of CGS-21680 (Satoh et al. 1999).
Moreover, CGS-21680-induced sleep is almost completely eliminated in A2AR
knockout mice, confirming the specificity of CGS-21680 for A2ARs (Urade et al.
2003).
A possible neural circuit of A2AR-mediated PGD2-induced sleep was mapped by
c-Fos-positive neurons detection (Scammell et al. 2001; Satoh et al. 1999). When
PGD2 or the A2AR agonist CGS-21680 was infused for 2 h into the PGD2-sensitive
zone of the subarachnoid space of the basal forebrain, the number of c-Fos-positive
cells was significantly increased in the leptomeningeal membrane as well as in the
ventrolateral preoptic (VLPO) area, where increases were concomitant with the
induction of NREM sleep (Scammell et al. 2001; Satoh et al. 1999). In contrast,
58 Z.-L. Huang et al.
the number of c-Fos-positive neurons decreased notably in the TMN of the posterior
hypothalamus. The VLPO is known to send specific inhibitory GABAergic and
galaninergic projections to the TMN, the neurons of which contain the ascending
histaminergic arousal system (Sherin et al. 1998).
Inhibition of the histaminergic system induces sleep. In vivo microdialysis
experiments showed that infusion of an adenosine A2AR agonist, CGS-21680, into
the subarachnoid space of the basal forebrain inhibited the release of histamine in
both the frontal cortex and the medial preoptic area in a dose-dependent manner, and
induced the GABA release selectively in the TMN but not in the frontal cortex
(Hong et al. 2005). The inhibition of histamine release induced by CGS-21680 can
be blocked by perfusion with a GABAA antagonist, picrotoxin, in the TMN,
indicating that the A2AR agonist promotes sleep through inhibiting the histaminergic
system by inducing GABA release in the TMN. These results support the original
proposal of the flip-flop mechanism, in which sleep is promoted by activating the
sleep neurons in the VLPO and meanwhile inhibiting the histaminergic wake
neurons in the TMN (Saper et al. 2005).
60 Z.-L. Huang et al.
Fig. 3.4 Time course of changes in wakefulness after caffeine 15 mg/kg treatment in WT mice (a),
A2AR KO (b), A1R WT mice (c), and A1R KO mice (d). Each circle represents the hourly
mean SEM (n ¼ 5–7). The arrows indicate the injection time (9 a.m.). *P < 0.05; **P < 0.01,
significantly different from the vehicle, by the paired t-test. Adapted from Huang et al. (2005) with
modification and permission
The A1Rs are broadly expressed in the brain cortex, hippocampus, thalamus, lateral
hypothalamus, basal ganglia, and TMN (Oishi et al. 2008; Yaar et al. 2005; Thakkar
et al. 2002). Because of the extensive distribution of A1Rs in the cortex and the
inhibition of excitatory neurotransmission following presynaptic A1R activation, it
has been generally presumed that adenosine influences sleep mainly by the A1R. A
few findings from pharmacological studies are consistent with this hypothesis. For
example, i.p. or i.c.v. administration of the A1R selective agonist N6-
cyclopentyladenosine (CPA) to rats induces NREM sleep, inhibits REM sleep, and
increases changes in the NREM sleep EEG similar to those brought about by
prolonged wakefulness (Benington et al. 1995; Schwierin et al. 1996). Furthermore,
microdialysis perfusion with A1R antisense oligonucleotides in the basal forebrain of
rats decreases NREM sleep and increases wakefulness (Thakkar et al. 2003). In vitro
electrophysiological studies showed that adenosine can postsynaptically inhibit
basal forebrain neurons and cholinergic neurons in the laterodorsal tegmental nuclei
via A1R (Arrigoni et al. 2006; Rainnie et al. 1994). Christie et al. reported that sleep
62 Z.-L. Huang et al.
loss induces adenosine in the basal forebrain by A1R, which leads to sleepiness and
impaired vigilance (Christie et al. 2008). During the rat Psychomotor Vigilance Task
performance, response latencies and performance lapses of rats increased signifi-
cantly after adenosine was dialyzed in the basal forebrain of rats when compared
with baseline (no dialysis) or vehicle dialysis sessions. The codialysis of A1R
antagonist, 8-cyclopentyltheophylline with adenosine completely blocked the effects
produced by adenosine alone, resulting in performance equivalent to that of the
vehicle sessions. These results suggest that adenosine-induced sleep is mediated via
A1R in the cholinergic neurons in the basal forebrain. In addition, A1R binding was
increased after prolonged wakefulness/sleep deprivation in both humans (Basheer
et al. 2007) and rats (Elmenhorst et al. 2009), presumably caused by increases of
adenosine, as proved for the basal forebrain (Basheer et al. 2007).
Although local administration of adenosine or A1R agonist into the basal fore-
brain induces NREM sleep, infusion of the A1R agonist CPA into the lateral
ventricle does not affect NREM and REM sleep in mice (Urade et al. 2003),
indicating that activation of A1R in other brain areas may promote wakefulness.
Methippara et al. (2005) investigated the effects of an adenosine transport inhibitor,
NBTI, and A1R agonists/antagonists on sleep by microdialyzing them into the lateral
preoptic area. The results revealed that A1R activation or inhibition of adenosine
transport by NBTI increased wakefulness (Methippara et al. 2005). Moreover, the
homeostatic component of sleep–wake regulation is not affected in A1R knockout
animals (Stenberg et al. 2003). These findings indicate that adenosine influences
sleep–wake cycles in a site- and receptor-dependent manner and A1Rs are probably
not necessary for sleep homeostasis.
Blanco-Centurion et al. (2006) reported that adenosine levels in the basal fore-
brain did not increase after 6 h of prolonged wakefulness in rats with 95% cholin-
ergic neurons lesion in the basal forebrain. The lesion rats had an intact sleep drive
after 6 and 12 h of prolonged wakefulness. In the absence of cholinergic neurons in
the basal forebrain, another selective A1R agonist, N6-cyclohexyladenosine, effec-
tively increases sleep after administration to the basal forebrain. Therefore, neither
the activity of cholinergic neurons nor the accumulation of adenosine in the basal
forebrain during wakefulness is necessary for the sleep drive. However, Kalinchuk
et al. (2008) found that lesions of the cholinergic neurons in the basal forebrain
eliminated both the increase of adenosine levels and the homeostatic sleep drive.
These findings leave open the possibility raised by both studies that A1R may
influence sleep through noncholinergic neurons and that adenosine could affect
sleep via the brain regions other than the basal forebrain (Nooralam et al. 2006).
To understand the roles of noncholinergic and cholinergic neurons of the basal
forebrain in sleep–wake and EEG, as well as in homeostatic sleep regulation, Kaur
et al. (2008) ablated noncholinergic neurons in the basal forebrain with ibotenate,
and cholinergic neurons with 192-IgG saporin, and found that the noncholinergic
neurons in the basal forebrain can activate cortex through inhibiting delta waves, that
cholinergic neurons in the basal forebrain are not exclusive in promoting wakeful-
ness, and that both types of neurons in the basal forebrain are critical for the increases
in NREM sleep and EEG delta power induced by sleep deprivation.
3 Prostaglandins, Adenosine, and Histaminergic System in the Regulation. . . 63
Although many hypnotic and antihypnotic drugs are on the market, most of these
compounds show various unwanted side effects. The molecular mechanisms under-
lying sleep–wake regulation by adenosine described in this chapter may provide a
valid and practical approach for the development of drugs to mitigate sleep disorders
based on the pharmacological use of enzyme inhibitors and receptor-specific thera-
pies. The concept of using adenosine receptor agonists as modulators for sleep
disorders is fascinating; however, in practice, this approach could be used in a
brain region-dependent manner (Jacobson and Gao 2006). It need to be emphasized
that much more work is still necessary to clarify the detailed mechanisms of sleep–
wake regulation in terms of these mediators.
The histaminergic neurons are located in the TMN (Lin 2000; Brown et al. 2001) and
histamine can induce cortical arousal possibly either through direct cortical pro-
jections or by tonic control over the sleep-generating mechanisms in the preoptic
anterior hypothalamus (Lin et al. 1990, 1996). It was found that GABA exists in
most of the neurons in the TMN. Selective siRNA knockdown of the vesicular
GABA transporter (Vgat) in histaminergic neurons induced hyperactive mice that
have an extraordinary amount of wakefulness. Ablation of the Vgat gene throughout
the TMN sharpened this phenotype even further (Yu et al. 2015). Histamine was
found to promote wakefulness by the activation of H1R (Lin et al. 1988, 1990, 1996;
64
Fig. 3.5 (a) Time courses of NREM and REM sleep in rats administered adenosine (Ado) at 4.5 nmol/; (b) ADA inhibitor coformycin (CF) at 4 nmol/side; or
(c) A1R agonist CPA at 1.5 nmol/side. Values are means SEM (n ¼ 5–8). *P < 0.05; **P < 0.01, significantly different from the vehicle injection. Adapted
from Oishi et al. (2008) with modification and permission
Z.-L. Huang et al.
3 Prostaglandins, Adenosine, and Histaminergic System in the Regulation. . . 65
Monti et al. 1990; Monti 1993). H1R KO mice provide a useful tool to investigate
the role of the histaminergic system in the arousal effect induced by orexin A. H1R
KO mice were originally generated by Inoue et al. (1996), and their behavior and
neuropharmacological characteristics also have been extensively studied (Inoue
et al. 1996; Yanai et al. 1998). It is reported that H1R KO mice showed a significant
decrease in ambulation in an open field and on an activity wheel, however, no
electrophysiological study has been carried out yet. We found that orexin A can
significantly promote wakefulness in WT mice but not in H1R KO mice, although
both genotypes of mice displayed basically the same amounts of sleep and wake-
fulness under basal conditions. This suggests that H1R is indispensable for orexin
A-induced wakefulness (Huang et al. 2001).
3.9 Conclusions
References
Borb AA, Achermann P. Sleep homeostasis and models of sleep regulation. J Biol Rhythm.
1999;14:559–70.
Brown RE, Stevens DR, Haas HL. The physiology of brain histamine. Prog Neurobiol. 2001;63:
637–72.
Brundege JM, Diao L, Proctor WR, Dunwiddie TV. The role of cyclic AMP as a precursor of
extracellular adenosine in the rat hippocampus. Neuropharmacology. 1997;36:1201–10.
Burnstock G. Physiology and pathophysiology of purinergic neurotransmission. Physiol Rev.
2007;87:659–797.
Christie MA, et al. Microdialysis elevation of adenosine in the basal forebrain produces vigilance
impairments in the rat psychomotor vigilance task. Sleep. 2008;31:1393.
Deussen A, Lloyd HG, Schrader J. Contribution of S-adenosylhomocysteine to cardiac adenosine
formation. J Mol Cell Cardiol. 1989;21:773–82.
Dunwiddie TV, Masino SA. The role and regulation of adenosine in the central nervous system.
Annu Rev Neurosci. 2001;24:31–55.
El Yacoubi M, Ledent C, Parmentier M, Costentin J, Vaugeois J. Caffeine reduces hypnotic effects
of alcohol through adenosine A2A receptor blockade. Neuropharmacology. 2003;45:977.
Elmenhorst D, et al. Sleep deprivation increases A1 adenosine receptor binding in the human brain:
a positron emission tomography study. J Neurosci. 2007;27:2410–5.
Elmenhorst D, Basheer R, Mccarley RW, Bauer A. Sleep deprivation increases A1 adenosine
receptor density in the rat brain. Brain Res. 2009;1258:53–8.
Feldberg W, Sherwood S. Injections of drugs into the lateral ventricle of the cat. J Physiol.
1954;123:148–67.
Ferre S, et al. Adenosine A2A receptors in ventral striatum, hypothalamus and nociceptive circuitry
Implications for drug addiction, sleep and pain. Prog Neurobiol. 2007;83:332–47.
Franken P, Chollet D, Tafti M. The homeostatic regulation of sleep need is under genetic control. J
Neurosci. 2001;21:2610–21.
Fredholm B. Adenosine, an endogenous distress signal, modulates tissue damage and repair. Cell
Death Differ. 2007;14:1315–23.
Fredholm BB, IJzerman AP, Jacobson KA, Klotz K-N, Linden J. International union of pharma-
cology. XXV Nomenclature and classification of adenosine receptors. Pharmacol Rev. 2001;53:
527–52.
Fredholm BB, Chen J-F, Cunha RA, Svenningsson P, Vaugeois J-M. Adenosine and brain function.
Int Rev Neurobiol. 2005;63:191–270.
Gallopin T, et al. The endogenous somnogen adenosine excites a subset of sleep-promoting neurons
via A2A receptors in the ventrolateral preoptic nucleus. Neuroscience. 2005;134:1377.
Geiger JD, Fyda DM. Adenosine transport in nervous system tissues. In: Adenosine in the nervous
system. New York: Academic Press; 1991. p. 1–23.
Gerashchenko D, Okano Y, Urade Y, Inoué S, Hayaishi O. Strong rebound of wakefulness follows
prostaglandin D2-or adenosine A2a receptor agonist-induced sleep. J Sleep Res. 2000;9:81–7.
Gu J, Foga I, Parkinson F, Geiger J. Involvement of Bidirectional Adenosine Transporters in the
Release of l-[3H] Adenosine from Rat Brain Synaptosomal Preparations. J Neurochem.
1995;64:2105–10.
Halassa MM, et al. Astrocytic modulation of sleep homeostasis and cognitive consequences of sleep
loss. Neuron. 2009;61:213–9.
Hong ZY, et al. An adenosine A2A receptor agonist induces sleep by increasing GABA release in
the tuberomammillary nucleus to inhibit histaminergic systems in rats. J Neurochem. 2005;92:
1542–9.
Huang Z-L, et al. Arousal effect of orexin A depends on activation of the histaminergic system. Proc
Natl Acad Sci U S A. 2001;98:9965–70. https://doi.org/10.1073/pnas.181330998.
Huang Z-L, et al. Adenosine A2A, but not A1, receptors mediate the arousal effect of caffeine. Nat
Neurosci. 2005;8:858–9.
Huang Z-L, Urade Y, Hayaishi O. Prostaglandins and adenosine in the regulation of sleep and
wakefulness. Curr Opin Pharmacol. 2007;7:33–8.
3 Prostaglandins, Adenosine, and Histaminergic System in the Regulation. . . 67
Huston J, et al. Extracellular adenosine levels in neostriatum and hippocampus during rest and
activity periods of rats. Neuroscience. 1996;73:99–107.
Inoue I, et al. Impaired locomotor activity and exploratory behavior in mice lacking histamine H1
receptors. Proc Natl Acad Sci U S A. 1996;93:13316–20.
Islam F, Watanabe Y, Morii H, Hayaishi O. Inhibition of rat brain prostaglandin D synthase by
inorganic selenocompounds. Arch Biochem Biophys. 1991;289:161–6.
Jacobson KA, Gao Z-G. Adenosine receptors as therapeutic targets. Nat Rev Drug Discov. 2006;5:
247–64.
Jones BE. Glia, adenosine, and sleep. Neuron. 2009;61:156–7.
Jordan W, et al. Prostaglandin D synthase (β-trace) in healthy human sleep. Sleep. 2004;27:867–74.
Jourdain P, et al. Glutamate exocytosis from astrocytes controls synaptic strength. Nat Neurosci.
2007;10:331–9.
Kalinchuk AV, McCarley RW, Stenberg D, Porkka-Heiskanen T, Basheer R. The role of cholin-
ergic basal forebrain neurons in adenosine-mediated homeostatic control of sleep: lessons from
192 IgG–saporin lesions. Neuroscience. 2008;157:238–53.
Kalinchuk A, McCarley R, Porkka-Heiskanen T, Basheer R. Sleep deprivation triggers inducible
nitric oxide-dependent nitric oxide production in wake-active basal forebrain neurons. J
Neurosci. 2010;30:13254.
Kaur S, Junek A, Black MA, Semba K. Effects of ibotenate and 192IgG-saporin lesions of the
nucleus basalis magnocellularis/substantia innominata on spontaneous sleep and wake states
and on recovery sleep after sleep deprivation in rats. J Neurosci. 2008;28:491–504.
Latini S, Pedata F. Adenosine in the central nervous system: release mechanisms and extracellular
concentrations. J Neurochem. 2001;79:463–84.
Li R, et al. Activation of adenosine A2A receptors in the olfactory tubercle promotes sleep in
rodents. Neuropharmacology. 2020;168:107923. https://doi.org/10.1016/j.neuropharm.2019.
107923.
Lin JS. Brain structures and mechanisms involved in the control of cortical activation and wake-
fulness, with emphasis on the posterior hypothalamus and histaminergic neurons. Sleep Med
Rev. 2000;4:471–503. https://doi.org/10.1053/smrv.2000.0116.
Lin JS, Sakai K, Jouvet M. Evidence for histaminergic arousal mechanisms in the hypothalamus of
cat. Neuropharmacology. 1988;27:111–22.
Lin JS, et al. Involvement of histaminergic neurons in arousal mechanisms demonstrated with
H3-receptor ligands in the cat. Brain Res. 1990;523:325–30.
Lin JS, Hou Y, Sakai K, Jouvet M. Histaminergic descending inputs to the mesopontine tegmentum
and their role in the control of cortical activation and wakefulness in the cat. J Neurosci.
1996;16:1523–37.
Marks G, Shaffery J, Speciale S, Birabil C. Enhancement of rapid eye movement sleep in the rat by
actions at A1 and A2a adenosine receptor subtypes with a differential sensitivity to atropine.
Neuroscience. 2003;116:913–20.
Matsumura H, Takahata R, Hayaishi O. Inhibition of sleep in rats by inorganic selenium com-
pounds, inhibitors of prostaglandin D synthase. Proc Natl Acad Sci U S A. 1991;88:9046–50.
Methippara MM, Kumar S, Alam MN, Szymusiak R, McGinty D. Effects on sleep of microdialysis
of adenosine A1 and A2a receptor analogs into the lateral preoptic area of rats. Am J Phys Regul
Integr Comp Phys. 2005;289:R1715–23.
Missale C, Nash SR, Robinson SW, Jaber M, Caron MG. Dopamine receptors: from structure to
function. Physiol Rev. 1998;78:189–225.
Mizoguchi A, et al. Dominant localization of prostaglandin D receptors on arachnoid trabecular
cells in mouse basal forebrain and their involvement in the regulation of non-rapid eye
movement sleep. Proc Natl Acad Sci U S A. 2001;98:11674–9.
Mohri I, Eguchi N, Suzuki K, Urade Y, Taniike M. Hematopoietic prostaglandin D synthase is
expressed in microglia in the developing postnatal mouse brain. Glia. 2003;42:263–74.
Monti JM. Involvement of histamine in the control of the waking state. Life Sci. 1993;53:1331–8.
68 Z.-L. Huang et al.
Monti JM, Orellana C, Boussard M, Jantos H, Olivera S. Sleep variables are unaltered by
zolantidine in rats: are histamine H2-receptors not involved in sleep regulation? Brain Res
Bull. 1990;25:229–31.
Monti J, Pandi-Perumal SR, Sinton CM, Sinton CW. Neurochemistry of sleep and wakefulness.
Cambridge: Cambridge University Press; 2008.
Murillo-Rodriguez E, Blanco-Centurion C, Gerashchenko D, Salin-Pascual R, Shiromani P. The
diurnal rhythm of adenosine levels in the basal forebrain of young and old rats. Neuroscience.
2004;123:361–70.
Nagy J, LaBella L, Buss M, Daddona P. Immunohistochemistry of adenosine deaminase: implica-
tions for adenosine neurotransmission. Science. 1984;224:166–8.
Narumiya S, Ogorochi T, Nakao K, Hayaishi O. Prostaglandin D2 in rat brain, spinal cord and
pituitary: basal level and regional distribution. Life Sci. 1982;31:2093.
Nooralam M, Szymusiak R, McGinty D. Adenosinergic regulation of sleep: multiple sites of action
in the brain. Sleep. 2006;29:1384–5.
Ogorochi T, et al. Regional distribution of prostaglandins D2, E2, and F2α and related enzymes in
postmortem human brain. J Neurochem. 1984;43:71–82.
Oishi Y, Huang Z-L, Fredholm BB, Urade Y, Hayaishi O. Adenosine in the tuberomammillary
nucleus inhibits the histaminergic system via A1 receptors and promotes non-rapid eye move-
ment sleep. Proc Natl Acad Sci U S A. 2008;105:19992–7.
Oishi Y, et al. Slow-wave sleep is controlled by a subset of nucleus accumbens core neurons in
mice. Nat Commun. 2017;8:734. https://doi.org/10.1038/s41467-017-00781-4.
Okada T, et al. Dominant localization of adenosine deaminase in leptomeninges and involvement of
the enzyme in sleep. Biochem Biophys Res Commun. 2003;312:29–34.
Onoe H, et al. Prostaglandin D2, a cerebral sleep-inducing substance in monkeys. Proc Natl Acad
Sci U S A. 1988;85:4082–6.
Pandey HP, Ram A, Matsumura H, Hayaishi O. Concentration of prostaglandin D2 in cerebrospinal
fluid exhibits a circadian alteration in conscious rats. Biochem Mol Biol Int. 1995;37:431–7.
Pascual O, et al. Astrocytic purinergic signaling coordinates synaptic networks. Science. 2005;310:
113–6.
Peng L, et al. Nucleoside transporter expression and function in cultured mouse astrocytes. Glia.
2005;52:25–35.
Peng W, et al. Regulation of sleep homeostasis mediator adenosine by basal forebrain glutamatergic
neurons. Science. 2020;369:eabb0556. https://doi.org/10.1126/science.abb0556.
Porkka-Heiskanen T, et al. Adenosine: a mediator of the sleep-inducing effects of prolonged
wakefulness. Science. 1997;276:1265–8.
Porkka-Heiskanen T, Strecker R, McCarley R. Brain site-specificity of extracellular adenosine
concentration changes during sleep deprivation and spontaneous sleep: an in vivo microdialysis
study. Neuroscience. 1999;99:507–17.
Qiu M, et al. D (1)/D (2) receptor-targeting L-stepholidine, an active ingredient of the Chinese herb
Stephonia, induces non-rapid eye movement sleep in mice. Pharmacol Biochem Behav.
2009;94:16.
Qu W-M, et al. Lipocalin-type prostaglandin D synthase produces prostaglandin D2 involved in
regulation of physiological sleep. Proc Natl Acad Sci U S A. 2006;103:17949–54.
Qu W-M, Huang Z-L, Xu X-H, Matsumoto N, Urade Y. Dopaminergic D1 and D2 receptors are
essential for the arousal effect of modafinil. J Neurosci. 2008;28:8462–9.
Qu W-M, et al. Essential role of dopamine D2 receptor in the maintenance of wakefulness, but not
in homeostatic regulation of sleep, in mice. J Neurosci. 2010;30:4382–9.
Radek RJ, Decker MW, Jarvis MF. The adenosine kinase inhibitor ABT-702 augments EEG slow
waves in rats. Brain Res. 2004;1026:74–83.
Radulovacki M, Virus R, Djuricic-Nedelson M, Green R. Hypnotic effects of deoxycorformycin in
rats. Brain Res. 1983;271:392–5.
Rai S, et al. A1 receptor mediated adenosinergic regulation of perifornical-lateral hypothalamic area
neurons in freely behaving rats. Neuroscience. 2010;167:40.
3 Prostaglandins, Adenosine, and Histaminergic System in the Regulation. . . 69
Rainnie DG, Grunze HC, McCarley RW, Greene RW. Adenosine inhibition of mesopontine
cholinergic neurons: implications for EEG arousal. Science (New York, NY). 1994;263:689.
Ram A, et al. CSF levels of prostaglandins, especially the level of prostaglandin D2, are correlated
with increasing propensity towards sleep in rats. Brain Res. 1997;751:81.
Retey J, et al. A functional genetic variation of adenosine deaminase affects the duration and
intensity of deep sleep in humans. Proc Natl Acad Sci U S A. 2005;102:15676–81.
Retey J, et al. A genetic variation in the adenosine A2A receptor gene (ADORA2A) contributes to
individual sensitivity to caffeine effects on sleep. Clin Pharmacol Ther. 2007;81:692–8.
Rosenberg PA, Li Y. Adenylyl cyclase activation underlies intracellular cyclic AMP accumulation,
cyclic AMP transport, and extracellular adenosine accumulation evoked by β-adrenergic recep-
tor stimulation in mixed cultures of neurons and astrocytes derived from rat cerebral cortex.
Brain Res. 1995;692:227–32.
Sala-Newby GB, Skladanowski AC, Newby AC. The mechanism of adenosine formation in cells
cloning of cytosolic 50 -nucleotidase-I. J Biol Chem. 1999;274:17789–93.
Saper CB, Scammell TE, Lu J. Hypothalamic regulation of sleep and circadian rhythms. Nature.
2005;437:1257–63.
Satoh S, Matsumura H, Suzuki F, Hayaishi O. Promotion of sleep mediated by the A2a-adenosine
receptor and possible involvement of this receptor in the sleep induced by prostaglandin D2 in
rats. Proc Natl Acad Sci U S A. 1996;93:5980–4.
Satoh S, Matsumura H, Hayaishi O. Involvement of adenosine A2A receptor in sleep promotion.
Eur J Pharmacol. 1998;351:155–62.
Satoh S, et al. Region-dependent difference in the sleep-promoting potency of an adenosine A2A
receptor agonist. Eur J Neurosci. 1999;11:1587–97.
Sawynok J, Liu XJ. Adenosine in the spinal cord and periphery: release and regulation of pain. Prog
Neurobiol. 2003;69:313–40.
Scammell T, et al. An adenosine A2a agonist increases sleep and induces Fos in ventrolateral
preoptic neurons. Neuroscience. 2001;107:653–63.
Schiffmann S, Fisone G, Moresco R, Cunha R, Ferre S. Adenosine A2A receptors and basal ganglia
physiology. Prog Neurobiol. 2007;83:277–92.
Schwierin B, Borbely A, Tobler I. Effects of N6-cyclopentyladenosine and caffeine on sleep
regulation in the rat. Eur J Pharmacol. 1996;300:163–72.
Sherin JE, Elmquist JK, Torrealba F, Saper CB. Innervation of histaminergic tuberomammillary
neurons by GABAergic and galaninergic neurons in the ventrolateral preoptic nucleus of the rat.
J Neurosci. 1998;18:4705–21.
Stenberg D, et al. Sleep and its homeostatic regulation in mice lacking the adenosine A1 receptor. J
Sleep Res. 2003;12:283–90.
Strecker RE, et al. Adenosinergic modulation of basal forebrain and preoptic/anterior hypothalamic
neuronal activity in the control of behavioral state. Behav Brain Res. 2000;115:183–204.
Takahata R, et al. Intravenous administration of inorganic selenium compounds, inhibitors of
prostaglandin D synthase, inhibits sleep in freely moving rats. Brain Res. 1993;623:65–71.
Thakkar MM, Winston S, McCarley RW. Orexin neurons of the hypothalamus express adenosine
A1 receptors. Brain Res. 2002;944:190–4.
Thakkar MM, Winston S, McCarley RW. A1 receptor and adenosinergic homeostatic regulation of
sleep-wakefulness: effects of antisense to the A1 receptor in the cholinergic basal forebrain. J
Neurosci. 2003;23:4278–87.
Thakkar M, Engemann S, Walsh K, Sahota P. Adenosine and the homeostatic control of sleep:
effects of A1 receptor blockade in the perifornical lateral hypothalamus on sleep–wakefulness.
Neuroscience. 2008;153:875–80.
Ueno R, Ishikawa Y, Nakayama T, Hayaishi O. Prostaglandin D2 induces sleep when microinjected
into the preoptic area of conscious rats. Biochem Biophys Res Commun. 1982;109:576.
Ueno R, Honda K, Inoué S, Hayaishi O. Prostaglandin D2, a cerebral sleep-inducing substance in
rats. Proc Natl Acad Sci U S A. 1983;80:1735–7.
70 Z.-L. Huang et al.
Urade Y, Hayaishi O. Crucial role of prostaglandin D2 and adenosine in sleep regulation: exper-
imental evidence from pharmacological approaches to gene-knockout mice. Future Neurol.
2010;5:363–76.
Urade Y, et al. Dominant expression of mRNA for prostaglandin D synthase in leptomeninges,
choroid plexus, and oligodendrocytes of the adult rat brain. Proc Natl Acad Sci. 1993;90:9070–
4.
Urade Y, et al. Sleep regulation in adenosine A2A receptor-deficient mice. Neurology. 2003;61:
S94–6.
Wang YQ, et al. Adenosine A2A receptors in the olfactory bulb suppress rapid eye movement sleep
in rodents. Brain Struct Funct. 2017;222:1351–66. https://doi.org/10.1007/s00429-016-1281-2.
Wang P, et al. Lipocalin-type prostaglandin D synthase levels increase in patients with narcolepsy
and idiopathic hypersomnia. Sleep. 2021;44:zsaa234. https://doi.org/10.1093/sleep/zsaa234.
Yaar R, Jones M, Chen JF, Ravid K. Animal models for the study of adenosine receptor function. J
Cell Physiol. 2005;202:9–20.
Yanai K, et al. Behavioural characterization and amounts of brain monoamines and their metabo-
lites in mice lacking histamine H1 receptors. Neuroscience. 1998;87:479–87.
Yu X, et al. Wakefulness is governed by GABA and histamine cotransmission. Neuron. 2015;87:
164–78. https://doi.org/10.1016/j.neuron.2015.06.003.
Yuan XS, et al. Striatal adenosine A2A receptor neurons control active-period sleep via
parvalbumin neurons in external globus pallidus. elife. 2017;6:e29055. https://doi.org/10.
7554/eLife.29055.
Zhang Z, et al. Regulated ATP release from astrocytes through lysosome exocytosis. Nat Cell Biol.
2007;9:945–53.
Zimmermann H. Extracellular metabolism of ATP and other nucleotides. Naunyn Schmiedeberg’s
Arch Pharmacol. 2000;362:299–309.
Zimmermann H, Braun N, Kegel B, Heine P. New insights into molecular structure and function of
ecto-nucleotidases in the nervous system. Neurochem Int. 1998;32:421–5.
Chapter 4
Sleep and Neuronal Plasticity
Marcos G. Frank
Abstract Sleep is considered to play an important role in neuronal plasticity yet the
underlying mechanisms remain controversial. In the last two decades, scientists have
made important advances in understanding these mechanisms in the developing and
adult brain. An emergent view is that sleep influences a number of types of plasticity
and this is determined by a number of factors, including preceding experience, and
ontogenetic status. Several theories of sleep function have also been proposed that
integrate older ideas about sleep and more recent discoveries in the field of synaptic
plasticity. In this chapter, I discuss these key findings and current theories that posit
different roles for sleep in neuronal plasticity.
4.1 Introduction
Sleep has long been suspected to play an important role in neuronal plasticity.
Scientists historically conceptualized and investigated the problem in terms of
what was known about classic Hebbian forms of plasticity such as long-term
synaptic potentiation (LTP) and depression (LTD) [reviewed in Benington and
Frank (2003) and Frank and Benington (2006)]. LTP and LTD refer to
use-dependent, persistent alterations in synaptic weights that strengthen (LTP) or
weaken (LTD)-specific synapses, respectively (Malenka and Bear 2004). They are
considered Hebbian because they are associative, input (synapse) specific and
require coordinated pre- and post-synaptic activity (Markram et al. 2011). Today,
LTP and LTD are believed to be cellular correlates (if not the substrates) of learning
and memory (Malenka and Bear 2004; Bear and Malenka 1994).
M. G. Frank (*)
Elson S. Floyd College of Medicine, Washington State University Spokane, Spokane, WA,
USA
e-mail: marcos.frank@wsu.edu
© Springer Nature B.V. and Shanghai Jiao Tong University Press 2022 71
A. I. Pack, Q. Y. Li (eds.), Sleep and its Disorders, Translational Medicine Research,
https://doi.org/10.1007/978-94-024-2168-2_4
72 M. G. Frank
More recently sleep has also been proposed to promote non-Hebbian (“homeo-
static”) forms of plasticity (Tononi and Cirelli 2003, 2006). In contrast to LTP and
LTD, these forms of plasticity can adjust all synapses in a neuron or network of
neurons upward or downward in response to global changes in activity (Turrigiano
1999, 2008; Pozo and Goda 2010). This type of plasticity is proposed to offset pure
Hebbian mechanisms in the brain, that if left unchecked would saturate synaptic
strength and prevent further learning. Homeostatic plasticity was originally
described in the late 1990s by non-sleep scientists [for review see Nelson and
Turrigiano (2008)], and conceptually incorporated into what came to be known as
the “synaptic homeostasis hypothesis” (SHY) in 2003 (Tononi and Cirelli 2001,
2003, 2006).
In the following sections, I discuss our current knowledge concerning interactions
between sleep and Hebbian and non-Hebbian synaptic plasticity in the developing
and adult brain. I also discuss these findings in the context of two theories of sleep
function that incorporate Hebbian and non-Hebbian forms of plasticity in
different ways.
Fig. 4.1 Effect of sleep on the magnitude of the ocular dominance shift induced by monocular
deprivation (MD). The first two columns show the experimental conditions of all kittens. The right-
most column schematically shows ocular dominance maps obtained from primary visual cortex
under various conditions. All kittens were monocularly deprived for 6 h, and one group was tested
immediately afterward (MD6h). A second group was allowed to sleep ad-lib during the following
6 h (MD+S), while a third group was kept awake in the dark (MD S). A fourth group was deprived
for 12 h and then tested (MD12h). The ocular dominance maps obtained by intrinsic-signal imaging
4 Sleep and Neuronal Plasticity 75
ERK) has no effect on circuit weakening in wakefulness, but when conducted during
subsequent sleep, this completely blocked sleep-dependent ODP (Seibt et al. 2012;
Dumoulin et al. 2015). Additional studies of cortical genes and proteins showed that
while plasticity-related mRNAs are upregulated by visual experience, they are not
translated into proteins until sleep occurs (Seibt et al. 2012). This suggests that the
transcription and translation of plasticity-related mRNAs are divided across sleep
and wake.
A two-stage process in ODP is further demonstrated by a study of single-neuron
activity in freely behaving cats. Aton et al. (2013) used chronic, stereotrode record-
ing of single visual cortical neurons to track their activity and interactions before,
during, and after a period of MD. In contrast to previous studies employing similar
longitudinal recording (Mioche and Singer 1989), neuronal activity was also
recorded across the sleep–wake cycle. MD in the awake animal caused a large
reduction in firing rate in fast-spiking neurons (i.e., putative GABAergic cells) in
the visual cortex. This decrease in activity was maintained during the first 6 h of
post-MD sleep and accompanied by an increase in firing in regular spiking (i.e.,
putative excitatory neurons). The decrease in fast-spiking activity was also propor-
tional to plastic changes in regular spiking neurons observed after sleep. This
suggests that in addition to changes in the deprived eye pathway, MD alters
intracortical inhibition which contributes to sleep-dependent changes in excitatory
circuits.
REM sleep has also been shown to influence a form of LTP in vitro that parallels the
critical period for ODP in rodents. In this type of LTP, high-frequency white matter
stimulation in cortical slices prepared from postnatal (P) day 28–30 rats produces
synaptic potentiation in cortical layers II/III. This form of LTP is only observed
during these particular ages and not in cortical slices from adult rats (Kirkwood et al.
1995). Shaffery et al. (2002) reported that 1 week of REM sleep deprivation
prolonged the critical period for the developmentally regulated form of LTP. Similar
manipulations of older animals outside of the critical period did not show the same
results. These investigators showed that this plasticity could be partially rescued if
REM sleep deprivation was administered near (or overlapping) the end of the critical
period (Shaffery et al. 2006; Shaffery and Roffwarg 2003). The effects of REM sleep
⁄
Fig. 4.1 (continued) display cortical regions dominated by the deprived eye in black, and those
dominated by the non-deprived eye in white. The MD+S group shows a loss of territory dominated
by the deprived eye well beyond that observed in the MD6h group, while the sleep-deprived group
(MD S) does not. In fact, the consolidation of the MD shift in the MD+S group amounts to about
the same magnitude as is observed after 12 h of monocular deprivation (MD12h). Reproduced with
permission from Sengpiel (2001)
76 M. G. Frank
The role of sleep in adult synaptic plasticity has historically been investigated using
classic tetany-based forms of Hebbian LTP and LTD [reviewed in Benington and
Frank (2003) and Hennevin et al. (2007)] in different brain states, or in slices of
brains after periods of sleep or sleep deprivation. Overall, hippocampal LTP can be
induced during REM sleep, whereas similar stimulus protocols during NREM sleep
have inconsistent effects or produce LTD [reviewed in Benington and Frank (2003)
and Hennevin et al. (2007)]. Conversely, sleep deprivation impairs the induction or
maintenance of LTP in vivo and in vitro, although this varies depending on the brain
area examined. For example, REM sleep deprivation and total sleep deprivation
impair the maintenance of LTP in the hippocampus but enhance this process in the
medial prefrontal cortex (Romcy-Pereira and Pavlides 2004). The effects of sleep
loss on hippocampal LTP in vivo have been replicated in other studies but the timing
of this effect varies, with some studies showing delayed impact on LTP (Marks and
Wayner 2005; Kim et al. 2005).
A large number of studies also show that in vitro hippocampal LTP (either the
induction or the maintenance) is reduced in rodents that undergo varying amounts of
REM sleep deprivation, total sleep deprivation, or sleep restriction prior to sacrifice
(Kopp et al. 2006; Arrigoni et al. 2009; Campbell et al. 2002; Davis et al. 2003;
Ishikawa et al. 2006; McDermott et al. 2003, 2006; Ravassard et al. 2006, 2009;
Tartar et al. 2006; Florian et al. 2011; Vecsey et al. 2009; Chen et al. 2006).
Interestingly, when REM sleep is restored (after prior deprivation) or increased in
rodents, this reverses deficits in hippocampal LTP (Ravassard et al. 2009, 2016).
The underlying mechanisms mediating the effects of sleep loss on LTP are not
completely understood. However, they do not appear to be simply due to indirect
effects of the sleep deprivation procedures. For example, these deficits can be
dissociated from changes in stress hormones (Kopp et al. 2006; Ravassard et al.
2009, 2016). Diminished plasticity may instead be linked to decrements in hippo-
campal NMDA receptor function (Kopp et al. 2006; McDermott et al. 2006; Chen
et al. 2006; Longordo et al. 2009) and ERK/MAPK activation (Ravassard et al.
2009) combined with reductions in plasticity-related mRNAs or proteins (Ravassard
4 Sleep and Neuronal Plasticity 77
et al. 2016; Guzman-Marin et al. 2006; Davis et al. 2006), and elevated concentra-
tions of PDE4 (Vecsey et al. 2009) and extracellular adenosine (Arrigoni et al. 2009;
Florian et al. 2011).
Sleep also promotes naturally occurring forms of synaptic potentiation in the adult
visual and motor cortex. Stimulus specific-response plasticity (SRP) is a form of
in vivo LTP that manifests as a potentiated response to an experienced (trained)
visual stimulus (Cooke and Bear 2010). SRP is only observed after a period of sleep,
and suppressed by sleep deprivation (Aton et al. 2014). A follow-up investigation
(Durkin and Aton 2016) showed that these changes could not be explained as a form
of “relative” synaptic weakening (Cirelli and Tononi 2015).
Interestingly, motor learning in adult mice promotes morphological signs of
synaptic potentiation. After the learning experience, sleep promotes the formation
of dendritic spines in motor cortex in a dendritic branch-dependent manner (Yang
et al. 2014). Similar sleep-dependent changes are reported in the hippocampus. Sleep
deprivation reduces structural changes in synapses normally induced by learning.
Conversely, during sleep these synaptic structures grow and expand (Havekes et al.
2016). However, these latter effects also appear to be highly dependent on the
dendritic branches and hippocampal regions under examination [reviewed in Frank
(2021)].
sets of circuits (ripples and sharp waves) (Buzsaki 1996; Schwindel and McNaugh-
ton 2011; Pfeiffer 2020).
The phenomenon appears quite robust, as variants have been found in the rodent
hippocampus, ventral striatum, and cortex (Yang et al. 2014; Wilson and McNaugh-
ton 1994; Skaggs and McNaughton 1996; Lee and Wilson 2002; Louie and Wilson
2001; Kudrimoti et al. 1999; Pennartz et al. 2004; Ji and Wilson 2007). Forms of
replay have been found in an impressive number of animal species, ranging from
birds (Dave and Margoliash 2000), cats (Dumoulin Bridi et al. 2015) to primates
(Hoffman and McNaughton 2002) and based on brain imaging, humans (Maquet
et al. 2000; Peigneux et al. 2004; Deuker et al. 2013). The animal studies are also
embedded in well-established paradigms of behavior, cellular physiology, and
plasticity [reviewed in Schwindel and McNaughton (2011), Pfeiffer (2020), and
Girardeau and Zugaro (2011)]. Although there is some evidence that forms of replay
occur during REM sleep (Dumoulin Bridi et al. 2015; Louie and Wilson 2001; Poe
et al. 2000), communication between the hippocampus and cortex is generally
conjectured to occur during NREM sleep. This is because during this state activity
in the hippocampus is consistent with outflow, rather than inflow (Buzsaki 1996;
Diekelmann and Born 2010; Graves et al. 2001; Hasselmo 1999). There are indeed
interesting correlations between ripples and sharp waves (hippocampal events when
a replay is reported) and thalamocortical spindles and cortical slow waves consistent
with this hypothesis (Siapas and Wilson 1998; Battaglia et al. 2004; Sirota et al.
2003). In addition, though quite rare, there are instances when hippocampal and
cortical replay occurs simultaneously (Ji and Wilson 2007; Qin et al. 1997).
ephemeral nature of replay. It is typically detectable only within the first 20–30 min
of sleep and then fades away. In some measures, it also accounts for only a fraction
of total variance in neuronal activity [reviewed in Frank (2007)].
The effects of novel experience on replay are less explored. Some studies show
that neuronal activity patterns associated with “novel” experience can appear in
sleep, but the novel tasks are often very similar to familiar tasks. For example, in one
study there was substantial overlap in cells active in the familiar versus novel maze
configurations (between 70 and 77%) (Kudrimoti et al. 1999). This issue seemed to
be resolved by studies reporting novelty-induced reactivation of waking activity
patterns in the sleeping rat forebrain (Ribeiro et al. 2004; Ribeiro 2007), but these
findings have been challenged on technical and methodological grounds (Tatsuno
et al. 2006). More recent findings, however, indicate that replay can occur following
a novel experience. In one study, rats were exposed to novel learning rules, and
medial prefrontal cortex ensemble recordings showed that patterns of activity
induced by learning “replayed” in subsequent NREM sleep (Peyrache et al. 2009).
Similar results were reported in rats trained on a brain–machine interface. In this
study, neuronal ensembles recruited in a learning task showed greater “reactivation”
during NREM sleep after only a few learning trials (Gulati et al. 2014).
A final consideration is whether replay in sleep actually has any function. Two
independent studies in rodents provide evidence that interrupting the hippocampal
bursts that convey replay impairs critically important behavior (learning and mem-
ory) (Ego-Stengel and Wilson 2010; Girardeau et al. 2009). These studies must be
cautiously interpreted because they involved disruption of the hippocampal ripples
and sharp waves, and not replay per se. It is also not clear if similar results would be
obtained if disruption were restricted to replay in wakefulness vs. sleep. Hippocam-
pal replay during sleep can also be triggered by presentation of auditory tones
present during experience—which suggests that replay represents a memory trace
(Bendor and Wilson 2012). The imposition of waking patterns of activity (associated
with a conditioned stimulus) in the olfactory bulb during sleep led to better perfor-
mance in an aversive training task (Barnes and Wilson 2014). Interestingly, similar
experiments in humans lead to greater performance on memory tasks (Schonauer
et al. 2014), and spontaneous replay can predict future performance (Deuker et al.
2013). These results strongly suggest that replay induces adaptive, functional plastic
changes in the brain.
Sleep has been variously hypothesized to stabilize (Kavanau 1996; Krueger and
Obal 1993), strengthen (Datta and Patteron 2003), or remove synapses (Crick and
Mitchison 1983; Giuditta et al. 1995). The synaptic Homeostasis Hypothesis (SHY)
is the most recent version of the latter idea. SHY proposes that sleep promotes “net”
synaptic weakening, which offsets synaptic strengthening during wakefulness
(Tononi and Cirelli 2003, 2006, 2014). Theoretically, this preserves the relative
80 M. G. Frank
strength between synapses, allows for further synaptic changes and prevents mal-
adaptive metabolic costs associated with excessive synaptogenesis or synapse main-
tenance. Therefore, SHY predicts that, overall, synapses should be weaker, not
stronger, after sleep.
As reviewed elsewhere (Tononi and Cirelli 2014), a number of findings are
consistent with SHY (but see (Frank 2021; Frank 2012; Frank and Cantera 2014;
Timofeev and Chauvette 2017). There are several changes in proteins, synaptic
efficacy, and dendrite morphology consistent with predictions of SHY (Vyazovskiy
et al. 2008; Maret et al. 2011; Liu et al. 2010). Briefly, markers of synaptic
potentiation (e.g., changes in AMPAR subunit number or phosphorylation) are
elevated in the brains of adult rats sacrificed at the end of the active phase (or after
sleep deprivation), relative to animals sacrificed at the end of the rest phase
(Vyazovskiy et al. 2008). Similar results are reported for measures of synaptic
efficacy (EPSPs and mini EPSPs), which are also elevated at the end of the active
phase (or after sleep deprivation) relative to sleep (Vyazovskiy et al. 2008; Liu et al.
2010). Two imaging studies of cortical dendrite spine morphology showed that the
ratio of spines eliminated vs. those formed was greater after a period of sleep than a
period of wakefulness (Maret et al. 2011; Yang and Gan 2012). Interestingly, these
results were restricted to stages of development when there is an overall pruning of
synapses, and were entirely absent in adult mice (Maret et al. 2011). It was then
shown using electron microscopy in fixed mouse tissue (layer 2–3 of the cortex) that
many synapses shrink in size when examined after a long period of sleep, relative to
sleep deprivation or the wake phase (de Vivo et al. 2017). In Drosophila, pre- and
postsynaptic proteins and proteins involved in neurotransmitter release are elevated
in the brain after extended waking periods or sleep deprivation (relative to sleep)
(Gilestro et al. 2009). Additional studies showed that presynaptic structures, axonal
arbors, and postsynaptic spines in Drosophila neurons expanded after extended
waking periods (or sleep deprivation); a process also reversed by extended periods
of sleep (Bushey et al. 2011). Similar results were observed in a separate study by
Donlea et al. (2011).
mechanism in SHY has been described using the same language used by scientists
who discovered synaptic scaling: “SWA, in turn, would promote synaptic down-
scaling (Turrigiano 1999)” [pg. 4503 (Cirelli et al. 2005)]. The consequences of
unchecked synaptic potentiation in SHY are also similar to the network instability
described in synaptic downscaling: “Sleep, and the accompanying downscaling of
synapses, would then be needed to interrupt the growth of synaptic strength associ-
ated with waking and prevent synaptic overload ” [pg. 55 (Tononi and Cirelli 2006)].
The renaming of this process to “synaptic renormalization” (Tononi and Cirelli
2012) did not change its basic similarities to synaptic scaling. Like the original
descriptions of synaptic downscaling, synaptic renormalization affects all or most
synapses and offsets LTP (or LTP-like plasticity) (Tononi and Cirelli 2003, 2006,
2014). Selective down-selection involves a modest variation in this concept by
adding the idea that some synapses are protected against this renormalization
process. Therefore, despite the changing names, it is reasonable to ask whether
sleep primarily promotes synaptic downscaling or other forms of non-Hebbian
synaptic weakening.
As reviewed elsewhere (Frank 2012), many molecular and electrophysiological
changes reported across the sleep–wake cycle are inconsistent with a primary
synaptic downscaling function for sleep. For example, it has been suggested that
global decreases in cortical activity (down-states) that occur during NREM sleep
might downscale synapses. However, the basic principle of synaptic scaling is that
global decreases in neuronal activity upscale synapses, while increases in neuronal
activity downscale synapses. Consequently, down-states in NREM sleep should
upscale, not downscale, synapses. Similarly, the neural expression of scaling factors
(Arc, Homer 1a, and tumor necrosis factor [tnfα]) across the sleep–wake cycle is
inconsistent with downscaling during sleep [reviewed in Frank (2012)]. While it has
been reported that Homer 1a mediates synaptic downscaling during sleep, this study
did not examine sleep per se. It instead measured changes in synapses at two vastly
different times of day in a circadian species (mice) in the absence of quantitative
measures of sleep or wakefulness or controls for circadian influences (Diering et al.
2017). Therefore, the results may be equally due to sleep or circadian rhythms.
Other mechanisms proposed in SHY to explain state-dependent synaptic
strengthening and weakening require greater scrutiny. For example, SHY proposes
that the neurotrophin BDNF is a key factor in the synaptic strengthening during
wake that increases sleep drive (Cirelli et al. 2005). This idea is supported by several
lines of evidence. BDNF promotes LTP (Yoshii and Constantine-Paton 2010) which
according to SHY increases sleep drive (Tononi and Cirelli 2014). BDNF mRNA
and protein levels increase with wakefulness (Huber et al. 2007; Hairston et al. 2004;
Taishi et al. 2001), Intraventricular/intracortical administration of exogenous BDNF
increases sleep time (Kushikata et al. 1999, 2003), and NREM SWA (Faraguna et al.
2008). The latter effect can be prevented pharmacologically with drugs that inhibit
protein kinase activity. However, these studies are principally correlative in nature,
involve nonphysiological means of altering NtrkB signaling, or rely on agents that
have broad effects on many kinases—not just those activated by BDNF.
82 M. G. Frank
changes mediated by peripheral clocks in neurons and glia [for review, see Frank
(2021) and Frank and Cantera (2014)].
There is also no convincing direct evidence that downscaling in sleep actually
causes adaptive changes in circuits or behavior (Tononi and Cirelli 2014). The
evidence that does exist is based almost entirely on computational models (Hill
et al. 2008; Olcese et al. 2010; Nere et al. 2013), not real biological findings.
Computational models can inform neurobiology but are not substitutes for direct
experimental observations. They depend critically on what variables are included in
the model and the assumptions one makes about how actual neurons operate in vivo.
Not surprisingly, there are other computational models of memory consolidation
which also posit a role for sleep that do not employ “selective down selection” or
“renormalization” as described in SHY (O’Donnell and Sejnowski 2014; Blanco
et al. 2015). Therefore, direct tests of how down-scaled synapses lead to adaptive
changes (behaviorally or otherwise) are now needed. One promising approach along
these lines is recent work in Drosophila (Donlea et al. 2011). It has been shown in
fruit flies that certain forms of experience can saturate synapse numbers, which
prevent certain forms of learning. Learning can be rescued after a period of sleep,
which also reduces synapses. It will therefore be important to determine if these
findings generalize to other circuits in Drosophila, and to other species.
4.5 Discussion
In the last decade, scientists have made important discoveries about how sleep and
sleep loss impact brain plasticity. There also remain several important challenges.
One important unanswered question is whether sleep-dependent plasticity in the
developing and adult brain is different. It has been suggested, for example, that
synaptic downscaling as described in SHY is equally important during early life
(Tononi and Cirelli 2012). This seems unlikely because the developmental ages
when sleep is maximal coincide with an overall gain of synapses (Frank and Heller
1997; Sur and Leamey 2001; Aghajanian and Bloom 1967; Thurber et al. 2008).
There is also no relationship between the developmental decline in NREM SWA and
a net pruning of cortical synapses, as measured by synaptic markers and spine
morphology in developing mice (de Vivo et al. 2014). Claims that sleep
renormalizes (downscales) synapses during these developmental periods (Cirelli
and Tononi 2020) have been challenged based on confounds in the experimental
design used in these experiments (Frank 2020).
One possibility is that sleep in developing brains promotes synaptic weakening
and strengthening at different times, and is partially determined by the kinds of
waking experience that precedes sleep (Genzel et al. 2014; Ribeiro 2012). In cats,
cortical kinase activation and protein synthesis necessary for LTP only occur in the
first 2–3 h of post-MD sleep (Aton et al. 2009; Seibt et al. 2012). After 5–6 h most of
these changes return to baseline or even drop below baseline values. This suggests
that under these conditions sleep first leads to synaptic potentiation, and then a
84 M. G. Frank
Fig. 4.2 A “Boom and Bust” model of sleep-dependent plasticity explains the effects of sleep on
ocular dominance plasticity (ODP). The initial effects of monocular deprivation (MD) in the cat are
a weakening of responses to the deprived eye during wakefulness. After sleep, there is no further
weakening in deprived eye circuits and instead, responses to the non-deprived eye become stronger.
(a) In the original description of the synaptic homeostasis hypothesis, sleep downscales synaptic
strength in a manner proportionate to the strength at each synapse. This produces no net potentiation
in the non-deprived circuits and increases depression in the deprived eye pathways. (b) According
to the Boom and Bust model, sleep immediately after experience leads to synaptic potentiation
(“Boom”). This is likely Hebbian, but may involve heterosynaptic changes due to synaptic tagging
and capture of plasticity-related proteins in neighboring synapses (Seibt and Frank 2019). As sleep
progresses, global downscaling ensues, which reduces synaptic strength proportionately at each
synapse (“Bust”). The net result is potentiation in the non-deprived eye pathways, and no further
modifications in the deprived eye circuits, which fits empirical data. For illustration purposes,
arbitrary units of synaptic strength are shown. Reproduced with permission from Frank (2015)
general synaptic weakening process (Fig. 4.2). This may also explain findings in
developing mice, where upscaling and downscaling of synaptic strength are highly
dependent on the initial effects of MD (or recovery from MD)—and not necessarily
in accordance with predictions of SHY (Cary and Turrigiano 2021; Hengen et al.
2016; Torrado Pacheco et al. 2021).
A second major challenge to the field is reconciling SHY with findings that show
that sleep in adult brains also increases synaptic strength or number, without an
accompanying “net” downscaling (Frank 2012; Puentes-Mestril and Aton 2017;
Havekes and Abel 2017). One possibility is that “replay-reactivation” occurs against
a background of global downscaling. For example, sleep during the early part of the
rest phase may express high levels of replay (leading to synaptic potentiation) that
then declines. Coincident with replay is a slower, non-Hebbian scaling event that
progressively asserts greater influence as replay fades. As this downscaling affects
all synapses in proportion to their strength, the relative differences in strength are
preserved. This is consistent with the time-course of replay during sleep and
properties of non-Hebbian synaptic scaling as originally described by Turrigiano
(2007). This is also predicted by the “Boom and Bust” model shown in Fig. 4.2 and
4 Sleep and Neuronal Plasticity 85
other theories that posit dual effects of sleep on synaptic strength (Giuditta et al.
1995; Blanco et al. 2015; Genzel et al. 2014; Ribeiro 2012).
A final consideration is that it is critical to conduct direct tests of hypothesized
relationships between synaptic plasticity and sleep function. If sleep need arises from
synaptic potentiation (or other changes in plasticity), then mutations in fruit flies or
mice that reduce synaptic potentiation or plasticity should also reduce sleep need.
There are a number of mutant mouse lines with profound reductions in LTP, but
these mice have not been examined with respect to sleep (Frank 2012). These
mutations can also now be experimentally and reversibly induced, particularly in
fruit flies, with increasingly fine temporal and spatial precision. These techniques
thus do not suffer from limitations of constitutive mutations (i.e., developmental
compensation in embryonic “knock-outs”) and can provide potentially powerful and
direct tests of current theories.
References
Aghajanian GK, Bloom FE. The formation of synaptic junctions in developing rat brain: a
quantitative electron microscopic study. Brain Res. 1967;6(4):716–27.
Arrigoni E, Lu J, Vetrivelan R, Saper CB. Long-term synaptic plasticity is impaired in rats with
lesions of the ventrolateral preoptic nucleus. Eur J Neurosci. 2009;30(11):2112–20.
Aton SJ, Seibt J, Dumoulin M, Jha SK, Steinmetz N, Coleman T, et al. Mechanisms of sleep-
dependent consolidation of cortical plasticity. Neuron. 2009;61(3):454–66.
Aton SJ, Broussard C, Dumoulin M, Seibt J, Watson A, Coleman T, et al. Visual experience and
subsequent sleep induce sequential plastic changes in putative inhibitory and excitatory cortical
neurons. Proc Natl Acad Sci U S A. 2013;110(8):3101–6.
Aton SJ, Suresh A, Broussard C, Frank MG. Sleep promotes cortical response potentiation
following visual experience. Sleep. 2014;37(7):1163–70.
Barnes DC, Wilson DA. Slow-wave sleep-imposed replay modulates both strength and precision of
memory. J Neurosci. 2014;34(15):5134–42.
Battaglia FP, Sutherland GR, McNaughton BL. Hippocampal sharp wave bursts coincide with
neocortical “up-state” transitions. Learn Mem. 2004;11(6):697–704.
Bear MF, Malenka RC. Synaptic plasticity: LTP and LTD. Curr Opin Neurobiol. 1994;4(3):
389–99.
Bendor D, Wilson MA. Biasing the content of hippocampal replay during sleep. Nat Neurosci.
2012;15(10):1439–44.
Benington JH, Frank MG. Cellular and molecular connections between sleep and synaptic plastic-
ity. Prog Neurobiol. 2003;69(2):71–101.
Blanco W, Pereira CM, Cota VR, Souza AC, Renno-Costa C, Santos S, et al. Synaptic homeostasis
and restructuring across the sleep-wake cycle. PLoS Comput Biol. 2015;11(5):e1004241.
Bushey D, Tononi G, Cirelli C. Sleep and synaptic homeostasis: structural evidence in Drosophila.
Science. 2011;332(6037):1576–81.
Buzsaki G. The hippocampo-neocortical dialogue. Cereb Cortex. 1996;6(2):81–92.
Campbell IG, Guinan MJ, Horowitz JM. Sleep deprivation impairs long-term potentiation in rat
hippocampal slices. J Neurophysiol. 2002;88(2):1073–6.
Cary BA, Turrigiano GG. Stability of neocortical synapses across sleep and wake states during the
critical period in rats. elife. 2021;10:e66304.
Chauvette S, Seigneur J, Timofeev I. Sleep oscillations in the thalamocortical system induce long-
term neuronal plasticity. Neuron. 2012;75(6):1105–13.
86 M. G. Frank
Foster DJ, Wilson MA. Reverse replay of behavioural sequences in hippocampal place cells during
the awake state. Nature. 2006;440(7084):680–3.
Frank MG. Hippocampal dreams, cortical wishes: a closer look at neuronal replay and the
hippocampal-neocortical dialogue during sleep. Cell Sci Rev. 2007;3(4):161–71.
Frank MG. Sleep and developmental brain plasticity: not just for kids. Prog Brain Res. 2011;193:
221–32.
Frank MG. Erasing synapses in sleep: is it time to be SHY? Neural Plast. 2012;2012:264378.
Frank MG. Why I’m not SHY: a reply to Tononi and Cirelli. Neural Plast. 2013;2013:394946.
Frank MG. Sleep and synaptic plasticity in the developing and adult brain. Curr Top Behav
Neurosci. 2015;25:123–49.
Frank MG. Sleep and plasticity in the visual cortex: more than meets the eye. Curr Opin Neurobiol.
2017;44:8–12.
Frank MG. The ontogenesis of mammalian sleep: form and function. Curr Sleep Med Rep. 2020;6
(4):267–79.
Frank MG. Renormalizing synapses in sleep: the clock is ticking. Biochem Pharmacol. 2021;191:
114533.
Frank MG, Benington JH. The role of sleep in memory consolidation and brain plasticity: dream or
reality? Neuroscientist. 2006;12(6):477–88.
Frank MG, Cantera R. Sleep, clocks, and synaptic plasticity. Trends Neurosci. 2014;37(9):
491–501.
Frank MG, Heller HC. Development of REM and slow wave sleep in the rat. Am J Phys. 1997;272
(6 Pt 2):R1792–9.
Frank MG, Issa NP, Stryker MP. Sleep enhances plasticity in the developing visual cortex. Neuron.
2001;30(1):275–87.
Genzel L, Kroes MC, Dresler M, Battaglia FP. Light sleep versus slow wave sleep in memory
consolidation: a question of global versus local processes? Trends Neurosci. 2014;37(1):10–9.
Gilestro GF, Tononi G, Cirelli C. Widespread changes in synaptic markers as a function of sleep
and wakefulness in Drosophila. Science. 2009;324(5923):109–12.
Girardeau G, Zugaro M. Hippocampal ripples and memory consolidation. Curr Opin Neurobiol.
2011;21(3):452–9.
Girardeau G, Benchenane K, Wiener SI, Buzsaki G, Zugaro MB. Selective suppression of hippo-
campal ripples impairs spatial memory. Nat Neurosci. 2009;12(10):1222–3.
Giuditta A, Ambrosini MV, Montagnese P, Mandile P, Cotugno M, Grassi Zucconi G, et al. The
sequential hypothesis of the function of sleep. Behav Brain Res. 1995;69(1–2):157–66.
Graves L, Pack A, Abel T. Sleep and memory: a molecular perspective. Trends Neurosci. 2001;24
(4):237–43.
Gulati T, Ramanathan DS, Wong CC, Ganguly K. Reactivation of emergent task-related ensembles
during slow-wave sleep after neuroprosthetic learning. Nat Neurosci. 2014;17(8):1107–13.
Guzman-Marin R, Ying Z, Suntsova N, Methippara M, Bashir T, Szymusiak R, et al. Suppression
of hippocampal plasticity-related gene expression by sleep deprivation in rats. J Physiol.
2006;575(Pt 3):807–19.
Hairston IS, Peyron C, Denning DP, Ruby NF, Flores J, Sapolsky RM, et al. Sleep deprivation
effects on growth factor expression in neonatal rats: a potential role for BDNF in the mediation
of delta power. J Neurophysiol. 2004;91(4):1586–95.
Hasselmo ME. Neuromodulation: acetylcholine and memory consolidation. Trends Cogn Sci.
1999;3(9):351–9.
Havekes R, Abel T. The tired hippocampus: the molecular impact of sleep deprivation on hippo-
campal function. Curr Opin Neurobiol. 2017;44:13–9.
Havekes R, Park AJ, Tudor JC, Luczak VG, Hansen RT, Ferri SL, et al. Sleep deprivation causes
memory deficits by negatively impacting neuronal connectivity in hippocampal area CA1. elife.
2016;5:e13424.
Hengen KB, Torrado Pacheco A, McGregor JN, Van Hooser SD, Turrigiano GG. Neuronal firing
rate homeostasis is inhibited by sleep and promoted by wake. Cell. 2016;165(1):180–91.
88 M. G. Frank
Hennevin E, Huetz C, Edeline JM. Neural representations during sleep: from sensory processing to
memory traces. Neurobiol Learn Mem. 2007;87(3):416–40.
Hill S, Tononi G, Ghilardi MF. Sleep improves the variability of motor performance. Brain Res
Bull. 2008;76(6):605–11.
Hoffman KL, McNaughton BL. Coordinated reactivation of distributed memory traces in primate
cortex. Science. 2002;297:2070–3.
Hubel DH, Wiesel TN. The period of susceptibility to the physiological effects of unilateral eye
closure in kittens. J Physiol. 1970;206(2):419–36.
Huber R, Tononi G, Cirelli C. Exploratory behavior, cortical BDNF expression, and sleep homeo-
stasis. Sleep. 2007;30(2):129–39.
Ishikawa A, Kanayama Y, Matsumura H, Tsuchimochi H, Ishida Y, Nakamura S. Selective rapid
eye movement sleep deprivation impairs the maintenance of long-term potentiation in the rat
hippocampus. Eur J Neurosci. 2006;24(1):243–8.
Ji D, Wilson MA. Coordinated memory replay in the visual cortex and hippocampus during sleep.
Nat Neurosci. 2007;10(1):100–7.
Jouvet-Mounier D, Astic L, Lacote D. Ontogenesis of the states of sleep in rat, cat, and guinea pig
during the first postnatal month. Dev Psychobiol. 1970;2(4):216–39.
Kavanau JL. Memory, sleep, and dynamic stabilization of neural circuitry: evolutionary perspec-
tives. Neurosci Biobehav Rev. 1996;20(2):289–311.
Kim EY, Mahmoud GS, Grover LM. REM sleep deprivation inhibits LTP in vivo in area CA1 of rat
hippocampus. Neurosci Lett. 2005;388(3):163–7.
Kirkwood A, Lee HK, Bear MF. Co-regulation of long-term potentiation and experience-dependent
synaptic plasticity in visual cortex by age and experience. Nature. 1995;375(6529):328–31.
Kopp C, Longordo F, Nicholson JR, Luthi A. Insufficient sleep reversibly alters bidirectional
synaptic plasticity and NMDA receptor function. J Neurosci. 2006;26(48):12456–65.
Krueger JM, Obal F. A neuronal group theory of sleep function. J Sleep Res. 1993;2(2):63–9.
Kudrimoti HS, Barnes CA, McNaughton BL. Reactivation of hippocampal cell assemblies: effects
of behavioral state, experience, and EEG dynamics. J Neurosci. 1999;19(10):4090–101.
Kushikata T, Fang J, Krueger JM. Brain-derived neurotrophic factor enhances spontaneous sleep in
rats and rabbits. AJP. 1999;276(5):R1334–8.
Kushikata T, Kubota T, Fang J, Krueger JM. Neurotrophins 3 and 4 enhance non-rapid eye
movement sleep in rabbits. Neurosci Lett. 2003;346(3):161–4.
Lee AK, Wilson MA. Memory of sequential experience in the hippocampus during slow wave
sleep. Neuron. 2002;36(6):1183–94.
Liu ZW, Faraguna U, Cirelli C, Tononi G, Gao XB. Direct evidence for wake-related increases and
sleep-related decreases in synaptic strength in rodent cortex. J Neurosci. 2010;30(25):8671–5.
Longordo F, Kopp C, Mishina M, Lujan R, Luthi A. NR2A at CA1 synapses is obligatory for the
susceptibility of hippocampal plasticity to sleep loss. J Neurosci. 2009;29(28):9026–41.
Louie K, Wilson MA. Temporally structured replay of awake hippocampal ensemble activity
during rapid eye movement sleep. Neuron. 2001;29(1):145–56.
Malenka RC, Bear MF. LTP and LTD: an embarrassment of riches. Neuron. 2004;44(1):5–21.
Maquet P, Laureys S, Peigneux P, Fuchs S, Petiau C, Phillips C, et al. Experience-dependent
changes in cerebral activation during human REM sleep. Nat Neurosci. 2000;3(8):831–6.
Maret S, Faraguna U, Nelson AB, Cirelli C, Tononi G. Sleep and waking modulate spine turnover
in the adolescent mouse cortex. Nat Neurosci. 2011;14(11):1418–20.
Markram H, Gerstner W, Sjostrom PJ. A history of spike-timing-dependent plasticity. Front
Synaptic Neurosci. 2011;3:4.
Marks CA, Wayner MJ. Effects of sleep disruption on rat dentate granule cell LTP in vivo. Brain
Res Bull. 2005;66(2):114–9.
Matsumoto S, Ohyama K, Diaz J, Yanagisawa M, Greene RW, Vogt KE. Enhanced cortical
responsiveness during natural sleep in freely behaving mice. Sci Rep. 2020;10(1):2278.
4 Sleep and Neuronal Plasticity 89
McDermott CM, LaHoste GJ, Chen C, Musto A, Bazan NG, Magee JC. Sleep deprivation causes
behavioral, synaptic, and membrane excitability alterations in hippocampal neurons. J Neurosci.
2003;23(29):9687–95.
McDermott CM, Hardy MN, Bazan NG, Magee JC. Sleep deprivation-induced alterations in
excitatory synaptic transmission in the CA1 region of the rat hippocampus. J Physiol.
2006;570(Pt 3):553–65.
Mioche L, Singer W. Chronic recordings from single sites of kitten striate cortex during experience-
dependent modifications of receptive-field properties. J Neurophysiol. 1989;62(1):185–97.
Muheim CM, Singletary KG, Frank MG. A chemical-genetic investigation of BDNF-NtrkB
signaling in mammalian sleep. Sleep. 2021;45:zsab237.
Nelson SB, Turrigiano GG. Strength through diversity. Neuron. 2008;60(3):477–82.
Nere A, Hashmi A, Cirelli C, Tononi G. Sleep-dependent synaptic down-selection (I): modeling the
benefits of sleep on memory consolidation and integration. Front Neurol. 2013;4:143.
O’Donnell C, Sejnowski TJ. Selective memory generalization by spatial patterning of protein
synthesis. Neuron. 2014;82(2):398–412.
O’Neill J, Senior T, Csicsvari J. Place-selective firing of CA1 pyramidal cells during sharp wave/
ripple network patterns in exploratory behavior. Neuron. 2006;49(1):143–55.
Olcese U, Esser SK, Tononi G. Sleep and synaptic renormalization: a computational study. J
Neurophysiol. 2010;104(6):3476–93.
Pavlides C, Winson J. Influences of hippocampal place cell firing in the awake state on the activity
of these cells during subsequent sleep. J Neurosci. 1989;9(8):2907–18.
Peigneux P, Laureys S, Fuchs S, Collette F, Perrin F, Reggers J, et al. Are spatial memories
strengthened in the human hippocampus during slow wave sleep? Neuron. 2004;44(3):535–45.
Pennartz CM, Lee E, Verheul J, Lipa P, Barnes CA, McNaughton BL. The ventral striatum in
off-line processing: ensemble reactivation during sleep and modulation by hippocampal ripples.
J Neurosci. 2004;24(29):6446–56.
Peyrache A, Khamassi M, Benchenane K, Wiener SI, Battaglia FP. Replay of rule-learning related
neural patterns in the prefrontal cortex during sleep. Nat Neurosci. 2009;12(7):919–26.
Pfeiffer BE. The content of hippocampal “replay”. Hippocampus. 2020;30(1):6–18.
Poe GR, Nitz DA, McNaughton BL, Barnes CA. Experience-dependent phase-reversal of hippo-
campal neuron firing during REM sleep. Brain Res. 2000;855(1):176–80.
Pozo K, Goda Y. Unraveling mechanisms of homeostatic synaptic plasticity. Neuron. 2010;66(3):
337–51.
Puentes-Mestril C, Aton SJ. Linking network activity to synaptic plasticity during sleep: hypotheses
and recent data. Front Neural Circuits. 2017;11:61.
Qin YL, McNaughton BL, Skaggs WE, Barnes CA. Memory reprocessing in cortiocortical and
hippocampocortical neuronal ensembles. Philos Trans R Soc Lond Ser B Biol Sci. 1997;352
(1360):1525–33.
Ravassard P, Pachoud B, Comte J, Gay N, Touret M, Luppi P, et al. Paradoxical sleep amount
modulates neuronal plasticity in adult rat hippocampus. J Sleep Res. 2006;15:191.
Ravassard P, Pachoud B, Comte JC, Mejia-Perez C, Scote-Blachon C, Gay N, et al. Paradoxical
(REM) sleep deprivation causes a large and rapidly reversible decrease in long-term potentia-
tion, synaptic transmission, glutamate receptor protein levels, and ERK/MAPK activation in the
dorsal hippocampus. Sleep. 2009;32(2):227–40.
Ravassard P, Hamieh AM, Joseph MA, Fraize N, Libourel PA, Lebarillier L, et al. REM sleep-
dependent bidirectional regulation of hippocampal-based emotional memory and LTP. Cereb
Cortex. 2016;26(4):1488–500.
Renouard L, Bridi MCD, Coleman T, Arckens L, Frank MG. Anatomical correlates of rapid eye
movement sleep-dependent plasticity in the developing cortex. Sleep. 2018;41(10):zsy124.
Ribeiro S. Novel experience induces persistent sleep-dependent plasticity in the cortex but not in the
hippocampus. Front Neurosci. 2007;1(1):43–55.
Ribeiro S. Sleep and plasticity. Pflugers Arch. 2012;463(1):111–20.
90 M. G. Frank
Ribeiro S, Gervasoni D, Soares ES, Zhou Y, Lin SC, Pantoja J, et al. Long-Lasting novelty-induced
neuronal reverberation during slow-Wave sleep in multiple forebrain areas. PLoS Biol. 2004;2
(1):E24.
Roffwarg HP, Muzio JN, Dement WC. Ontogenetic development of the human sleep-dream cycle.
Science. 1966;152(3722):604–19.
Romcy-Pereira R, Pavlides C. Distinct modulatory effects of sleep on the maintenance of hippo-
campal and medial prefrontal cortex LTP. Eur J Neurosci. 2004;20(12):3453–62.
Schonauer M, Geisler T, Gais S. Strengthening procedural memories by reactivation in sleep. J
Cogn Neurosci. 2014;26(1):143–53.
Schwindel CD, McNaughton BL. Hippocampal-cortical interactions and the dynamics of memory
trace reactivation. Prog Brain Res. 2011;193:163–77.
Seibt J, Frank MG. Primed to sleep: the dynamics of synaptic plasticity across brain states. Front
Syst Neurosci. 2019;13:2.
Seibt J, Dumoulin MC, Aton SJ, Coleman T, Watson A, Naidoo N, et al. Protein synthesis during
sleep consolidates cortical plasticity in vivo. Curr Biol. 2012;22(8):676–82.
Sengpiel F. Cortical plasticity: learning while you sleep? Curr Biol. 2001;11(16):R647–R50.
Shaffery JP, Roffwarg HP. Rapid eye-movement sleep deprivation does not ‘rescue’ developmen-
tally regulated long-term potentiation in visual cortex of mature rats. Neurosci Lett. 2003;342
(3):196–200.
Shaffery JP, Sinton CM, Bissette G, Roffwarg HP, Marks GA. Rapid eye movement sleep
deprivation modifies expression of long-term potentiation in visual cortex of immature rats.
Neuroscience. 2002;110(3):431–43.
Shaffery JP, Lopez J, Bissette G, Roffwarg HP. Rapid eye movement sleep deprivation revives a
form of developmentally regulated synaptic plasticity in the visual cortex of post-critical period
rats. Neurosci Lett. 2006;391(3):96–101.
Shaffery JP, Lopez J, Roffwarg HP. Brain-derived neurotrophic factor (BDNF) reverses the effects
of rapid eye movement sleep deprivation (REMSD) on developmentally regulated, long-term
potentiation (LTP) in visual cortex slices. Neurosci Lett. 2012;513(1):84–8.
Siapas AG, Wilson MA. Coordinated interactions between hippocampal ripples and cortical
spindles during slow-wave sleep. Neuron. 1998;21(5):1123–8.
Sirota A, Csicsvari J, Buhl D, Buzsaki G. Communication between neocortex and hippocampus
during sleep in rodents. Proc Natl Acad Sci U S A. 2003;100(4):2065–9.
Skaggs WE, McNaughton BL. Replay of neuronal firing sequences in rat hippocampus during sleep
following spatial experience. Science. 1996;271(5257):1870–3.
Smith GB, Heynen AJ, Bear MF. Bidirectional synaptic mechanisms of ocular dominance plasticity
in visual cortex. Philos Trans R Soc Lond Ser B Biol Sci. 2009;364(1515):357–67.
Spolidoro M, Sale A, Berardi N, Maffei L. Plasticity in the adult brain: lessons from the visual
system. Exp Brain Res. 2009;192(3):335–41.
Steriade M, Timofeev I. Neuronal plasticity in thalamocortical networks during sleep and waking
oscillations. Neuron. 2003;37(4):563–76.
Sur M, Leamey CA. Development and plasticity of cortical areas and networks. Nat Rev Neurosci.
2001;2(4):251–62.
Taishi P, Sanchez C, Wang Y, Fang J, Harding JW, Krueger JM. Conditions that affect sleep alter
the expression of molecules associated with synaptic plasticity. Am J Phys. 2001;281:R839–
R45.
Tartar JL, Ward CP, McKenna JT, Thakkar M, Arrigoni E, McCarley RW, et al. Hippocampal
synaptic plasticity and spatial learning are impaired in a rat model of sleep fragmentation. Eur J
Neurosci. 2006;23(10):2739–48.
Tatsuno M, Lipa P, McNaughton BL. Methodological considerations on the use of template
matching to study long-lasting memory trace replay. J Neurosci. 2006;26(42):10727–42.
Thurber A, Jha SK, Coleman T, Frank MG. A preliminary study of sleep ontogenesis in the ferret
(Mustela putorius furo). Behav Brain Res. 2008;189(1):41–51.
4 Sleep and Neuronal Plasticity 91
Timofeev I, Chauvette S. Sleep slow oscillation and plasticity. Curr Opin Neurobiol. 2017;44:116–
26.
Timofeev I, Chauvette S. Sleep, anesthesia, and plasticity. Neuron. 2018;97(6):1200–2.
Tononi G. Slow wave homeostasis and synaptic plasticity. J Clin Sleep Med. 2009;5(2 Suppl):
S16–9.
Tononi G, Cirelli C. Some considerations on sleep and neural plasticity. Arch Ital Biol. 2001;139
(3):122–41.
Tononi G, Cirelli C. Sleep and synaptic homeostasis: a hypothesis. Brain Res Bull. 2003;62(2):
143–50.
Tononi G, Cirelli C. Sleep function and synaptic homeostasis. Sleep Med Rev. 2006;10(1):49–62.
Tononi G, Cirelli C. Time to be SHY? Some comments on sleep and synaptic homeostasis. Neural
Plast. 2012;2012:415250.
Tononi G, Cirelli C. Sleep and the price of plasticity: from synaptic and cellular homeostasis to
memory consolidation and integration. Neuron. 2014;81(1):12–34.
Torrado Pacheco A, Bottorff J, Gao Y, Turrigiano GG. Sleep promotes downward riring rate
homeostasis. Neuron. 2021;109(3):530–44.e6.
Tropea D, Van Wart A, Sur M. Molecular mechanisms of experience-dependent plasticity in visual
cortex. Philos Trans R Soc Lond Ser B Biol Sci. 2009;364(1515):341–55.
Tsanov M, Manahan-Vaughan D. The adult visual cortex expresses dynamic synaptic plasticity that
is driven by the light/dark cycle. J Neurosci. 2007;27(31):8414–21.
Turrigiano GG. Homeostatic plasticity in neuronal networks: the more things change, the more they
stay the same. Trends Neurosci. 1999;22(5):221–7.
Turrigiano G. Homeostatic signaling: the positive side of negative feedback. Curr Opin Neurobiol.
2007;17(3):318–24.
Turrigiano GG. The self-tuning neuron: synaptic scaling of excitatory synapses. Cell. 2008;135(3):
422–35.
Vecsey CG, Baillie GS, Jaganath D, Havekes R, Daniels A, Wimmer M, et al. Sleep deprivation
impairs cAMP signalling in the hippocampus. Nature. 2009;461(7267):1122–5.
Vyazovskiy VV, Cirelli C, Pfister-Genskow M, Faraguna U, Tononi G. Molecular and electro-
physiological evidence for net synaptic potentiation in wake and depression in sleep. Nat
Neurosci. 2008;11(2):200–8.
Wiesel TN, Hubel DH. Single cell responses in striate cortex of kittens deprived of vision in one
eye. J Neurophysiol. 1963;28:1029–40.
Wilson MA, McNaughton BL. Reactivation of hippocampal ensemble memories during sleep.
Science. 1994;265(5172):676–9.
Yang G, Gan WB. Sleep contributes to dendritic spine formation and elimination in the developing
mouse somatosensory cortex. Dev Neurobiol. 2012;72(11):1391–8.
Yang G, Lai CS, Cichon J, Ma L, Li W, Gan WB. Sleep promotes branch-specific formation of
dendritic spines after learning. Science. 2014;344(6188):1173–8.
Yoshii A, Constantine-Paton M. Postsynaptic BDNF-TrkB signaling in synapse maturation, plas-
ticity, and disease. Dev Neurobiol. 2010;70(5):304–22.
Zhou Y, Lai CSW, Bai Y, Li W, Zhao R, Yang G, et al. REM sleep promotes experience-dependent
dendritic spine elimination in the mouse cortex. Nat Commun. 2020;11(1):4819.
Part II
Insufficient Sleep and Its Consequences
Chapter 5
Epidemiology of Insufficient Sleep
Michael A. Grandner
Abstract Insufficient sleep duration has emerged as a key behavioral risk factor for
cardiometabolic disease risk, daytime functioning deficits, and other adverse out-
comes, including mortality. Epidemiologic estimates of insufficient sleep vary,
likely due to variability in how sleep is assessed. Most population-based estimates
are based on single item retrospective reports of habitual sleep duration. Based on
these reports, approximately 30–35% of the adult population reports 6 h of sleep or
less and approximately 8–12% report 9 h or more, with the plurality (approximately
50–60%) reporting the normative 7–8 h. No nationally representative data exist
using objective measures, nor do they exist using prospective self-report (diary), nor
do they exist using validated questionnaires. These estimates, since they rely on
retrospective self-report are likely subject to validity issues, recall biases, and other
methodologic problems. They may better reflect time in bed than physiologic sleep.
Still, these reports have demonstrated utility. Self-reported habitual sleep duration
has been reliably associated in epidemiologic studies with incident mortality, as well
as obesity, heart disease, diabetes, and daytime dysfunction across a range of
domains. Habitual sleep duration also distinguishes sociodemographic groups,
with patterns associated with age (increased sleep duration in younger and older
adults), education level (insufficient sleep associated with less education), income
(insufficient sleep associated with poverty), and race/ethnicity (insufficient sleep
more likely among minority groups).
M. A. Grandner (*)
Sleep and Health Research Program, Department of Psychiatry, University of Arizona College
of Medicine, Tucson, AZ, USA
e-mail: grandner@email.arizona.edu
© Springer Nature B.V. and Shanghai Jiao Tong University Press 2022 95
A. I. Pack, Q. Y. Li (eds.), Sleep and its Disorders, Translational Medicine Research,
https://doi.org/10.1007/978-94-024-2168-2_5
96 M. A. Grandner
Since as early as 1964 (Hammond 1964), there has been disagreement regarding the
definition of “insufficient sleep” as it applies to the general population. Part of the
reason for this confusion is due to the various sources of scientific information on
“insufficient sleep (Grandner et al. 2010).” In general, two main types of studies
have explored this question, including those performed under the controlled condi-
tions of the laboratory (where sleep is systematically manipulated) and those
performed in the field, often using survey-based methods.
The laboratory studies that have explored the physiological impacts of alterations
in sleep opportunity have explored “total sleep deprivation” (complete loss of sleep
for at least 24 h), as well as “partial sleep deprivation” (systematic reduction in sleep
opportunity over days or weeks. These studies sometimes characterize this approach
as “sleep restriction.” Although total sleep deprivation studies are frequently useful
for probing sleep loss in extreme scenarios, it is less useful as a model for real-world
insufficient sleep. Partial sleep deprivation, or sleep restriction, serves as a better
model, because it consists of sleep durations that are frequently seen in real-world
settings (e.g., 4–6 h). Although these experimental protocols can be useful
approaches to revealing the physiologic effects of acute changes in sleep duration,
they often lack external validity. As such, these approaches have improved precision
for observing subtle effects but lack generalizability as a consequence (Grandner
et al. 2010; Grandner 2016; Consensus Conference Panel et al. 2015a). Therefore,
suppositions regarding what is “insufficient” based on laboratory studies may not
generalize to real-world settings where issues such as compensation/countermea-
sures, self-selection, and the presence of confounding variables may cloud predictive
accuracy.
In addition to controlled studies in the laboratory, the other primary source of
research on sleep duration and its impacts comes from population-based methods. In
contrast to the laboratory studies that characterize acute sleep loss relative to a
baseline, these studies are better suited for characterizing habitual sleep behaviors
in context. Regarding sleep duration, this is often classified as “short sleep duration”
or “long sleep duration,” as compared to “normal sleep duration.” The criteria for
classification may vary by the study and population examined. These studies, when
longitudinal, can model chronic sleep loss or changes in sleep duration, as well as
other aspects of sleep health. These may include sleep quality, sleep continuity, and
sleep timing. Sleep duration might be obtained by self-report of a number of hours,
or it might be calculated, based on time in bed, subtracting sleep latency and wake
time after sleep onset. Although most of these studies are survey-based, these
dimensions of sleep may be assessed retrospectively (e.g., through surveys and
questionnaires) or prospectively. Prospective assessment strategies can include
subjective (e.g., sleep diary) or objective (e.g., wearables) methods. These studies
often sacrifice precision for generalizability (Grandner et al. 2010; Grandner 2016;
Consensus Conference Panel et al. 2015a), in contrast to the laboratory studies.
Thus, there are many terms that could refer to “insufficient sleep” from laboratory
protocols of experimental sleep deprivation to population studies of habitual sleep
5 Epidemiology of Insufficient Sleep 97
Perhaps the most current estimates of insufficient sleep come from the Behavioral
Risk Factor Surveillance System (BRFSS). The BRFSS is an annual survey
conducted by the Centers for Disease Control and Prevention (CDC) (http://www.
cdc.gov/brfss). It is state-based, with population-weighted samples representing each
strata of age, sex, race/ethnicity, and geographic region. Sleep duration in the
BRFSS is assessed with the item, “On average, how many hours of sleep do you
get in a 24-hour period?” Responses are coded in whole numbers. Liu and colleagues
reported population-weighted prevalence estimates for sleep duration around a
cutoff of 7 h (based on the consensus statement (Centers for Disease Control and
Prevention 2015) from the 2014 BRFSS (N ¼ 444,306). Overall, the age-adjusted
estimated prevalence of insufficient sleep (6 h) was reported to be 35.1% of the US
population. Grandner and colleagues (2016a) reported prevalence estimates also
using the 2014 BRFSS. Estimated prevalence by hour was calculated, such that
the estimated prevalence by hour of sleep duration was 1.12% for 3 h, 3.19% for
4 h, 7.75% for 5 h, 23.55% for 6 h, 28.72% for 7 h, 27.64% for 8 h, 4.42% for 9 h,
2.35% for 10 h, and 1.27% for 11 h.
Other prevalence estimates have also been calculated using the National Health
and Nutrition Examination Survey (NHANES). The NHANES is a survey that is
also conducted by the CDC that includes a nationally representative sample (http://
www.cdc.gov/nchs/nhanes). The sample size is much smaller than BRFSS, though
the reliability of data may be better since surveys were administered in person rather
than over the phone. Similar to the BRFSS, NHANES assesses sleep duration by a
whole number (no partial hours). Unlike the BRFSS, though, NHANES assesses
sleep duration with the item, “How much sleep do you usually get at night on
weekdays or workdays?” Thus, this item may capture modal nighttime sleep, rather
than 24-h sleep (which may include naps). Using the 2007–2008 wave of NHANES,
Grandner and colleagues calculated prevalence estimates for sleep duration by
1
This section is adapted from Grandner, M. A. (2019). Epidemiology of insufficient sleep and poor
sleep quality. In: M. A. Grandner (Ed.) Sleep and Health. Oxford: Academic Press.
5 Epidemiology of Insufficient Sleep 99
28.72%
days have you felt that you did not get enough rest or sleep?” Based on this variable,
Mcknight-Eily and colleagues (2009) reported prevalence estimates based on
responses to this variable. They estimate that 30.7% of the population reports 0/30
days of insufficient sleep, with 1–13 days reported by 41.3% of the population,
14–29 days reported by 16.8% of the population, and 30/30 days reported by 11.1%
of the population. Based on these estimates, 27.9% of the US population reports
perceived sleep insufficiency at least 2 weeks out of the month.
Table 5.1 reports sleep duration prevalence by age, using population-weighted data
from the 2013 BRFSS. Chi-square analysis indicated that sleep duration differed by
age group ( p < 0.0001). Several studies have examined age-related differences.
Based on BRFSS data, Liu and colleagues (2016) provided age-based prevalence
estimates for insufficient sleep (6 h). They reported estimated of 32.2% for those
age 18–24, 37.9% for 25–34, 38.3% for 35–44, 37.3% for 45–64, and 26.3% for
those 65 years or above. Of note, the lowest rate of insufficient sleep was seen among
the oldest adults. This is consistent with other studies that showed that perceived
insufficient sleep declines with age (Grandner et al. 2015b), as does self-reported
sleep disturbance (Grandner et al. 2012a; Soldatos et al. 2005; Zilli et al. 2009). This
is in contrast to more objective sleep disturbances, which are well-characterized to
increase in older adults (Ohayon et al. 2004; Lindstrom et al. 2012; Cooke and
Ancoli-Israel 2011). There are a number of potential reasons for this, including
retirement offering greater sleep opportunities and differing expectations regarding
sleep (Grandner et al. 2012b).
Prevalence estimates of sleep duration by age in NHANES were reported by
Grandner and colleagues (2015a). Among teenagers aged 16–17 years, prevalence of
sleep duration was 0.63% for 4 h, 19.38% for 5–6 h, 62.47% for 7–8 h, and
17.52% for 9 h. For younger adults aged 18–30 years, prevalence was 4.83% for
4 h, 31.02% for 5–6 h, 54.44% for 7–8 h, and 9.81% for 9 h. For adults aged
30–50 years, prevalence was 5.86% for 4 h, 33.61% for 5–6 h, 55.49% for 7–8 h,
and 5.03% for 9 h. For adults aged 50–65 years, prevalence was 4.95% for 4 h,
35.41% for 5–6 h, 56.04% for 7–8 h, and 3.61% for 9 h. For older adults 65 and
older, prevalence was 4.17% for 4 h, 28.31% for 5–6 h, 55.58% for 7–8 h, and
11.94% for 9 h. Thus, prevalence of insufficient sleep (6 h) was reported to be
20.01% for those aged 16–17 years, 35.85% for those aged 18–30 years, 39.47% for
adults 30–50, 40.36% for adults aged 50–65 years, and 32.48% for older adults over
65 years. Again, the prevalence of insufficient sleep is highest in working age adults.
Using the ATUS data, Basner and colleagues (2014) found that, compared to
15–24 year olds, increased likelihood of insufficient sleep (6 h) was seen in those
aged 25–34 (OR ¼ 1.38; 95% CI ¼ 1.18;1.61), 35–44 (OR ¼ 1.40; 95%
CI ¼ 1.22;1.62), 45–54 (OR ¼ 1.68; 95% CI ¼ 1.44;1.94), and 55–64
Table 5.1 Distribution of sleep duration by age and sex, using the 2013 BRFSS
Stratified by age
Complete 18– 25– 30– 35– 40– 45– 50– 55– 60– 65– 70– 75–
Sample 24 29 34 39 44 49 54 59 64 69 74 79 80
All
Very Short (4 h) (%) 4.31 3.88 4.73 4.62 4.50 5.04 4.92 5.18 4.93 4.17 3.46 2.76 2.64 2.67
5 Epidemiology of Insufficient Sleep
Short (5–6 h) (%) 31.30 29.20 33.76 33.79 34.27 34.35 35.41 34.71 32.72 29.59 26.41 23.65 23.35 22.37
Normal (7–8 h) (%) 56.36 55.42 54.36 54.72 55.28 55.38 54.40 54.31 56.29 58.60 60.33 61.90 61.08 59.10
Long (9 h) (%) 8.03 11.49 7.15 6.87 5.96 5.23 5.28 5.80 6.06 7.64 9.79 11.69 12.93 15.85
Men
Very Short (4 h) (%) 4.46 4.60 5.21 4.88 4.77 4.95 4.77 5.22 4.52 4.24 3.56 2.56 2.31 2.48
Short (5–6 h) (%) 31.68 28.86 34.89 34.61 36.57 34.99 37.26 35.04 32.78 29.51 25.54 21.30 21.16 19.87
Normal (7–8 h) (%) 56.34 55.33 53.84 54.76 53.93 55.14 53.48 54.46 57.16 58.82 60.92 64.05 62.41 60.25
Long (9 h) (%) 7.52 11.22 6.06 5.76 4.72 4.92 4.50 5.28 5.54 7.43 9.98 12.08 14.12 17.39
Women
Very Short (4 h) (%) 4.17 3.12 4.19 4.37 4.23 5.14 5.06 5.14 5.33 4.10 3.37 2.93 2.87 2.80
Short (5–6 h) (%) 30.93 29.57 32.52 33.01 32.04 33.70 33.57 34.39 32.67 29.67 27.20 25.65 24.86 24.01
Normal (7–8 h) (%) 56.38 55.53 54.94 54.69 56.57 55.63 55.32 54.17 55.45 58.38 59.80 60.07 60.17 58.34
Long (9 h) (%) 8.52 11.79 8.35 7.94 7.16 5.53 6.05 6.30 6.55 7.85 9.63 11.36 12.10 14.84
101
102 M. A. Grandner
(OR ¼ 1.41; 95% CI ¼ 1.18;1.68), but not those 65 years or older. Similarly,
shortest sleep durations were seen in working age adults.
Using self-reported insufficiency from the BRFSS, Mcknight-Eily and colleagues
(2009) report that the prevalence of self-reported insufficient sleep at least 14 of the
past 30 days was reported by 31.3% of 18–24 year olds. The estimated prevalence
was 34.2% for 35–34 year olds, 32.1% for 35–44 year olds, 27.2% for 45–64 year
olds, and 15.0% for those 65 years or older.
Several studies have examined sex relative to insufficient sleep. Liu and colleagues
report that based on the 2014 BRFSS data, insufficient sleep (6 h) is reported by
35.4% of men and 34.8% of women (Liu et al. 2016). Using data from 2007 to 2008
NHANES, Whinnery and colleagues report no sex differences in the likelihood of
insufficient sleep (though they report that women are 35% less likely to report long
sleep duration after adjusting for covariates) (Whinnery et al. 2014). Using NHIS
data, Kruger and Friedman report that men are 7% less likely to report 5 vs. 7 h of
sleep (Krueger and Friedman 2009). Basner and colleagues report that men are more
likely to report insufficient sleep (OR ¼ 1.27; 95% CI ¼ 1.20;1.35) (Basner et al.
2014). McKnight-Eily reports that self-reported insufficient sleep at least 14 out of
the past 30 days was reported by 25.5% of men and 30.4% of women (McKnight-
Eily et al. 2009). Taken together, sex differences in insufficient sleep are likely small
and difficult to observe. This is in contrast to self-reported sleep disturbances, which
are much more prevalent in women (Schredl and Reinhard 2011; Subramanian et al.
2011; Zhang and Wing 2006).
See Table 5.1 for population-weighted estimates of habitual sleep duration,
broken down by age and sex, derived from the 2013 BRFSS. A chi-square test
found that the distribution of sleep duration categories differed between men and
women ( p < 0.0001), with slightly more short sleep in men and slightly more long
sleep in women.
100%
5.12% 6.48% 7.91% 8.55% 9.07% 10.14% 11.23% 11.06%
90%
80%
70%
51.51% 50.08% 47.15% 45.34%
63.33% 58.74% 56.36% 54.75%
60%
50%
40%
30%
32.73%
33.69% 33.80%
32.42% 34.13%
20% 32.24% 32.03%
29.37%
10%
7.83% 10.87%
2.18% 2.53% 3.70% 4.28% 5.29% 6.09%
0%
≥ $75,000 $50,000 - $35,000 - $25,000 - $20,000 - $15,000 - $10,000 - <$10,000
$74,999 $49,999 $34,999 $24,999 $19,999 $14,999
Very Short (≤4 hours) Short (5-6 hours) Normal (7-8 hours) Long (≥9 hours)
(OR ¼ 4.3) or 5–6 (OR ¼ 1.6) hours, and those with some college education were
also more likely than college graduates to report <5 (OR ¼ 3.6) or 5–6 (OR ¼ 1.6)
hours of sleep (Whinnery et al. 2014). Another socioeconomic indicator evaluated in
this study was lack of access to healthcare, which was more common among those
reporting <5 h of sleep. Food insecurity—a measure of inability to financially
provide healthy access to enough food—was also more common among those
reporting <5 and 5–6 h of sleep (Whinnery et al. 2014).
Figure 5.2 depicts the distribution of income categories across sleep duration
categories, using BRFSS 2013 data. These values are population weighted. A
chi-square test showed that the distribution of sleep duration categories differed
across income groups ( p < 0.0001).
Fig. 5.3 Geographic distribution of insufficient sleep [based on data reported in Grandner et al.
(2015c)]
abnormally low levels of insufficient sleep were seen in the northern Midwest
(Wisconsin/Minnesota/Iowa), central Texas, central Virginia, and areas along the
West Coast.
Rather than examine statistical hotspots of perceived insufficient sleep,
researchers at the CDC used BRFSS data to map prevalence of 6 h of sleep across
the USA (Liu et al. 2016). The US states with the highest prevalence were (in order)
Hawaii (43.9%), Kentucky (39.7%), Maryland (38.9%), Alabama (38.8%), Georgia
(38.7%), and Michigan (38.7%). The US states with the lowest prevalence were
(in order) South Dakota (28.4%), Colorado (28.5%), Minnesota (29.2%), Nebraska
(30.4%), and Idaho (30.6%).
Many epidemiologic studies have examined the overlapping incidence and preva-
lence of insufficient sleep with a number of other health outcomes.
106 M. A. Grandner
Since 1964 (Hammond 1964), over 50 studies have examined relationships between
insufficient sleep and mortality. These have been summarized in narrative reviews
(Bixler 2009; Bliwise and Young 2007; Grandner and Patel 2009; Youngstedt and
Kripke 2004) and meta-analyses (Gallicchio and Kalesan 2009; Cappuccio et al.
2010). Although the meta-analyses used slightly different methodologies, they
generally come to similar conclusions. Gallicchio and Kalesan (2009) found an
increased risk for mortality associated with short sleep duration (RR ¼ 1.10; 95%
CI [1.06,1.15], p < 0.05), as well as longer sleep duration (RR ¼ 1.23; 95%CI
[1.17,1.30], p < 0.05). Similarly, Cappuccio and colleagues (2010) found significant
increased risk associated with both short (RR ¼ 1.12; 95%CI [1.06,1.18]; p < 0.01)
and long sleep (RR ¼ 1.30; 95%CI [1.22,1.38]; p < 0.0001). The analysis by
Cappuccio conducted post-tests and found that relationships between mortality and
short sleep did not differ by age, sex, socioeconomic status, the definition of sleep
duration category, duration of follow-up, or geographic location.
Many studies have documented associations between insufficient sleep duration and
obesity. These have been summarized in both narrative reviews (St-Onge et al. 2016;
Grandner et al. 2016a, b; St-Onge 2013; Morselli et al. 2012; Akinnusi et al. 2012;
Zimberg et al. 2012; Nielsen et al. 2011; Horne 2011; Chaput 2011; Beccuti and
Pannain 2011) and meta-analyses (Cappuccio et al. 2008; Chen et al. 2008). Overall,
habitual short sleep duration is associated with obesity prevalence, which persists
whether obesity is subjectively or objectively measured. Further, habitual short sleep
is associated with increased weight gain and other measures of adiposity over time in
epidemiologic studies (Nagai et al. 2013; Watanabe et al. 2010; Chaput et al. 2008;
Patel et al. 2006; Ayas et al. 2003). These findings have led to increased epidemi-
ologic study of other cardiometabolic disease outcomes associated with habitual
short sleep (see below), as well as laboratory and other studies of mechanisms
potentially linking sleep duration and obesity (Grandner et al. 2016a, b; Yi et al.
2013; Bornhorst et al. 2012). Although these are outside the scope of an epidemi-
ology review, this line of research is an example of how research across the
translational spectrum can influence work at all other levels. In some cases, more
basic science is influencing epidemiologic study and in others, epidemiologic trends
are inspiring hypotheses at the basic science level.
5 Epidemiology of Insufficient Sleep 107
Laboratory studies have shown that otherwise healthy adults, when sleep deprived in
the laboratory, would demonstrate metabolic dysregulation suggestive of diabetes
risk (Van Cauter et al. 2008; Spiegel et al. 2004). Since then, several studies have
demonstrated that habitual short sleep duration in the population is associated with
diabetes (Grandner et al. 2014, 2016a; Buxton and Marcelli 2010; Perelis et al.
2016). In addition to cross-sectional analyses, meta-analyses suggest that individuals
who habitually get 6 h or less of sleep are approximately 30% more likely to develop
diabetes over time (Anothaisintawee et al. 2016; Shan et al. 2015). Several reviews
have discussed these patterns of findings relative to proposed mechanisms (Grandner
et al. 2016a; Perelis et al. 2016; Anothaisintawee et al. 2016; Lee et al. 2017;
Rangaraj and Knutson 2016; Upala et al. 2015).
of laboratory literature that documents with great precision the effects of acute sleep
loss on neurocognition. Taken together, sleep deprivation is associated with imme-
diate and cumulative deficits in sustained attention (Banks and Dinges 2007; Dinges
2006; Dinges and Banks 2009; Durmer and Dinges 2005; Lim and Dinges 2008;
Rogers et al. 2003; Van Dongen et al. 2005), which can have profound impacts in the
domains of vigilance and safety-sensitive activities such as driving. Other studies
have shown that acute sleep loss impairs decision-making (Jackson et al. 2013;
Killgore et al. 2012; Pace-Schott et al. 2012), planning (Killgore 2010), working
memory (Verweij et al. 2014; Drummond et al. 2012; Joo et al. 2012; Lo et al. 2012;
Jiang et al. 2011), and other cognitive domains. Although these are difficult to study
at a population level, several studies have examined these relationships. For exam-
ple, Maia and colleagues (2013) found that habitual short sleep duration was
associated with drowsy driving, even if those individuals felt fully rested. These
findings are supported by work by Abe and colleagues (2012) who found that short
sleep duration was associated with increased drowsy driving in a large sample of
Japanese adults.
Several issues continue to limit the robustness and reliability of population estimates
of insufficient sleep. First, there are no direct measures of sleep, so all assessment
strategies contain some elements of measurement error. Even polysomnography is
an indirect measure of sleep, so there is no way to measure sleep duration in a
definitive way. For example, sleep diaries are the gold standard for insomnia, which
reflects the individual’s experience. Yet, it suffers from recall and other biases.
Objective methods (e.g., wearables) do not have the same recall biases but may
fail to capture important elements of sleep that correlate with outcomes and also have
their own inherent limitations. Questionnaires may be easy to administer and
efficient for capturing a wide range of outcomes, but their retrospective nature and
imprecision limit interpretability. Retrospective assessments of sleep duration may
better approximate time in bed than physiologic sleep. There still exists no
nationally-representative sample with sampling via prospective, validated measure-
ments. Therefore, all presumptions about population-level estimates should be made
with caution.
Second, the definition of insufficient sleep still varies greatly among different
studies. Some studies examine sleep by clock hour, but most use categorical
groupings. Given the recent consensus statements, some uniformity may be emerg-
ing, but all studies should report how cutoffs were determined and interpretations
should be cautious when comparing results based on different definitions of insuf-
ficient sleep. It is still not clear, for example, whether sleep insufficiency is more
reliably determined based on subjective or objective sleep assessment methods.
5 Epidemiology of Insufficient Sleep 109
Third, even if definitions become more standardized, they are still based on
population-level recommendations which fail to take into account individual differ-
ences in sleep need, sleep ability, and resilience to sleep loss. All of these factors may
contribute to individual differences in what is “insufficient.” Also, general cutoffs
typically fail to resolve insufficiency relative to any specific outcome. For example,
the amount of sleep that is sufficient for an individual to experience optimal
cardiovascular health may be different than for mental health, cognitive function,
immunity, or other outcomes. More information is needed to personalize recom-
mendations of sleep duration, matched with relevant outcomes. It is possible that the
amount of sleep needed may differ depending on the outcome assessed.
References
Abe T, Komada Y, Inoue Y. Short sleep duration, snoring and subjective sleep insufficiency are
independent factors associated with both falling asleep and feeling sleepiness while driving.
Intern Med. 2012;51(23):3253–60.
Adedayo AM, Olafiranye O, Smith D, Hill A, Zizi F, Brown C, et al. Obstructive sleep apnea and
dyslipidemia: evidence and underlying mechanism. Sleep Breath ¼ Schlaf Atmung. 2014;18(1):
13–8.
Akinnusi ME, Saliba R, Porhomayon J, El-Solh AA. Sleep disorders in morbid obesity. Eur J Int
Med. 2012;23(3):219–26.
Altman NG, Izci-Balserak B, Schopfer E, Jackson N, Rattanaumpawan P, Gehrman PR, et al. Sleep
duration versus sleep insufficiency as predictors of cardiometabolic health outcomes. Sleep
Med. 2012;13(10):1261–70.
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. 2016;30:11–24.
Ayas NT, White DP, Al-Delaimy WK, Manson JE, Stampfer MJ, Speizer FE, et al. A prospective
study of self-reported sleep duration and incident diabetes in women. Diabetes Care. 2003;26
(2):380–4.
Banks S, Dinges DF. Behavioral and physiological consequences of sleep restriction. J Clin Sleep
Med. 2007;3(5):519–28.
Basner M, Spaeth AM, Dinges DF. Sociodemographic characteristics and waking activities and
their role in the timing and duration of sleep. Sleep. 2014;37(12):1889–906.
Beccuti G, Pannain S. Sleep and obesity. Curr Opin Clin Nutr Metab Care. 2011;14(4):402–12.
Bixler E. Sleep and society: an epidemiological perspective. Sleep Med. 2009;10(Suppl 1):S3–6.
Bliwise DL, Young TB. The parable of parabola: what the U-shaped curve can and cannot tell us
about sleep. Sleep. 2007;30(12):1614–5.
Bornhorst C, Hense S, Ahrens W, Hebestreit A, Reisch L, Barba G, et al. From sleep duration to
childhood obesit—what are the pathways? Eur J Pediatr. 2012;171(7):1029–38.
Buxton OM, Marcelli E. Short and long sleep are positively associated with obesity, diabetes,
hypertension, and cardiovascular disease among adults in the United States. Soc Sci Med.
2010;71(5):1027–36.
Cappuccio FP, Taggart FM, Kandala NB, Currie A, Peile E, Stranges S, et al. Meta-analysis of short
sleep duration and obesity in children and adults. Sleep. 2008;31(5):619–26.
Cappuccio FP, D’Elia L, Strazzullo P, Miller MA. Sleep duration and all-cause mortality: a
systematic review and meta-analysis of prospective studies. Sleep. 2010;33(5):585–92.
110 M. A. Grandner
Cappuccio FP, Cooper D, D’Elia L, Strazzullo P, Miller MA. Sleep duration predicts cardiovascular
outcomes: a systematic review and meta-analysis of prospective studies. Eur Heart J. 2011;32
(12):1484–92.
Centers for Disease Control and Prevention. Behavioral risk factor surveillance system 2014
codebook report. Atlanta, GA: CDC; 2015.
Chaput JP. Short sleep duration as a cause of obesity: myth or reality? Obesity Rev. 2011;12(5):
e2–3.
Chaput JP, Despres JP, Bouchard C, Tremblay A. The association between sleep duration and
weight gain in adults: a 6-year prospective study from the Quebec Family Study. Sleep. 2008;31
(4):517–23.
Chen X, Beydoun MA, Wang Y. Is sleep duration associated with childhood obesity? A systematic
review and meta-analysis. Obesity. 2008;16(2):265–74.
Consensus Conference Panel, Watson NF, Badr MS, Belenky G, Bliwise DL, Buxton OM, 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. 2015a;11(8):931–52.
Consensus Conference Panel, Watson NF, Badr MS, Belenky G, Bliwise DL, Buxton OM, 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. 2015b;11(6):591–2.
Cooke JR, Ancoli-Israel S. Normal and abnormal sleep in the elderly. In: Montagna P,
Chokroverty S, editors. Sleep disorders. Handbook of clinical neurology. Edinburgh: Elsevier;
2011. p. 653–65.
Dinges DF. The state of sleep deprivation: From functional biology to functional consequences.
Sleep Med Rev. 2006;10(5):303–5.
Dinges DF, Banks S. Sleep deprivation: cognitive performance. In: Amlaner CJ, Fuller PM, editors.
Basics of sleep guide. 2nd ed. Westchester, IL: Sleep Research Society; 2009. p. 257–64.
Drummond SP, Anderson DE, Straus LD, Vogel EK, Perez VB. The effects of two types of sleep
deprivation on visual working memory capacity and filtering efficiency. PLoS One. 2012;7(4):
e35653.
Durmer JS, Dinges DF. Neurocognitive consequences of sleep deprivation. Semin Neurol. 2005;25
(1):117–29.
Eguchi K, Hoshide S, Ishikawa S, Shimada K, Kario K. Short sleep duration is an independent
predictor of stroke events in elderly hypertensive patients. J Am Soc Hypertens. 2010;4(5):
255–62.
Ertel KA, Berkman LF, Buxton OM. Socioeconomic status, occupational characteristics, and sleep
duration in African/Caribbean immigrants and US White health care workers. Sleep. 2011;34
(4):509–18.
Gallicchio L, Kalesan B. Sleep duration and mortality: a systematic review and meta-analysis. J
Sleep Res. 2009;18(2):148–58.
Grandner MA. Sleep deprivation: societal impact and long-term consequences. In: Chokroverty S,
Billiard M, editors. Sleep medicine: a comprehensive guide to its development, clinical mile-
stones, and advances in treatment. New York: Springer; 2016. p. 495–509.
Grandner MA, Patel NP. From sleep duration to mortality: implications of meta-analysis and future
directions. J Sleep Res. 2009;18(2):145–7.
Grandner MA, Patel NP, Gehrman PR, Perlis ML, Pack AI. Problems associated with short sleep:
bridging the gap between laboratory and epidemiological studies. Sleep Med Rev. 2010;14(4):
239–47.
Grandner MA, Martin JL, Patel NP, Jackson NJ, Gehrman PR, Pien G, et al. Age and sleep
disturbances among American men and women: data from the U.S. Behavioral Risk Factor
Surveillance System. Sleep. 2012a;35(3):395–406.
Grandner MA, Patel NP, Gooneratne NS. Difficulties sleeping: a natural part of growing older?
Aging Health. 2012b;8(3):219–21.
5 Epidemiology of Insufficient Sleep 111
Grandner MA, Sands-Lincoln MR, Pak VM, Garland SN. Sleep duration, cardiovascular disease,
and proinflammatory biomarkers. Nat Sci sleep. 2013;5:93–107.
Grandner MA, Chakravorty S, Perlis ML, Oliver L, Gurubhagavatula I. Habitual sleep duration
associated with self-reported and objectively determined cardiometabolic risk factors. Sleep
Med. 2014;15(1):42–50.
Grandner MA, Schopfer EA, Sands-Lincoln M, Jackson N, Malhotra A. Relationship between sleep
duration and body mass index depends on age. Obesity. 2015a;23(12):2491–8.
Grandner MA, Jackson NJ, Izci-Balserak B, Gallagher RA, Murray-Bachmann R, Williams NJ,
et al. Social and behavioral determinants of perceived insufficient sleep. Front Neurol. 2015b;6:
112.
Grandner MA, Smith TE, Jackson N, Jackson T, Burgard S, Branas C. Geographic distribution of
insufficient sleep across the United States: a county-level hotspot analysis. Sleep Health.
2015c;1(3):158–65.
Grandner MA, Seixas A, Shetty S, Shenoy S. Sleep duration and diabetes risk: population trends
and potential mechanisms. Curr Diab Rep. 2016a;16(11):106.
Grandner MA, Alfonso-Miller P, Fernandez-Mendoza J, Shetty S, Shenoy S, Combs D. Sleep:
important considerations for the prevention of cardiovascular disease. Curr Opin Cardiol.
2016b;31(5):551–65.
Hale L, Do DP. Racial differences in self-reports of sleep duration in a population-based study.
Sleep. 2007;30(9):1096–103.
Hammond EC. Some preliminary findings on physical complaints from a prospective study of
1,064,004 men and women. Am J Public Health Nations Health. 1964;54:11–23.
Hirshkowitz M, Whiton K, Albert SM, Alessi C, Bruni O, DonCarlos L, et al. National Sleep
Foundation’s updated sleep duration recommendations: final report. Sleep Health. 2015a;1(4):
233–43.
Hirshkowitz M, Whiton K, Albert SM, Alessi C, Bruni O, DonCarlos L, et al. National Sleep
Foundation’s sleep time duration recommendations: methodology and results summary. Sleep
Health. 2015b;1(1):40–3.
Hoevenaar-Blom MP, Spijkerman AM, Kromhout D, Verschuren WM. Sufficient sleep duration
contributes to lower cardiovascular disease risk in addition to four traditional lifestyle factors:
the MORGEN study. Eur J Prev Cardiol. 2014;21(11):1367–75.
Horne J. Obesity and short sleep: unlikely bedfellows? Obes Rev. 2011;12(5):e84–94.
Irwin MR, Olmstead R, Carroll JE. Sleep disturbance, sleep duration, and inflammation: a system-
atic review and meta-analysis of cohort studies and experimental sleep deprivation. Biol
Psychiatry. 2016;80(1):40–52.
Jackson ML, Gunzelmann G, Whitney P, Hinson JM, Belenky G, Rabat A, et al. Deconstructing
and reconstructing cognitive performance in sleep deprivation. Sleep Med Rev. 2013;17(3):
215–25.
Jean-Louis G, Kripke DF, Ancoli-Israel S, Klauber MR, Sepulveda RS. Sleep duration, illumina-
tion, and activity patterns in a population sample: effects of gender and ethnicity. Biol Psychi-
atry. 2000;47(10):921–7.
Jean-Louis G, Grandner MA, Youngstedt SD, Williams NJ, Zizi F, Sarpong DF, et al. Differential
increase in prevalence estimates of inadequate sleep among black and white Americans. BMC
Public Health. 2015a;15:1185.
Jean-Louis G, Youngstedt S, Grandner M, Williams NJ, Sarpong D, Zizi F, et al. Unequal burden of
sleep-related obesity among black and white Americans. Sleep Health. 2015b;1(3):169–76.
Jiang F, VanDyke RD, Zhang J, Li F, Gozal D, Shen X. Effect of chronic sleep restriction on
sleepiness and working memory in adolescents and young adults. J Clin Exp Neuropsychol.
2011;33(8):892–900.
Joo EY, Yoon CW, Koo DL, Kim D, Hong SB. Adverse effects of 24 hours of sleep deprivation on
cognition and stress hormones. J Clin Neurol. 2012;8(2):146–50.
112 M. A. Grandner
Killgore WDS. Effects of sleep deprivation on cognition. In: Kerkhof GA, Van Dongen HPA,
editors. Human sleep and cognition, part I: basic research. Amsterdam: Elsevier; 2010.
p. 105–29.
Killgore WD, Grugle NL, Balkin TJ. Gambling when sleep deprived: don’t bet on stimulants.
Chronobiol Int. 2012;29(1):43–54.
Krueger PM, Friedman EM. Sleep duration in the United States: a cross-sectional population-based
study. Am J Epidemiol. 2009;169(9):1052–63.
Kurina LM, McClintock MK, Chen JH, Waite LJ, Thisted RA, Lauderdale DS. Sleep duration and
all-cause mortality: a critical review of measurement and associations. Ann Epidemiol. 2013;23
(6):361–70.
Lauderdale DS, Knutson KL, Yan LL, Rathouz PJ, Hulley SB, Sidney S, et al. Objectively
measured sleep characteristics among early-middle-aged adults: the CARDIA study. Am J
Epidemiol. 2006;164(1):5–16.
Lauderdale DS, Knutson KL, Yan LL, Liu K, Rathouz PJ. Self-reported and measured sleep
duration: how similar are they? Epidemiology. 2008;19(6):838–45.
Lee SWH, Ng KY, Chin WK. The impact of sleep amount and sleep quality on glycemic control in
type 2 diabetes: a systematic review and meta-analysis. Sleep Med Rev. 2017;31:91–101.
Lim J, Dinges DF. Sleep deprivation and vigilant attention. Ann N Y Acad Sci. 2008;1129:305–22.
Lindstrom V, Andersson K, Lintrup M, Holst G, Berglund J. Prevalence of sleep problems and pain
among the elderly in Sweden. J Nutr Health Aging. 2012;16(2):180–3.
Liu Y, Wheaton AG, Chapman DP, Cunningham TJ, Lu H, Croft JB. Prevalence of Healthy Sleep
Duration among Adults—United States, 2014. MMWR Morb Mortal Wkly Rep. 2016;65(6):
137–41.
Lo JC, Groeger JA, Santhi N, Arbon EL, Lazar AS, Hasan S, et al. Effects of partial and acute total
sleep deprivation on performance across cognitive domains, individuals and circadian phase.
PLoS One. 2012;7(9):e45987.
Maia Q, Grandner MA, Findley J, Gurubhagavatula I. Short and long sleep duration and risk of
drowsy driving and the role of subjective sleep insufficiency. Accid Anal Prev. 2013;59:618–22.
McKnight-Eily LR, Liu Y, Perry GS, Presley-Cantrell LR, Strine TW, Lu H, et al. Perceived
insufficient rest or sleep among adults—United States, 2008. MMWR Morb Mortal Wkly Rep.
2009;58(42):1175–9.
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. 2013;36(11):985–95.
Morselli LL, Guyon A, Spiegel K. Sleep and metabolic function. Pflugers Arch. 2012;463(1):
139–60.
Mukherjee S, Patel SR, Kales SN, Ayas NT, Strohl KP, Gozal D, et al. An official American
Thoracic Society statement: the importance of healthy sleep. Recommendations and future
priorities. Am J Respir Crit Care Med. 2015;191(12):1450–8.
Nagai M, Tomata Y, Watanabe T, Kakizaki M, Tsuji I. Association between sleep duration, weight
gain, and obesity for long period. Sleep Med. 2013;14(2):206–10.
Nielsen LS, Danielsen KV, Sorensen TI. Short sleep duration as a possible cause of obesity: critical
analysis of the epidemiological evidence. Obes Rev. 2011;12(2):78–92.
Nunes J, Jean-Louis G, Zizi F, Casimir GJ, von Gizycki H, Brown CD, et al. Sleep duration among
black and white Americans: results of the National Health Interview Survey. J Natl Med Assoc.
2008;100(3):317–22.
Ohayon MM, Carskadon MA, Guilleminault C, Vitiello MV. Meta-analysis of quantitative sleep
parameters from childhood to old age in healthy individuals: developing normative sleep values
across the human lifespan. Sleep. 2004;27(7):1255–73.
Pace-Schott EF, Nave G, Morgan A, Spencer RM. Sleep-dependent modulation of affectively
guided decision-making. J Sleep Res. 2012;21(1):30–9.
Patel SR, Malhotra A, White DP, Gottlieb DJ, Hu FB. Association between reduced sleep and
weight gain in women. Am J Epidemiol. 2006;164(10):947–54.
5 Epidemiology of Insufficient Sleep 113
Perelis M, Ramsey KM, Marcheva B, Bass J. Circadian transcription from beta Cell function to
diabetes pathophysiology. J Biol Rhythm. 2016;31(4):323–36.
Rangaraj VR, Knutson KL. Association between sleep deficiency and cardiometabolic disease:
implications for health disparities. Sleep Med. 2016;18:19–35.
Rogers NL, Dorrian J, Dinges DF. Sleep, waking and neurobehavioural performance. Front Biosci.
2003;8:s1056–67.
Sabanayagam C, Shankar A. Sleep duration and hypercholesterolaemia: results from the National
Health Interview Survey 2008. Sleep Med. 2012;13(2):145–50.
Schredl M, Reinhard I. Gender differences in nightmare frequency: a meta-analysis. Sleep Med
Rev. 2011;15(2):115–21.
Shan Z, Ma H, Xie M, Yan P, Guo Y, Bao W, et al. Sleep duration and risk of type 2 diabetes: a
meta-analysis of prospective studies. Diabetes Care. 2015;38(3):529–37.
Soldatos CR, Allaert FA, Ohta T, Dikeos DG. How do individuals sleep around the world? Results
from a single-day survey in ten countries. Sleep Med. 2005;6(1):5–13.
Spiegel K, Tasali E, Penev P, Van Cauter E. Brief communication: sleep curtailment in healthy
young men is associated with decreased leptin levels, elevated ghrelin levels, and increased
hunger and appetite. Ann Intern Med. 2004;141(11):846–50.
Stamatakis KA, Kaplan GA, Roberts RE. Short sleep duration across income, education, and race/
ethnic groups: population prevalence and growing disparities during 34 years of follow-up. Ann
Epidemiol. 2007;17(12):948–55.
St-Onge MP. The role of sleep duration in the regulation of energy balance: effects on energy
intakes and expenditure. J Clin Sleep Med. 2013;9(1):73–80.
St-Onge MP, Grandner MA, Brown D, Conroy MB, Jean-Louis G, Coons M, et al. Sleep duration
and quality: impact on lifestyle behaviors and cardiometabolic health: a scientific statement
from the American Heart Association. Circulation. 2016;134(18):e367–e86.
Subramanian S, Guntupalli B, Murugan T, Bopparaju S, Chanamolu S, Casturi L, et al. Gender and
ethnic differences in prevalence of self-reported insomnia among patients with obstructive sleep
apnea. Sleep Breath ¼ Schlaf Atmung. 2011;15(4):711–5.
Upala S, Sanguankeo A, Congrete S, Romphothong K. Sleep duration and insulin resistance in
individuals without diabetes mellitus: a systematic review and meta-analysis. Diabetes Res Clin
Pract. 2015;109(3):e11–2.
Van Cauter E, Spiegel K, Tasali E, Leproult R. Metabolic consequences of sleep and sleep loss.
Sleep Med. 2008;9(Suppl 1):S23–8.
Van Dongen HP, Vitellaro KM, Dinges DF. Individual differences in adult human sleep and
wakefulness: leitmotif for a research agenda. Sleep. 2005;28(4):479–96.
Verweij IM, Romeijn N, Smit DJ, Piantoni G, Van Someren EJ, van der Werf YD. Sleep
deprivation leads to a loss of functional connectivity in frontal brain regions. BMC Neurosci.
2014;15:88.
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. 2012;7(1):e30972.
Watanabe M, Kikuchi H, Tanaka K, Takahashi M. Association of short sleep duration with weight
gain and obesity at 1-year follow-up: a large-scale prospective study. Sleep. 2010;33(2):161–7.
Watson NF, Badr MS, Belenky G, Bliwise DL, Buxton OM, Buysse D, 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. Sleep. 2015;38(6):843–4.
Whinnery J, Jackson N, Rattanaumpawan P, Grandner MA. Short and long sleep duration associ-
ated with race/ethnicity, sociodemographics, and socioeconomic position. Sleep. 2014;37(3):
601–11.
Yi S, Nakagawa T, Yamamoto S, Mizoue T, Takahashi Y, Noda M, et al. Short sleep duration in
association with CT-scanned abdominal fat areas: the Hitachi Health Study. Int J Obes. 2013;37
(1):129–34.
114 M. A. Grandner
Youngstedt SD, Kripke DF. Long sleep and mortality: rationale for sleep restriction. Sleep Med
Rev. 2004;8(3):159–74.
Zhang B, Wing YK. Sex differences in insomnia: a meta-analysis. Sleep. 2006;29(1):85–93.
Zilli I, Ficca G, Salzarulo P. Factors involved in sleep satisfaction in the elderly. Sleep Med.
2009;10(2):233–9.
Zimberg IZ, Damaso A, Del Re M, Carneiro AM, de Sa SH, de Lira FS, et al. Short sleep duration
and obesity: mechanisms and future perspectives. Cell Biochem Funct. 2012;30(6):524–9.
Chapter 6
Social Factors in Insufficient Sleep
Mathias Basner
Abstract Insufficient sleep (i.e., not getting adequate amounts of daily sleep rela-
tive to individual sleep need on a chronic basis) has not only been linked to
neurobehavioral performance decrements and physiological changes considered
precursors of disease (e.g., decreased insulin sensitivity), but also to negative health
outcomes (e.g., diabetes) and all-cause mortality. Sleep can be regarded as a flexible
commodity that can be traded for waking activities considered more important or
pressing. The high prevalence of habitual short sleep and its association with
morbidity and mortality warrant the identification of social factors and waking
activities that predispose to insufficient sleep and that could be targeted in interven-
tion programs. Several demographic (e.g., age, gender, and race) and social (e.g.,
income, educational attainment, and employment status) factors of insufficient sleep
have been identified and are discussed in detail in this chapter. Waking activities
predominantly exchanged for less sleep (i.e., working, traveling, socializing and
communicating, grooming, and watching TV) are also discussed. The relationships
between social factors and insufficient sleep demonstrate complex interactions with
psychological, behavioral, health, cultural, and environmental factors. Findings
reported in the literature therefore often disagree depending on the study design
and the degree of adjustment for the above-mentioned confounders. More research is
needed that more comprehensively adjusts for social, behavioral, and economic
factors when modeling the relation between sleep and other health or mortality
outcomes.
M. Basner (*)
Unit for Experimental Psychiatry, Division of Sleep and Chronobiology, Department of
Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
e-mail: basner@pennmedicine.upenn.edu
© Springer Nature B.V. and Shanghai Jiao Tong University Press 2022 115
A. I. Pack, Q. Y. Li (eds.), Sleep and its Disorders, Translational Medicine Research,
https://doi.org/10.1007/978-94-024-2168-2_6
116 M. Basner
6.1 Introduction
social factors that predispose to insufficient sleep and that could be targeted in
intervention programs. Below, several demographic and social factors and their
relationship to insufficient sleep will be discussed, as well as the relationship of
several waking activities to insufficient sleep. This chapter focuses on sleep duration,
but it is acknowledged that sleep quality and the timing and variability of sleep also
play important roles in health.
6.2 Age
There are systematic changes in sleep across the lifespan, with a delay in the
circadian phase during adolescence and a gradual shift to earlier timing during
healthy aging (Skeldon et al. 2016). Homeostatic sleep pressure and slow-wave
sleep continuously decline with increasing age. Sleep fragmentation, wake periods,
and the amount of superficial sleep stages S1 and S2 increase, while the amount of
REM sleep decreases with age (Bonnet and Arand 2007). Data from the American
Time Use Survey (ATUS) suggest a U-shaped relationship between age and sleep
duration, with long sleep in very young (15–24 years) and older (>64 years)
respondents, and the lowest amounts of sleep in the 35–64 year age bracket (Basner
et al. 2014). A similar U-shaped relationship was found for long sleep (>9 h relative
to 7 h) in the National Health Interview Survey, a nationally representative survey of
N ¼ 110,441 Americans 18 years and older (Krueger and Friedman 2009). The
striking difference between weekday and weekend sleep time decreases notably with
older age, likely related to retirement (Basner et al. 2007). The finding that weekday
sleep time starts to increase again in 65-year-old respondents is somewhat at odds
with the findings of a meta-analysis of quantitative sleep parameters across the
human lifespan, which reported continuously decreasing total sleep time with
increasing age (Ohayon et al. 2004). However, this meta-analysis concentrated on
polysomnographically measured sleep during the main sleep period, often recorded
in laboratory contexts. The ATUS data suggest that once the obligation to arrive at
work in the morning is no longer present during retirement, older subjects begin to
sleep more, both in the morning and in the afternoon. This strongly supports the
concept of social jetlag during the years of employment, and its reversibility during
retirement (Wittmann et al. 2006).
6.3 Sex
(>11 h). The latter observation agrees with the National Health Interview Survey,
where males had reduced odds of sleeping 5 or fewer hours or 8 or more hours
compared to females (Krueger and Friedman 2009). However, sex differences were
no longer significant after a more comprehensive adjustment for confounding.
6.4 Race/Ethnicity
In the ATUS survey (Basner et al. 2014), Black, Hispanic, and Asian respondents
slept on average significantly longer than white respondents and were also signifi-
cantly more likely to be long sleepers (defined as 11 h per 24 h in this analysis).
Only Black respondents were also more likely to be short sleepers (defined as 6 h
per 24 h in this analysis) than white respondents, while Hispanic and Asian respon-
dents were less likely to be short sleepers, indicating some degree of heterogeneity in
Black respondents. Several studies have shown that Black respondents are more
likely to report short sleep and sometimes also long sleep compared to White
respondents (Stamatakis et al. 2007; Grandner et al. 2014; Maslowsky and Ozer
2014; Jackson et al. 2013; Adenekan et al. 2013; Hale and Do 2007; Knutson et al.
2010). In one prospective study (Stamatakis et al. 2007), the age-adjusted odds ratio
for short sleep in Black respondents was reduced by 32% after adjusting for
household living conditions, income, and education, and only marginally reduced
(12–13%) after adjustment for chronic health conditions, health risk behaviors, and
depression. The ATUS data showed that the high odds for a short sleep in Black
respondents persisted across nearly all other sociodemographic strata. Future studies
are needed to identify in detail what characteristics and behaviors predispose Black
respondents to become short or long sleepers, and to what extent these behaviors are
driven by sociocultural factors and beliefs about the value of sleep. As Knutson
(2013) points out, the broad “racial” or “ethnic” categories typically used in research
are likely heterogeneous, and sleep time estimates within racial or ethnic groups
accordingly reflect this heterogeneity.
6.5 Education
In ATUS (Basner et al. 2014), sleep duration was negatively correlated with
educational attainment, with college graduates or those with a higher degree
obtaining on average significantly less sleep than high school graduates. However,
higher educational attainment was also associated with lower odds of being a short or
long sleeper, suggesting less variability in sleep duration in those with higher
education compared to high school graduates. In contrast, respondents with less
than a high school degree slept significantly longer than those with a high school
degree, and were less likely to be a short sleeper and more likely to be a long sleeper.
In the National Health Interview Survey, high levels of education were also
6 Social Factors in Insufficient Sleep 119
associated with lower odds of short or long sleep (Krueger and Friedman 2009).
These findings are at odds with the finding of Stamatakis et al. (2007) who found a
higher odds of short sleep (<7 h) in those with less than a high school degree. This
association was reduced by 31–33% in separate models adjusting for living condi-
tions, health risk behaviors, and depression.
Stamatakis et al. (2007) found increased odds for short sleep in the lowest income
quintile, but this association was substantially reduced by 69% after adjusting for
living conditions, race/ethnicity and education, reduced by 42% after adjustment for
depression, and reduced by 31% after adjustment for chronic health conditions. After
adjustment for multiple covariates related to health behaviors and health status, a
study by Stranges et al. (2008) found a significant association between lower
socioeconomic position and short sleep in the UK but not in the USA. In the National
Health Interview Survey, high income levels were associated with lower odds of
short or long sleep (Krueger and Friedman 2009). In ATUS (Basner et al. 2014),
family income and sleep duration were negatively correlated, and the odds of being a
short sleeper increased while the odds of being a long sleeper decreased with
increasing family income, the former on weekdays only. During weekdays, respon-
dents in the higher income categories were less likely to sleep in the morning
120 M. Basner
6.8 Employment
In ATUS (Basner et al. 2014), sleep time and the odds of being a short or long
sleeper did not differ between the private-sector and government employees. In
contrast, self-employed respondents obtained significantly more sleep, had signifi-
cantly lower odds of being a short sleeper and also higher odds of being a long
sleeper compared to private-sector employees on weekdays. Interestingly, this
pattern was reversed on weekends/holidays. Relative to private-sector employees,
full-time high school students obtained significantly less sleep (0.28 h) on week-
days, but obtained significantly more sleep (+0.45 h) on weekends/holidays. Full-
time college or university students were more likely to be long sleepers on week-
days, but did otherwise not differ from private-sector employees. Those employed
but absent from work obtained almost 1 additional hour of sleep, and were signif-
icantly less likely to be short sleepers and more likely to be long sleepers on
weekdays. Respondents who were unemployed, retired, or not in the labor force
obtained significantly more sleep, were less likely to be short sleepers, and were
more likely to be long sleepers. In the National Health Interview Survey, those who
worked 41 or more hours were 40% more likely to sleep 5 h or less and 59% less
likely to sleep 9 or more hours relative to those working 1–34 h (Krueger and
Friedman 2009). Those who were not working had increased odds of both short and
long sleep. However, after adjusting for health behaviors and health status,
non-working individuals no longer had increased odds of short sleep.
In ATUS (Basner et al. 2014) working multiple jobs was associated with the
highest observed odds ratio for being a short sleeper (adjusted OR 1.61 on weekdays
and OR 1.72 on weekends/holidays) compared to all other sociodemographic char-
acteristics, and this was true across virtually all sociodemographic strata (approxi-
mately 1 out of 10 employed respondents worked more than one job). Sleep
restriction and high workload associated with working multiple jobs are known
risk factors for increased levels of fatigue that may not only affect job performance
but also affect safety both on the job and during the commute. This is extremely
troubling in subjects working jobs that immediately affect other people’s lives, like
medical doctors [so-called moonlighting (McNeeley et al. 2014)] or bus drivers.
6 Social Factors in Insufficient Sleep 121
Oftentimes, employers do not know about other jobs of their employees, and even if
work hour regulations for the primary job are not violated, they would be if the other
job was taken into account. ATUS data suggest that reducing the number of those
who work multiple jobs could have a profound impact on the amount of sleep
obtained.
The ATUS data (Basner et al. 2014) point to work as the dominant waking activity
that is performed instead of sleep in short sleepers (1.55 h more on weekdays and
1.86 h more on weekends/holidays compared to average sleepers), while long
sleepers spent much less time working compared to average sleepers (2.66 h on
weekdays and 0.90 h on weekends/holidays). Working ranked as the primary
waking activity that was performed instead of sleep across all sociodemographic
strata, with the exception of respondents retired, unemployed, or otherwise not in the
labor force. Furthermore, age 25–64 years, male sex, high income, and employment
per se (i.e., sociodemographic characteristics usually associated with paid work)
were also consistently associated with short sleep. The timing of work in short
sleepers compared to average sleepers suggests that short sleepers stop working
later at night and start working earlier in the morning, which directly impacts their
ability to obtain sufficient amounts of sleep. Between 2003 and 2011 sleep time and
work time were significantly negatively correlated (Basner et al. 2007). The longest
sleep times were observed in the economic crisis years 2009, 2010, and 2011, which
were characterized by layoffs and a decrease in average work time. Prior research
also suggests that respondents working long hours get up earlier in the morning than
those not working or working fewer hours, but that these groups do not differ in
bedtime (Basner and Dinges 2009). For individuals who need to work long hours,
going to bed at an earlier time rather than engaging in other activities (e.g., social-
izing or watching television) would thus be one way to prolong sleep duration
(Basner and Dinges 2009). The association between long work hours and short
sleep has been identified in earlier research (Basner et al. 2007; Biddle and
Hamermesh 1990). Knutson et al. found a significant increase in the odds of short
sleep between 1975 and 2006 for full-time workers only (Knutson et al. 2010).
In terms of intervention strategies, it may be difficult to reduce work time in order
to increase sleep time. However, postponing work start time or making it more
flexible may help increase sleep time; even if the total time spent working is kept
constant. According to ATUS (Basner et al. 2014), with every hour of work or class
starting later in the morning, sleep time increased by approximately 20 min (for class
after 8 am only). Other studies have shown that later class start times may increase
students’ sleep time, attention and cognitive performance (Lufi et al. 2011; Boergers
et al. 2014), and decrease teen automobile crash rates (Vorona et al. 2011), although
one study on college students showed that taking later classes was associated with
increased alcohol consumption and lower academic performance (Onyper et al.
122 M. Basner
2012). More flexible work (and class) start times may be possible without decreasing
work time and productivity, and would also accommodate individuals with late
circadian preferences who cannot fall asleep early but have to get up before their
biological wake time to meet social demands (a condition coined “social jet lag”)
(Wittmann et al. 2006).
Two other waking activities observed more frequently in short sleepers and less
frequently in long sleepers were grooming, socializing, and communicating. These
activities are typically performed late at night or early in the morning and thus
directly “compete” with sleep for a time. In an analysis that concentrated on 2-h
periods pre and post-bed, grooming accounted for 6.5% of the 2 h spent before going
to bed in the evening and for 20.2% of the 2 h after getting out of bed in the morning
(Basner and Dinges 2009). Although a certain level of body hygiene is important for
social and physical well-being, excessive time spent in these activities may reduce
sleep time at both ends of the sleep period.
In ATUS (Basner et al. 2014), the role of watching TV during short and long
sleep was less straightforward, as both short and long sleepers watched more TV
than normal sleepers. However, watching TV in the pre-bed hours was a very
prevalent behavior in short, normal, and long sleepers, also in the 2 h pre-bedtime
(Basner and Dinges 2009). Although short sleepers only watched an average of
4 min more TV than normal sleepers, they started and stopped watching TV much
later at night compared to normal and especially long sleepers. This high prevalence
of watching TV late at night in short sleepers suggests that considerable amounts of
sleep could be gained by turning the TV off earlier at night. Turning off the TV and
other electronic devices may also have the added benefit of reducing exposure to
bright light during late evening hours, which has been shown to suppress melatonin
excretion and delay melatonin onset, and may even delay circadian phase and thus
aggravate social jetlag (Burgess 2013; Gooley et al. 2011). Importantly, the extent to
which watching TV was exchanged for less sleep varied greatly depending on
sociodemographic characteristics. It was more likely in those 45 years, in females,
in Black respondents, in those with lower educational attainment, in respondents
without a partner, in those with family income <$25,000, and in respondents
without work.
6.10 Conclusions
The high prevalence of habitual short sleep and its association with morbidity and
mortality warrant the identification of social factors and waking activities that
predispose to insufficient sleep and that could be targeted in intervention programs.
This chapter illustrates that several demographic and social factors, as well as
waking activities, are related to insufficient sleep. However, the relationships often
demonstrate complex interactions with psychological, behavioral, health, cultural,
and environmental factors. Findings reported in the literature therefore often dis-
agree depending on the study design and the degree of adjustment for the above-
6 Social Factors in Insufficient Sleep 123
Disclosure of Financial Support Supported by NIH NR004281 and by the National Space
Biomedical Research Institute through NASA NCC 9-58.
References
Adenekan B, Pandey A, McKenzie S, Zizi F, Casimir GJ, Jean-Louis G. Sleep in America: role of
racial/ethnic differences. Sleep Med Rev. 2013;17(4):255–62.
Banks S, Dinges DF. Behavioral and physiological consequences of sleep restriction. J Clin Sleep
Med. 2007;3(5):519–28.
Basner M, Dinges DF. Dubious bargain: trading sleep for Leno and Letterman. Sleep. 2009;32(6):
747–52.
Basner M, Fomberstein KM, Razavi FM, Banks S, William JH, Rosa RR, et al. American time use
survey: sleep time and its relationship to waking activities. Sleep. 2007;30(9):1085–95.
Basner M, Spaeth AM, Dinges DF. Sociodemographic characteristics and waking activities and
their role in the timing and duration of sleep. Sleep. 2014;37(12):1889–906.
Beihl DA, Liese AD, Haffner SM. Sleep duration as a risk factor for incident type 2 diabetes in a
multiethnic cohort. Ann Epidemiol. 2009;19(5):351–7.
Biddle JE, Hamermesh DS. Sleep and the allocation of time. J Polit Econ. 1990;98(5):922–43.
Boergers J, Gable CJ, Owens JA. Later school start time is associated with improved sleep and
daytime functioning in adolescents. J Dev Behav Pediatr. 2014;35(1):11–7.
Bonnet MH, Arand DL. EEG arousal norms by age. J Clin Sleep Med. 2007;3(3):271–4.
Burgess HJ. Evening ambient light exposure can reduce circadian phase advances to morning light
independent of sleep deprivation. J Sleep Res. 2013;22(1):83–8.
Buxton OM, Pavlova M, Reid EW, Wang W, Simonson DC, Adler GK. Sleep restriction for 1 week
reduces insulin sensitivity in healthy men. Diabetes. 2010;59(9):2126–33.
Cappuccio FP, D’Elia L, Strazzullo P, Miller MA. Sleep duration and all-cause mortality: a
systematic review and meta-analysis of prospective studies. Sleep. 2010;33(5):585–92.
Centers for Disease Control and Prevention. Effect of short sleep duration on daily activities—
United States, 2005–2008. MMWR Morb Mortal Wkly Rep. 2011;60(8):239–42.
Colten HR, Altevogt BM. In: Colten HR, Altevogt BM, editors. Sleep disorders and sleep
deprivation: an unmet public health problem. Washington, DC: National Academic Press; 2006.
Diekelmann S, Born J. The memory function of sleep. Nat Rev Neurosci. 2010;11(2):114–26.
Ferrie JE, Shipley MJ, Akbaraly TN, Marmot MG, Kivimaki M, Singh-Manoux A. Change in sleep
duration and cognitive function: findings from the Whitehall II Study. Sleep. 2011;34(5):
565–73.
Gallicchio L, Kalesan B. Sleep duration and mortality: a systematic review and meta-analysis. J
Sleep Res. 2009;18(2):148–58.
Gooley JJ, Chamberlain K, Smith KA, Khalsa SB, Rajaratnam SM, Van Reen E, et al. Exposure to
room light before bedtime suppresses melatonin onset and shortens melatonin duration in
humans. J Clin Endocrinol Metab. 2011;96(3):E463–72.
Grandner MA, Chakravorty S, Perlis ML, Oliver L, Gurubhagavatula I. Habitual sleep duration
associated with self-reported and objectively determined cardiometabolic risk factors. Sleep
Med. 2014;15(1):42–50.
124 M. Basner
Vorona RD, Szklo-Coxe M, Wu A, Dubik M, Zhao Y, Ware JC. Dissimilar teen crash rates in two
neighboring southeastern Virginia cities with different high school start times. J Clin Sleep Med.
2011;7(2):145–51.
Wang Q, Xi B, Liu M, Zhang Y, Fu M. Short sleep duration is associated with hypertension risk
among adults: a systematic review and meta-analysis. Hypertens Res. 2012;35(10):1012–8.
Watson NF, Badr MS, Belenky G, Bliwise DL, Buxton OM, Buysse D, 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. Sleep. 2015;38
(8):1161–83.
Wittmann M, Dinich J, Merrow M, Roenneberg T. Social jetlag: misalignment of biological and
social time. Chronobiol Int. 2006;23(1–2):497–509.
Chapter 7
Sleep Loss and the Unfolded Protein
Response
Nirinjini Naidoo
Abstract Sleep loss leads to activation of the unfolded protein response (UPR) not
only in the brain but also in peripheral organs (heart, lung, and pancreas). This has
been shown in multiple species including mice, rats, Drosophila, and migratory
birds. The unfolded protein response to sleep loss changes with age. In older mice,
there is activation of the maladaptive response to sleep loss with increased expres-
sion of genes in the pro-apoptotic pathway. UPR is not simply a consequence of
sleep loss but also affects sleep behavior. Alteration of the molecular machinery of
the UPR alters baseline sleep as well as the degree of sleep recovery following sleep
deprivation. Moreover, administration of drugs that alter the UPR can reduce the
sleep fragmentation that occurs with age. UPR response to sleep loss has implica-
tions for neurodegenerative disorders and other medical conditions.
7.1 Introduction
Sleep loss is common in the American population. Sleep deprivation can result from
a period of acute sleep loss or from insufficient sleep day after day. Some profes-
sional groups, such as health care workers (physicians and nurses) and investment
banking analysts/interns (http://news.efinancialcareers.com/us-en/196881/10-worst-
banks-working-hours/), (ABC News http://abcnews.go.com/Business/bank-amer
ica-investment-banking-analysts-jumping-joy-firms/story; http://www.
businessinsider.com/investment-banking-internship-nightmare-2013-8) are particu-
larly at risk for sleep loss because of their work schedules. Insufficient sleep has been
the focus of two IOM reports; Sleep Disorders and Sleep Deprivation: An Unmet
N. Naidoo (*)
Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University
of Pennsylvania, Philadelphia, PA, USA
e-mail: naidoo@pennmedicine.upenn.edu
© Springer Nature B.V. and Shanghai Jiao Tong University Press 2022 127
A. I. Pack, Q. Y. Li (eds.), Sleep and its Disorders, Translational Medicine Research,
https://doi.org/10.1007/978-94-024-2168-2_7
128 N. Naidoo
Public Health Problem and Optimizing Graduate Medical Trainee (Resident) Hours
and Work Schedule to Improve Patient Safety (Ulmer et al. 2009; Colten et al. 2006).
Sleep loss has a number of consequences. It leads to what has been termed wake-
state instability (Chee et al. 2006), in which individuals are unable to maintain stable
wakefulness and instead drift in and out of sleep. This results in lapses in cognitive
performance and compromises aspects of cognitive function such as executive
attention and working memory (Dinges 1992; Goel et al. 2009). It is estimated
that almost 20% of motor vehicle crashes are attributed to driver sleepiness inde-
pendent of alcohol effects (Connor et al. 2002). In addition to behavioral conse-
quences, sleep loss/deprivation also has important metabolic and cardiovascular
repercussions. Epidemiological studies indicate an association between sleep loss
and increased rates of obesity, type 2 diabetes, and an increased risk of cardiovas-
cular disease (Grandner et al. 2014; Knutson et al. 2010; Knutson 2012; Grandner
2014). People reporting short sleep duration (typically less than 6 h/night) display an
increased prevalence of type 2 diabetes, hypertension, obesity, cardiovascular dis-
ease, and stroke (Qureshi et al. 1997; Elliott et al. 2014; Ayas et al. 2003; Spiegel
et al. 2005; Chen et al. 2008). Several microarrays and transcriptomic—and to a
lesser extent proteomic studies—over the past few years have begun to elucidate
some of the molecular correlates of sleep loss. For example, downregulation of
macromolecular synthetic processes with acute sleep loss has been described
(Mackiewicz et al. 2007). Concomitantly, the upregulation of immediate early
genes and molecules involved in energy regulation and metabolism and an adaptive
cellular stress response to acute sleep loss is consistently observed among all studies
(for details and references see Table 7.1). Specifically, the molecular chaperone BiP
(Immunoglobulin Binding Protein) is upregulated in all studied species in response
to sleep loss (Naidoo 2009). BiP is a sentinel marker of the unfolded protein
response (UPR), which is a cytoprotective cellular stress response. Upregulation of
other components of the UPR signaling pathways (described below) during sleep
loss have also been described by several groups (see Table 7.1). This chapter will
provide an overview of the UPR from its initial cytoprotective response to the later
pro-apoptotic signaling pathway. The chapter will also summarize data from sleep
deprivation and intermittent hypoxia studies that demonstrate a role of the UPR in
sleep. Understanding the pathways activated by sleep loss and the mechanisms by
which they occur will aid in the development of therapies to protect the brain during
sleep loss and to treat sleep disorders, including those associated with aging.
The unfolded protein response (UPR) is a coordinated set of signaling pathways that
are triggered when endoplasmic reticulum (ER) homeostasis is perturbed. The ER is
an organelle within which all secretory and integral membrane proteins are folded
and post-translationally modified in ATP-dependent chaperone-mediated processes
(Walter and Ron 2011). The ER is also the site of steroid, cholesterol, and lipid
7 Sleep Loss and the Unfolded Protein Response 129
Table 7.1 UPR associated genes that change with sleep loss and sleep fragmentation
Gene name Function References
BiP/HSPA5/ ER chaperone, folding, ERAD, ATPase Mackiewicz et al. (2007), Naidoo
GRP78 activity, anti-apoptotic et al. (2005, 2008), Maret et al.
(2007), Terao et al. (2006), Jones
et al. (2008), Cirelli et al. (2006), and
Shaw et al. (2000b)
GRP94 Heat shock 90 family, ER chaperone, Maret et al. (2007), Cirelli and
folding Tononi (2000), and Terao et al. (2003)
CHOP/ Apoptotic signaling Cirelli and Tononi (2000)
GADD153
ERP72 ER chaperone, folding Terao et al. (2006) and Cirelli and
Tononi (2000)
PERK Kinase, protein translation inhibition, Cirelli et al. (2004)
antioxidant response
Calcineurin Calcium activated phosphatase Cirelli et al. (2004)
FK506 bind- Peptidyl propyl cis/trans isomerases, Cirelli et al. (2004)
ing protein12 conformational change, translational
control
IP3 receptor Calcium receptor signaling Cirelli et al. (2004)
XBP-1 Transcription factor, ER chaperone Mackiewicz et al. (2007) and Maret
induction et al. (2007)
Calreticulin Calcium binding, ER chaperone of Mackiewicz et al. (2007) Maret et al.
glycoproteins (2007), and Cirelli et al. (2006)
eIF4B Translation initiation factor Mackiewicz et al. (2007)
Ribosomal Protein translation Mackiewicz et al. (2007)
S6 kinase
Dnajc1 DnaJ (Hsp40) homolog, ER chaper- Mackiewicz et al. (2007)
one)—Protein synthesis and folding—
ERAD, translocation, HSP70 assis-
tance, ATPase activity
Dnajb5 HSP40; binds unfolded ER proteins Mackiewicz et al. (2007)
Dnajc3 HSP40 family Mackiewicz et al. (2007)
Dnajb11 HSP40 family Mackiewicz et al. (2007)
Gadd45a (Demethylation—DNA repair) Mackiewicz et al. (2007)
Gadd45b Mackiewicz et al. (2007)
Calpain 5 ER protease, caspase cleavage Mackiewicz et al. (2007)
Mannosidase ER degradation enhancing—EDEM/ Mackiewicz et al. (2007)
2, alpha B1 ERAD
Hsp90ab1 Signal transduction, protein folding and Maret et al. (2007)
degradation, and morphological
evolution
Hsp60 Heat shock protein family Cirelli and Tononi (2000) and Cirelli
(2002)
Hsp70 Heat shock protein family Cirelli and Tononi (2000) and Cirelli
(2002)
Hspb1 Stress resistance and actin organization Maret et al. (2007) and Cirelli et al.
(2006), and Conti et al. (2007)
(continued)
130 N. Naidoo
biosynthesis and is the major signal-transducing organelle in the cell that continu-
ously responds to environmental cues to release calcium (Kaufman 2002). Since the
endoplasmic reticulum is a complex membranous network that extends throughout
the cytoplasm and is contiguous with the nuclear envelope, it can sense and transmit
signals that originate in any cellular sub-compartment. Thus, perturbing ER homeo-
stasis disrupts protein folding and leads to the accumulation of unfolded proteins and
protein aggregates, which are detrimental to cell survival. The UPR conveys signals
from the ER to the nucleus and cytosol through the activation of three signal
transducers; PERK (PKR like ER kinase), ATF6 (Activating Transcription Factor
6), and IRE1 (Inositol Requiring Element 1) (see Fig. 7.1). These three molecules are
7 Sleep Loss and the Unfolded Protein Response 131
Fig. 7.1 Schematic showing the three major pathways of the unfolded protein response. Misfolded
or unfolded proteins titrate BiP away from the three transducers of ER stress: PERK, IRE1, and
ATF6. Activated PERK phosphorylates eIF2α to attenuate protein translation and phosphorylates
Nrf-2 to upregulate the antioxidant response. Cleaved activated ATF6 leads to induction of
molecular chaperones like BiP and GRP94. IRE1 activation leads to XBP-1 splicing, transcriptional
activation of chaperones, and stimulation of protein and transcript degradation through ERAD and
RIDD, respectively. The various ER chaperones, such as BiP and GRP94 are protective and control
protein folding and components of the UPR, while ATF4 induction leads to GADD34 production as
well as autophagy
held in an inactive state by binding to a molecular chaperone, BiP on the luminal side
of the ER. BiP, also known as GRP78 and Hspa5, is an ATPase and member of the
heat shock 70 family of proteins that binds preferentially to nascent and misfolded
proteins. Perturbation of ER homeostasis caused by events such as reduced energy,
changes in calcium flux, redox changes, ischemia, hyperhomocysteinemia, viral
infections, and mutations (Kaufman 2002; Ron 2002) leads to protein misfolding.
When this occurs BiP dissociates from PERK, IRE1 and ATF6 bind to the misfolded
proteins and assist in their refolding or to escort these proteins out of the ER for
degradation. The dissociation of BiP from these three molecules (PERK, IRE1, and
ATF6) leads to the activation of their three respective signaling cascades that
transduce the ER stress signal to the cytosol and nucleus. The duration and response
of each signaling cascade are dependent on the duration of the ER stress signal. The
response is initially adaptive, however, sustained ER stress eventually leads to a
132 N. Naidoo
ATF6 represents a group of ER stress transducers that encode basic leucine zipper
(bZIP) transcription factors, including ATF6α, ATF6β, LUMAN (also known
as CREB3), old astrocyte specifically induced substance (OASIS; also known as
CREB3L1), BBF2 human homologue on chromosome 7 (BBF2H7; also known as
CREB3L2), cyclic AMP-responsive element-binding protein hepatocyte (CREBH;
also known as CREB3L3) and CREB4 (also known as CREB3L4) (Asada et al.
2011). Under ER stress conditions ATF6 dissociates from BiP and is exported to the
Golgi where it is cleaved by site-1 protease (S1P) and S2P releasing its cytosolic
domain which is a potent transcription factor. The 50-kDa cleaved ATF6α then
translocates to the cell nucleus, where it binds to the ER stress response element
CCAAT(N)9CCACG (Yoshida et al. 1998) in genes encoding ER chaperone pro-
teins such as BiP and GRP94. GRP94 is a member of the heat shock90 family of
chaperones. This binding results in increased transcription of these proteins and
hence increased protein folding activity in the ER (Yoshida et al. 1998; Okada et al.
2002). Other important targets regulated by ATF6 include XBP-1, CHOP, HERP
(hyperhomocysteinemia-induced ER stress-responsive protein), and PDI (Protein
disulfide isomerase).
134 N. Naidoo
Fig. 7.2 Sustained ER stress leads to pro-apoptotic signaling. Prolonged UPR activation leads to
ER calcium release and cell death signaling. Activated IRE-1 complexes with TRAF2 and ASK1 to
activate JNK and caspases. ATF4-dependent transcription leads to increases in CHOP. CHOP is a
pro-apoptotic transcription factor. CHOP inhibits BCl-2 leading to calcium release; higher calcium
levels sensitize mitochondria to other insults inducing cell death. BCl-2 exerts an anti-apoptotic
function in the ER.CHOP and JNK also promote the translocation of Bax to the mitochondria where
it facilitates the release of cytochrome c required for caspase activation and apoptosis
7 Sleep Loss and the Unfolded Protein Response 135
The induction of the molecular chaperone BiP by sleep loss is conserved across
species. Several studies have shown that acute sleep deprivation increases
BiP/GRP78 expression in the brains of mice (Mackiewicz et al. 2007; Naidoo
et al. 2005; Maret et al. 2007), rats (Cirelli et al. 2004; Terao et al. 2006), birds
(Jones et al. 2008), and fruitflies (Shaw et al. 2000a; Naidoo et al. 2007; Williams
et al. 2007). Sleep loss activates the PERK pathway in the Drosophila brain (Brown
et al. 2014) and also in the cerebral cortex of mice (Naidoo et al. 2005, 2008). PERK
cerebellar transcript levels were also found to be higher in wakefulness than in sleep
(Cirelli et al. 2004). Activation of IRE1 activity through xbp1 mRNA splicing has
been observed in sleep-deprived fly brains (Brown et al. 2014); Naidoo et al.
unpublished observations). Xbp 1 transcript levels have also been reported to
increase in mouse brain (Mackiewicz et al. 2007). Transcripts of several molecular
chaperones besides BiP are also increased in brain following sleep loss. ERP72,
GRP94, HSP27, HSP70-1, and HSP84 mRNA levels are all increased in cortex,
basal forebrain, hypothalamus cerebellum, and medulla during sleep deprivation,
whereas increased mRNA levels during recovery sleep were limited to the cortex and
medulla (Terao et al. 2006). These chaperones are downstream targets of UPR
136 N. Naidoo
transcription factors XBP1 and ATF6. Other UPR-specific transcripts that change
with sleep deprivation include DNA-J which is a co-chaperone of BiP, calreticulin,
caspase-9, ATF4, ATF6 (Mackiewicz et al. 2007), and ERo1L (Williams et al.
2007). Several UPR-activated pro-inflammatory molecules are also induced by
sleep deprivation. These include NfkB and JNK (Williams et al. 2007). For a
complete list of UPR factors upregulated by sleep loss, see Table 7.1.
Aged animals exhibit more fragmented sleep (Naidoo et al. 2008; Welsh et al. 1986;
Shiromani et al. 2000) and display basal levels of ER stress in tissues examined
(Brown et al. 2014; Naidoo et al. 2011). The adaptive ER stress response to sleep
deprivation is impaired in aged mice cerebral cortices (Naidoo et al. 2008) as well as
in aged Drosophila brains (Brown et al. 2014). There is a decrease in the adaptive
UPR and an increase in inflammatory/pro-apoptotic signaling (Brown et al. 2014).
Wake-active neurons in aged mice exhibit considerable ER stress and PERK path-
way activation when compared with similar regions in young mice (Naidoo et al.
2011). In a study comparing wake in young and old mice, we found wake instability
and impaired wake responses to novelty in older mice that were accompanied by ER
stress in orexinergic and noradrenergic wake neurons (Naidoo et al. 2011). The ER
stress response in orexin and noradrenergic neurons was evidenced by the presence
of activated phosphorylated PERK, nuclear translocation of CHOP, and increased
GADD34 expression. The specific wake impairments identified in aged mice were
consistent with orexinergic and noradrenergic neuronal injury. Surprisingly, recov-
ery sleep following sleep deprivation is less in older animals than in young. This has
been shown in humans (Bonnet 1985; Carskadon and Dement 1987) and in rats
(Shiromani et al. 2000; Mendelson and Bergmann 2000). It is not known whether the
UPR plays a role in the mammalian recovery sleep response to sleep deprivation and
is currently an area of investigation.
While acute sleep deprivation leads to induction of the adaptive UPR, it appears that
chronic sleep loss or long-term sleep deprivation as assessed by an increase in BiP
transcript levels does not. A study by Cirelli et al. (2006) showed that rat cerebral
cortex BiP mRNA levels do not increase after long-term (7 days) sleep deprivation
as much as after short-term (8 h) sleep deprivation. It is possible that 7 days of sleep
deprivation results in sustained ER stress much like that experienced by aged
animals, which results in a shutdown of the adaptive ER stress response. Thus, it
is likely to be more injurious. Further studies are needed to address this. A more
recent study indicates that long-term REM sleep loss (6–10 days) does lead to
7 Sleep Loss and the Unfolded Protein Response 137
increased apoptosis in several regions of the rat brain (Biswas et al. 2006). Using
amino cupric staining as a marker of neuronal degeneration, TUNEL (TdT-mediated
dUPT nick end labeling) assays, and the ratio of Bcl-2 to Bax as indices of apoptosis
this study demonstrates that neurons in locus coeruleus (LC), laterodorsal tegmen-
tum (LDT), and medial preoptic area (MPO) but not in the lateral septum undergo
degeneration with REM sleep loss (Biswas et al. 2006). The increase in Bax positive
neurons over BCl-2 positive neurons observed in this study suggests that the
apoptotic phase of the UPR is activated in the REM sleep-deprived animals. The
absence of any apoptotic factors in the lateral septum following REM sleep loss
illustrates that there is differential vulnerability between neuronal groups to stress.
Most studies have focused on UPR induction in the brain as a result of sleep loss.
There are a few studies that indicate that sleep loss does activate the UPR in
peripheral tissues. BiP is upregulated in several peripheral tissues with acute sleep
deprivation; these include liver (Maret et al. 2007), skeletal muscle (Cirelli et al.
2006), heart (Anafi et al. 2013), lung (Anafi et al. 2013), and pancreas (Naidoo et al.
2014). A transcriptomics study on heart and lung examined the temporal profile of
gene expression during 3, 6, 9, and 12 h of sleep deprivation (Anafi et al. 2013).
They found several ER stress and UPR transcripts that were repressed during sleep
and enhanced during sleep loss. Notably, protein folding and the UPR was the major
pathway identified in this study. In addition to BiP, other transcripts identified
included XBP1, ATF4, GADD34, GRP94, DNAjb, ER Mannosidase I, Calreticulin,
HSP40, IRE1, ATF6, and UGGT (Anafi et al. 2013) (Table 7.1). A later study in the
pancreas demonstrated that BiP protein expression was significantly increased with
acute sleep deprivation (Naidoo et al. 2014). Induction of the UPR was confirmed
using additional markers, including cleavage of ATF6 and phosphorylation of eIF2a,
both of which were significantly increased following sleep loss. Expression of
CHOP also trended higher with sleep loss, though CHOP expression exhibited a
high degree of variation between animals. This study also demonstrated that there is
a loss of the adaptive UPR with age and that this affects insulin sensitivity in aged
animals providing a link between age-related sleep disturbances and metabolic
dysfunction (Naidoo et al. 2014).
Sleep fragmentation that occurs with aging, disease, and sleep apnea also induces the
UPR. Exposure to intermittent cyclical hypoxia/reoxygenation similar to that which
occurs in obstructive sleep apnea results in ER stress in select brainstem motor
neurons (Zhu et al. 2008). Motor neurons process large amounts of secretory and
138 N. Naidoo
membrane proteins that must be properly folded within the ER (Shaw et al. 2000b),
and as such are prone to ongoing ER stress and UPR activation. Both short-term
(3 days) and longer-term intermittent hypoxia over 8 weeks selectively activates the
PERK pathway of the UPR in the hypoglossal and facial motor nuclei (Zhu et al.
2008). Additionally, pro-apoptotic proteins, CHOP, GADD34, cleaved caspase-7,
and caspase-3, are all increased with longer-term intermittent hypoxia in these motor
neurons. As a result, these motor neurons undergo ER stress even at baseline which
is then exacerbated by long-term intermittent hypoxia. The presence of CHOP and
subsequently GADD34 leads to dephosphorylation of p-eIF2α, increased protein
synthesis, and greater ER stress in these neurons. The inability of these neurons to
relieve the ER stress leads to a neural injury that can be observed at the ultrastructural
level; the ER is swollen and distorted and there is disaggregation of ribosomes and
degranulation of rough ER (Zhu et al. 2008).
A recent study from the Gozal group showed that chronic sleep fragmentation
also induced temporal changes in ER stress in the hypothalamus, across the three
major UPR pathways (Hakim et al. 2015). There is an increase in cleaved ATF6
expression, splicing of XBP1 mRNA (indicative of IRE1 activation), and induction
of the PERK pathway evidenced by increased p-eIF2α expression. The UPR chap-
erones BiP, HSP70, and HSP90 also exhibit increased expression levels in hypo-
thalamic extracts after sleep fragmentation compared with sleep control conditions.
This study also found that chronic sleep fragmentation leads to sustained ER stress
and induction of leptin resistance (Hakim et al. 2015) identifying a possible mech-
anism by which sleep fragmentation impacts metabolism.
Whether or not the effect of sleep loss on UPR induction in the periphery is a
direct effect or occurs via the brain is not known and remains to be determined.
Not only is ER stress and the UPR activated with sleep loss, but this pathway also
appears to be involved in the sleep homeostatic response. In Drosophila, there is an
increase in BiP with sleep loss and a diminution of expression with recovery sleep
(Naidoo et al. 2007). BiP protein levels return to baseline levels over 24 h with
recovery sleep following an almost threefold increase with 6 h of sleep deprivation.
Overexpression of BiP through genetic means leads to an increase in the amount of
sleep recovered after sleep loss (Naidoo et al. 2007). Whether the altered amounts of
recovery sleep when BiP levels are manipulated due to BiP itself or more indirectly
through other effects on the UPR remains to be determined. Recent data indicates
that reducing ER stress in aged animals improves sleep quality. While inducing ER
stress fragments sleep (Brown et al. 2014), treatment with 4-phenyl butyrate
(PBA)—a chemical chaperone with properties similar to BiP—consolidates sleep
in aging Drosophila (Brown et al. 2014). Aged flies, like aged mice, display reduced
expression of BiP. Thus, supplementing chaperone levels ameliorates fragmented
sleep that is typically observed during aging. PBA treatment also improves the
7 Sleep Loss and the Unfolded Protein Response 139
recovery of sleep response to sleep deprivation in aged flies recapitulating the effect
of BiP overexpression in young flies (Brown et al. 2014). The effect of the chemical
chaperone on sleep is not limited to its effect in aged flies. PBA improves sleep
quality in the short sleeping mutant, sleepless. Sleep in this extreme short sleeping
mutant is also very fragmented (Koh et al. 2006); treatment with PBA both consol-
idated and increased sleep in the sleepless mutant (Brown et al. 2014). In all of the
studies described, the chemical chaperone acted analogously to BiP to reduce
activation of the PERK and IRE1 pathways. Recent data indicates that the PERK
pathway itself has a role in sleep regulation. Pharmacological knockdown of PERK
has been shown to reduce sleep in zebrafish and Drosophila indicating an evolu-
tionarily conserved mechanism (Ly et al. 2020). This effect on sleep is recapitulated
by genetic pan-neuronal knockdown of PERK and more specifically in wake-
promoting PDF (pigment dispersing factor) neurons. PERK overexpression within
the same PDF circuit increases sleep during the night (Ly et al. 2020). Further,
genetic manipulation of PERK within the PDF neurons alters PDF expression
suggesting that PERK signaling directly impacts wake-promoting neuropeptide
expression, revealing a mechanism through which proteostasis pathways can affect
sleep and wake behavior. The impact of the direct manipulation of the IRE1 pathway
upon sleep is currently being investigated.
The UPR is an integral part of the cell’s protein homeostatic machinery and is critical
for cellular integrity. Protein folding, processing, and secretion are central to cell
function. The ER is exquisitely sensitive to changes in the cell’s milieu. The
upregulation of the UPR by sleep disturbances indicates that losing sleep causes a
perturbation in the cellular environment of the various tissues of affected organisms
and that sleep loss is a cellular stressor. The duration of cellular stress/sleep loss also
determines the type of response. Prolonged or chronic sleep loss and fragmentation
lead to inflammatory and pro-apoptotic signaling which are detrimental to cell
survival.
Sleep loss induces the unfolded protein response in the brain as well as peripheral
organs and the implications of UPR induction by sleep loss are multifold. First, ER
stress and the UPR are known to be the mechanism underlying B-cell death in
diabetes (Hotamisligil 2005). ER stress and the UPR could be the link between
metabolic dysfunction and sleep deprivation (see model in Fig. 7.3). Many epide-
miological studies have indicated that sleep deprivation leads to insulin resistance.
Our study in aged mice indicates that chronic sleep loss reduces insulin sensitivity
(Naidoo et al. 2014). ER stress and induction of the UPR have also been implicated
in abnormal protein processing and neuronal death in age-associated diseases, as
well as neurodegenerative diseases [see reviews Forman et al. (2003), Johnson et al.
(2008), Rumpf and Pazos (2013), Hetz and Mollereau (2014), and Scheper and
Hoozemans (2015)]. Accumulation of misfolded proteins that lead to alterations in
140 N. Naidoo
Fig. 7.3 Model of interaction between sleep loss and protein misfolding. Sleep loss/disruption
leads to an adaptive robust UPR response in young animals while in aged animals the UPR is
impaired. This leads to protein misfolding persisting, activation of pro-inflammatory and
pro-apoptotic signaling, and increased oxidative stress that promotes more ER stress and protein
misfolding creating a vicious cycle
Acknowledgments Thanks to Sarah Ly for helpful comments and edits during the writing of the
manuscript and Michael Paolini for assistance with figures. This study was supported by
NIH/AG17628.
142 N. Naidoo
References
Ameri K, Harris AL. Activating transcription factor 4. Int J Biochem Cell Biol. 2008;40(1):14–21.
Anafi RC, Pellegrino R, Shockley KR, Romer M, Tufik S, Pack AI. Sleep is not just for the brain:
transcriptional responses to sleep in peripheral tissues. BMC Genomics. 2013;14:362.
Asada R, Kanemoto S, Kondo S, Saito A, Imaizumi K. J Biochem. 2011;149(5):507–518. https://
doi.org/10.1093/jb/mvr041
Ayas NT, White DP, Manson JE, Stampfer MJ, Speizer FE, Malhotra A, et al. A prospective study
of sleep duration and coronary heart disease in women. Arch Intern Med. 2003;163(2):205–9.
Basseri S, Austin RC. Endoplasmic reticulum stress and lipid metabolism: mechanisms and
therapeutic potential. Biochem Res Int. 2012;2012:841362.
Behrman S, Acosta-Alvear D, Walter P. A CHOP-regulated microRNA controls rhodopsin expres-
sion. J Cell Biol. 2011;192(6):919–27.
Biswas S, Mishra P, Mallick BN. Increased apoptosis in rat brain after rapid eye movement sleep
loss. Neuroscience. 2006;142(2):315–31.
Bonnet MH. Effect of sleep disruption on sleep, performance, and mood. Sleep. 1985;8(1):11–9.
Brown M, Chan M, Zimmerman J, Pack A, Jackson N, Naidoo N. ER stress alters sleep and sleep
homeostasis during aging. Neurobiol Aging. 2014;35(6):1431–41.
Calfon M, Zeng H, Urano F, Till JH, Hubbard SR, Harding HP, et al. IRE1 couples endoplasmic
reticulum load to secretory capacity by processing the XBP-1 mRNA. [erratum appears in
Nature 2002 Nov 14;420(6912):202]. Nature. 2002;415(6867):92–6.
Carskadon MA, Dement WC. Daytime sleepiness: quantification of a behavioral state. Neurosci
Biobehav Rev. 1987;11(3):307–17.
Chan K, Han XD, Kan YW. An important function of Nrf2 in combating oxidative stress:
detoxification of acetaminophen. Proc Natl Acad Sci U S A. 2001;98(8):4611–6.
Chee MW, Chuah LY, Venkatraman V, Chan WY, Philip P, Dinges DF. Functional imaging of
working memory following normal sleep and after 24 and 35 h of sleep deprivation: correlations
of fronto-parietal activation with performance. NeuroImage. 2006;31(1):419–28.
Chen JC, Brunner RL, Ren H, Wassertheil-Smoller S, Larson JC, Levine DW, et al. Sleep duration
and risk of ischemic stroke in postmenopausal women. Stroke. 2008;39(12):3185–92.
Cirelli C. How sleep deprivation affects gene expression in the brain: a review of recent findings. J
Appl Physiol (1985). 2002;92(1):394–400.
Cirelli C, Tononi G. Gene expression in the brain across the sleep-waking cycle. Brain Res.
2000;885(2):303–21.
Cirelli C, Gutierrez CM, Tononi G. Extensive and divergent effects of sleep and wakefulness on
brain gene expression. Neuron. 2004;41(1):35–43.
Cirelli C, Faraguna U, Tononi G. Changes in brain gene expression after long-term sleep depriva-
tion. J Neurochem. 2006;98(5):1632–45.
Colbert RA, DeLay ML, Klenk EI, Layh-Schmitt G. From HLA-B27 to spondyloarthritis: a journey
through the ER. Immunol Rev. 2010;233(1):181–202.
Colten HR, Altevogt BM, Institute of Medicine Committee on Sleep Medicine and Research. Sleep
disorders and sleep deprivation: an unmet public health problem. Washington, DC: Institute of
Medicine, National Academies Press; 2006.
Connor J, Norton R, Ameratunga S, Robinson E, Civil I, Dunn R, et al. Driver sleepiness and risk of
serious injury to car occupants: population based case control study. BMJ. 2002;324(7346):
1125.
Conti B, Maier R, Barr AM, Morale MC, Lu X, Sanna PP, et al. Region-specific transcriptional
changes following the three antidepressant treatments electro convulsive therapy, sleep depri-
vation and fluoxetine. Mol Psychiatry. 2007;12(2):167–89.
Cullinan SB, Zhang D, Hannink M, Arvisais E, Kaufman RJ, Diehl JA. Nrf2 is a direct PERK
substrate and effector of PERK-dependent cell survival. Mol Cell Biol. 2003;23(20):7198–209.
7 Sleep Loss and the Unfolded Protein Response 143
Dinges D. Probing the limits of functional capability: the effects of sleep loss on short-duration
tasks. In: Broughton RJ, Ogilvie RD, editors. Sleep, arousal, and performance. Boston, MA:
Birkhäuser; 1992. p. 177–88.
Dufey E, Sepulveda D, Rojas-Rivera D, Hetz C. Cellular mechanisms of endoplasmic reticulum
stress signaling in health and disease. 1. An overview. Am J Physiol Cell Physiol. 2014;307(7):
C582–94.
Elliott AS, Huber JD, O’Callaghan JP, Rosen CL, Miller DB. A review of sleep deprivation studies
evaluating the brain transcriptome. Springerplus. 2014;3:728.
Forman MS, Lee VMY, Trojanowski JQ. ‘Unfolding’ pathways in neurodegenerative disease.
Trends Neurosci. 2003;26(8):407–10.
Fu Y, Li J, Lee AS. GRP78/BiP inhibits endoplasmic reticulum BIK and protects human breast
cancer cells against estrogen starvation-induced apoptosis. Cancer Res. 2007;67(8):3734–40.
Goel N, Rao H, Durmer JS, Dinges DF. Neurocognitive consequences of sleep deprivation. Semin
Neurol. 2009;29(4):320–39.
Gotoh T, Mori M. Regulation of apoptosis by molecular chaperones. Tanpakushitsu Kakusan Koso.
2004;49(7 Suppl):1010–3.
Grandner MA. Addressing sleep disturbances: an opportunity to prevent cardiometabolic disease?
Int Rev Psychiatry. 2014;26(2):155–76.
Grandner MA, Chakravorty S, Perlis ML, Oliver L, Gurubhagavatula I. Habitual sleep duration
associated with self-reported and objectively determined cardiometabolic risk factors. Sleep
Med. 2014;15(1):42–50.
Hakim F, Wang Y, Carreras A, Hirotsu C, Zhang J, Peris E, et al. Chronic sleep fragmentation
during the sleep period induces hypothalamic endoplasmic reticulum stress and PTP1b-
mediated leptin resistance in male mice. Sleep. 2015;38(1):31–40.
Harding HP, Zhang YH, Bertolotti A, Zeng HQ, Ron D. Perk is essential for translational regulation
and cell survival during the unfolded protein response. Mol Cell. 2000;5(5):897–904.
Hasnain SZ, Lourie R, Das I, Chen AC, McGuckin MA. The interplay between endoplasmic
reticulum stress and inflammation. Immunol Cell Biol. 2012;90(3):260–70.
He CH, Gong P, Hu B, Stewart D, Choi ME, Choi AM, et al. Identification of activating
transcription factor 4 (ATF4) as an Nrf2-interacting protein. Implication for heme oxygenase-
1 gene regulation. J Biol Chem. 2001;276(24):20858–65.
Healy SJ, Gorman AM, Mousavi-Shafaei P, Gupta S, Samali A. Targeting the endoplasmic
reticulum-stress response as an anticancer strategy. Eur J Pharmacol. 2009;625(1–3):234–46.
Heazlewood CK, Cook MC, Eri R, Price GR, Tauro SB, Taupin D, et al. Aberrant mucin assembly
in mice causes endoplasmic reticulum stress and spontaneous inflammation resembling ulcer-
ative colitis. PLoS Med. 2008;5(3):e54.
Hersey P, Zhang XD. Adaptation to ER stress as a driver of malignancy and resistance to therapy in
human melanoma. Pigment Cell Melanoma Res. 2008;21(3):358–67.
Hetz C, Mollereau B. Disturbance of endoplasmic reticulum proteostasis in neurodegenerative
diseases. Nat Rev Neurosci. 2014;15(4):233–49.
Hetz C, Martinon F, Rodriguez D, Glimcher LH. The unfolded protein response: integrating stress
signals through the stress sensor IRE1alpha. Physiol Rev. 2011;91(4):1219–43.
Hollien J, Weissman JS. Decay of endoplasmic reticulum-localized mRNAs during the unfolded
protein response. Science. 2006;313(5783):104–7.
Hotamisligil GS. Role of endoplasmic reticulum stress and c-Jun NH2-terminal kinase pathways in
inflammation and origin of obesity and diabetes. Diabetes. 2005;54(Suppl 2):S73–8.
Hotamisligil GS. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease.
Cell. 2010;140(6):900–17.
Hu P, Han Z, Couvillon AD, Kaufman RJ, Exton JH. Autocrine tumor necrosis factor alpha links
endoplasmic reticulum stress to the membrane death receptor pathway through IRE1alpha-
mediated NF-kappaB activation and down-regulation of TRAF2 expression. Mol Cell Biol.
2006;26(8):3071–84.
144 N. Naidoo
Huang RF, Huang SM, Lin BS, Wei JS, Liu TZ. Homocysteine thiolactone induces apoptotic DNA
damage mediated by increased intracellular hydrogen peroxide and caspase 3 activation in
HL-60 cells. Life Sci. 2001;68(25):2799–811.
Hybiske K, Fu Z, Schwarzer C, Tseng J, Do J, Huang N, et al. Effects of cystic fibrosis
transmembrane conductance regulator and DeltaF508CFTR on inflammatory response, ER
stress, and Ca2+ of airway epithelia. Am J Physiol Lung Cell Mol Physiol. 2007;293(5):
L1250–60.
Ishii T, Itoh K, Takahashi S, Sato H, Yanagawa T, Katoh Y, et al. Transcription factor Nrf2
coordinately regulates a group of oxidative stress-inducible genes in macrophages. J Biol Chem.
2000;275(21):16023–9.
Itoh K, Wakabayashi N, Katoh Y, Ishii T, Igarashi K, Engel JD, et al. Keap1 represses nuclear
activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal
Neh2 domain. Genes Dev. 1999;13(1):76–86.
Johnson JA, Johnson DA, Kraft AD, Calkins MJ, Jakel RJ, Vargas MR, et al. The Nrf2-ARE
pathway: an indicator and modulator of oxidative stress in neurodegeneration. Ann N Y Acad
Sci. 2008;1147:61–9.
Jones S, Pfister-Genskow M, Benca RM, Cirelli C. Molecular correlates of sleep and wakefulness in
the brain of the white-crowned sparrow. J Neurochem. 2008;105(1):46–62.
Kammoun HL, Chabanon H, Hainault I, Luquet S, Magnan C, Koike T, et al. GRP78 expression
inhibits insulin and ER stress-induced SREBP-1c activation and reduces hepatic steatosis in
mice. J Clin Invest. 2009;119(5):1201–15.
Kaufman RJ. Orchestrating the unfolded protein response in health and disease. J Clin Invest.
2002;110(10):1389–98.
Knutson KL. Does inadequate sleep play a role in vulnerability to obesity? Am J Hum Biol.
2012;24(3):361–71.
Knutson KL, Van Cauter E, Rathouz PJ, DeLeire T, Lauderdale DS. Trends in the prevalence of
short sleepers in the USA: 1975-2006. Sleep. 2010;33(1):37–45.
Koh K, Evans JM, Hendricks JC, Sehgal A. A Drosophila model for age-associated changes in
sleep: wake cycles. Proc Natl Acad Sci U S A. 2006;103(37):13843–7.
Kuo TH, Williams JA. Acute sleep deprivation enhances post-infection sleep and promotes survival
during bacterial infection in Drosophila. Sleep. 2014;37(5):859–69.
Kuo TH, Pike DH, Beizaeipour Z, Williams JA. Sleep triggered by an immune response in
Drosophila is regulated by the circadian clock and requires the NFkappaB Relish. BMC
Neurosci. 2010;11:17.
Lee AS. GRP78 induction in cancer: therapeutic and prognostic implications. Cancer Res. 2007;67
(8):3496–9.
Lee AS, Hendershot LM. ER stress and cancer. Cancer Biol Ther. 2006;5(7):721–2.
Lee K, Tirasophon W, Shen X, Michalak M, Prywes R, Okada T, et al. IRE1-mediated unconven-
tional mRNA splicing and S2P-mediated ATF6 cleavage merge to regulate XBP1 in signaling
the unfolded protein response. Genes Dev. 2002;16(4):452–66.
Lee K, Neigeborn L, Kaufman RJ. The unfolded protein response is required for haploid tolerance
in yeast. J Biol Chem. 2003;278(14):11818–27.
Li J, Lee AS. Stress induction of GRP78/BiP and its role in cancer. Curr Mol Med. 2006;6(1):
45–54.
Ly S, Lee DA, Strus E, Prober DA, Naidoo N. Evolutionarily conserved regulation of sleep by the
protein translational regulator PERK. Curr Biol. 2020;30(9):1639–48 e3.
Mackiewicz M, Shockley KR, Romer MA, Galante RJ, Zimmerman JE, Naidoo N, et al. Macro-
molecule biosynthesis: a key function of sleep. Physiol Genomics. 2007;31(3):441–57.
Maret S, Dorsaz S, Gurcel L, Pradervand S, Petit B, Pfister C, et al. Homer1a is a core brain
molecular correlate of sleep loss. Proc Natl Acad Sci U S A. 2007;104(50):20090–5.
Maurel M, Chevet E, Tavernier J, Gerlo S. Getting RIDD of RNA: IRE1 in cell fate regulation.
Trends Biochem Sci. 2014;39(5):245–54.
7 Sleep Loss and the Unfolded Protein Response 145
McCullough KD, Martindale JL, Klotz LO, Aw TY, Holbrook NJ. Gadd153 sensitizes cells to
endoplasmic reticulum stress by down-regulating Bcl2 and perturbing the cellular redox state.
Mol Cell Biol. 2001;21(4):1249–59.
McGuckin MA, Eri RD, Das I, Lourie R, Florin TH. ER stress and the unfolded protein response in
intestinal inflammation. Am J Physiol Gastrointest Liver Physiol. 2010;298(6):G820–32.
Mendelson WB, Bergmann BM. Age-dependent changes in recovery sleep after 48 hours of sleep
deprivation in rats. Neurobiol Aging. 2000;21(5):689–93.
Mhaille AN, McQuaid S, Windebank A, Cunnea P, McMahon J, Samali A, et al. Increased
expression of endoplasmic reticulum stress-related signaling pathway molecules in multiple
sclerosis lesions. J Neuropathol Exp Neurol. 2008;67(3):200–11.
Motohashi H, Yamamoto M. Nrf2-Keap1 defines a physiologically important stress response
mechanism. Trends Mol Med. 2004;10(11):549–57.
Naidoo N. Cellular stress/the unfolded protein response: relevance to sleep and sleep disorders.
Sleep Med Rev. 2009;13(3):195–204.
Naidoo N, Giang W, Galante RJ, Pack AI. Sleep deprivation induces the unfolded protein response
in mouse cerebral cortex. J Neurochem. 2005;92(5):1150–7.
Naidoo N, Casiano V, Cater J, Zimmerman J, Pack AI. A role for the molecular chaperone protein
BiP/GRP78 in Drosophila sleep homeostasis. Sleep. 2007;30(5):557–65.
Naidoo N, Ferber M, Master M, Zhu Y, Pack AI. Aging impairs the unfolded protein response to
sleep deprivation and leads to proapoptotic signaling. J Neurosci. 2008;28(26):6539–48.
Naidoo N, Zhu J, Zhu Y, Fenik P, Lian J, Galante R, et al. Endoplasmic reticulum stress in wake-
active neurons progresses with aging. Aging Cell. 2011;10(4):640–9.
Naidoo N, Davis JG, Zhu J, Yabumoto M, Singletary K, Brown M, et al. Aging and sleep
deprivation induce the unfolded protein response in the pancreas: implications for metabolism.
Aging Cell. 2014;13(1):131–41.
Nechushtan A, Smith CL, Hsu YT, Youle RJ. Conformation of the Bax C-terminus regulates
subcellular location and cell death. EMBO J. 1999;18(9):2330–41.
Nguyen DT, Kebache S, Fazel A, Wong HN, Jenna S, Emadali A, et al. Nck-dependent activation
of extracellular signal-regulated kinase-1 and regulation of cell survival during endoplasmic
reticulum stress. Mol Biol Cell. 2004;15(9):4248–60.
Okada T, Yoshida H, Akazawa R, Negishi M, Mori K. Distinct roles of activating transcription
factor 6 (ATF6) and double-stranded RNA-activated protein kinase-like endoplasmic reticulum
kinase (PERK) in transcription during the mammalian unfolded protein response. Biochem
J. 2002;366(Pt 2):585–94.
Outinen PA, Sood SK, Pfeifer SI, Pamidi S, Podor TJ, Li J, et al. Homocysteine-induced endo-
plasmic reticulum stress and growth arrest leads to specific changes in gene expression in human
vascular endothelial cells. Blood. 1999;94(3):959–67.
Pirot P, Naamane N, Libert F, Magnusson NE, Orntoft TF, Cardozo AK, et al. Global profiling of
genes modified by endoplasmic reticulum stress in pancreatic beta cells reveals the early
degradation of insulin mRNAs. Diabetologia. 2007;50(5):1006–14.
Pyrko P, Schonthal AH, Hofman FM, Chen TC, Lee AS. The unfolded protein response regulator
GRP78/BiP as a novel target for increasing chemosensitivity in malignant gliomas. Cancer Res.
2007;67(20):9809–16.
Qureshi AI, Giles WH, Croft JB, Bliwise DL. Habitual sleep patterns and risk for stroke and
coronary heart disease: a 10-year follow-up from NHANES I. Neurology. 1997;48(4):904–11.
Rao RV, Peel A, Logvinova A, del Rio G, Hermel E, Yokota T, et al. Coupling endoplasmic
reticulum stress to the cell death program: role of the ER chaperone GRP78. FEBS Lett.
2002;514(2–3):122–8.
Reddy PH, Williams M, Tagle DA. Recent advances in understanding the pathogenesis of
Huntington’s disease. Trends Neurosci. 1999;22(6):248–55.
Reddy RK, Mao C, Baumeister P, Austin RC, Kaufman RJ, Lee AS. Endoplasmic reticulum
chaperone protein GRP78 protects cells from apoptosis induced by topoisomerase inhibitors:
146 N. Naidoo
role of ATP binding site in suppression of caspase-7 activation. J Biol Chem. 2003;278(23):
20915–24.
Ron D. Hyperhomocysteinemia and function of the endoplasmic reticulum. J Clin Invest. 2001;107
(10):1221–2.
Ron D. Translational control in the endoplasmic reticulum stress response. J Clin Invest. 2002;110
(10):1383–8.
Rumpf RC, Pazos JJ. Optimization of planar self-collimating photonic crystals. J Opt Soc Am A
Opt Image Sci Vis. 2013;30(7):1297–304.
Santos CX, Tanaka LY, Wosniak J, Laurindo FR. Mechanisms and implications of reactive oxygen
species generation during the unfolded protein response: roles of endoplasmic reticulum
oxidoreductases, mitochondrial electron transport, and NADPH oxidase. Antioxid Redox Sig-
nal. 2009;11(10):2409–27.
Scheper W, Hoozemans JJ. The unfolded protein response in neurodegenerative diseases: a
neuropathological perspective. Acta Neuropathol. 2015;130(3):315–31.
Shaw PJ, Cirelli C, Greenspan RJ, Tononi G. Correlates of sleep and waking in Drosophila
melanogaster. Science. 2000a;287(5459):1834–7.
Shaw MK, He CY, Roos DS, Tilney LG. Proteasome inhibitors block intracellular growth and
replication of Toxoplasma gondii. Parasitology. 2000b;121(Pt 1):35–47.
Shiromani PJ, Lu J, Wagner D, Thakkar J, Greco MA, Basheer R, et al. Compensatory sleep
response to 12 h wakefulness in young and old rats. Am J Physiol Regul Integr Comp Physiol.
2000;278(1):R125–33.
Spiegel K, Knutson K, Leproult R, Tasali E, Van Cauter E. Sleep loss: a novel risk factor for insulin
resistance and Type 2 diabetes. J Appl Physiol (1985). 2005;99(5):2008–19.
Terao A, Steininger TL, Hyder K, Apte-Deshpande A, Ding J, Rishipathak D, et al. Differential
increase in the expression of heat shock protein family members during sleep deprivation and
during sleep. Neuroscience. 2003;116(1):187–200.
Terao A, Wisor JP, Peyron C, Apte-Deshpande A, Wurts SW, Edgar DM, et al. Gene expression in
the rat brain during sleep deprivation and recovery sleep: an Affymetrix GeneChip study.
Neuroscience. 2006;137(2):593–605.
Ulmer CS, Calhoun PS, Edinger JD, Wagner HR, Beckham JC. Sleep disturbance and baroreceptor
sensitivity in women with posttraumatic stress disorder. J Trauma Stress. 2009;22(6):643–7.
Upton JP, Wang L, Han D, Wang ES, Huskey NE, Lim L, et al. IRE1alpha cleaves select
microRNAs during ER stress to derepress translation of proapoptotic Caspase-2. Science.
2012;338(6108):818–22.
Urano F, Bertolotti A, Ron D. IRE1 and efferent signaling from the endoplasmic reticulum. J Cell
Sci. 2000;113(Pt 21):3697–702.
Vandewynckel YP, Laukens D, Geerts A, Bogaerts E, Paridaens A, Verhelst X, et al. The paradox
of the unfolded protein response in cancer. Anticancer Res. 2013;33(11):4683–94.
Venugopal R, Jaiswal AK. Nrf1 and Nrf2 positively and c-Fos and Fra1 negatively regulate the
human antioxidant response element-mediated expression of NAD(P)H: quinone oxidoreduc-
tase1 gene. Proc Natl Acad Sci U S A. 1996;93(25):14960–5.
Walter P, Ron D. The unfolded protein response: from stress pathway to homeostatic regulation.
Science. 2011;334(6059):1081–6.
Wang M, Kaufman RJ. The impact of the endoplasmic reticulum protein-folding environment on
cancer development. Nat Rev Cancer. 2014;14(9):581–97.
Welsh DK, Richardson GS, Dement WC. Effect of age on the circadian pattern of sleep and
wakefulness in the mouse. J Gerontol. 1986;41(5):579–86.
Williams JA, Sathyanarayanan S, Hendricks JC, Sehgal A. Interaction between sleep and the
immune response in Drosophila: a role for the NFkappaB relish. Sleep. 2007;30(4):389–400.
Yoshida H, Haze K, Yanagi H, Yura T, Mori K. Identification of the cis-acting endoplasmic
reticulum stress response element responsible for transcriptional induction of mammalian
glucose-regulated proteins. Involvement of basic leucine zipper transcription factors. [erratum
appears in J Biol Chem 1999 Jan 22;274(4):2592]. J Biol Chem. 1998;273(50):33741–9.
7 Sleep Loss and the Unfolded Protein Response 147
Yoshida H, Matsui T, Yamamoto A, Okada T, Mori K. XBP1 mRNA is induced by ATF6 and
spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell.
2001;107(7):881–91.
Yoshida H, Matsui T, Hosokawa N, Kaufman RJ, Nagata K, Mori K. A time-dependent phase shift
in the mammalian unfolded protein response. Dev Cell. 2003;4(2):265–71.
Zhang K, Kaufman RJ. From endoplasmic-reticulum stress to the inflammatory response. Nature.
2008;454(7203):455–62.
Zhang LH, Zhang X. Roles of GRP78 in physiology and cancer. J Cell Biochem. 2010;110(6):
1299–305.
Zhang C, Cai Y, Adachi MT, Oshiro S, Aso T, Kaufman RJ, et al. Homocysteine induces
programmed cell death in human vascular endothelial cells through activation of the unfolded
protein response. J Biol Chem. 2001;276(38):35867–74.
Zhu Y, Fenik P, Zhan G, Sanfillipo-Cohn B, Naidoo N, Veasey SC. Eif-2a protects brainstem
motoneurons in a murine model of sleep apnea. J Neurosci. 2008;28(9):2168–78.
Part III
Sleep Apnea
Chapter 8
Biological and Genetic Mechanisms
of Sleepiness in Obstructive Sleep Apnea
and Cardiovascular Disease
Victoria M. Pak
8.1 Introduction
V. M. Pak (*)
Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, USA
Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA,
USA
e-mail: Victoria.m.pak@emory.edu
© Springer Nature B.V. and Shanghai Jiao Tong University Press 2022 151
A. I. Pack, Q. Y. Li (eds.), Sleep and its Disorders, Translational Medicine Research,
https://doi.org/10.1007/978-94-024-2168-2_8
152 V. M. Pak
women and 16% of men experience excessive daytime sleepiness (EDS) (Young
et al. 1993). EDS can lead to substantial impairment in quality of life and cognitive
function (Findley et al. 1986). However, not all OSA patients develop EDS at any
level of disease severity (Engleman et al. 1997), which suggests biological and
genetic factors may be involved.
EDS is defined in the International Classification of Sleep Disorders (ICSD),
based on behavior of falling asleep, or difficulty maintaining alertness or wakeful-
ness and unintentionally falling asleep. EDS is the most common daytime symptom
of OSA (Pagel 2009). Daytime sleepiness associated with OSA has been found to be
the only sleep disturbance symptom associated with total and cardiovascular mor-
tality in adults, although the mechanism is unidentified (Newman et al. 2000). The
mechanism for the development of EDS in OSA and how this may play a role in
cardiovascular risk is unknown. Understanding mechanisms of EDS associated with
OSA have great significance for public health in order to improve treatment options,
symptom management, and reduce cardiovascular risk.
I review selected genetic and biological markers of oxidative stress and inflamma-
tion that show promise in translational medicine. Exploration of these select path-
ways will identify future molecular targets in order to improve treatment options and
reduce cardiovascular disease risk. There is a need for further studies to examine the
utility of the most promising biomarkers to predict sleepiness phenotype and
response to treatment.
Several candidate genes may be involved in oxidative stress pathway and sleepiness
symptoms.
NOX-2 (CYBB Gene) Measurable amounts of reactive oxygen species (ROS) in the
serum can be largely attributed to NOX-2 (Violi et al. 2006). The observed variabil-
ity in ROS might be partly due to genetic variability of the NOX-2 enzymatic
complex (Bedard et al. 2009). Increased levels of ROS in rodent models are
associated with the presence of intermittent hypoxia-induced dysfunction of the
central nervous system (Wang et al. 2010). Association studies exploring genetic
variability in this gene and the link to sleep symptoms and cardiovascular risk will be
important.
NOX p22phox Gene Overproduction of ROS is associated with neurodegenerative
disorders and cardiovascular impairments (Tabner et al. 2005). The major sources of
ROS in the vasculature are the NOX oxidases whose sole function is to generate
ROS (Selemidis et al. 2008). Several SNPs related to NOX expression or activity
have been identified (Gozal et al. 2012; Pierola et al. 2011). SNPs in the NOX
p22phox gene may account for differences among OSA children with and without
cognitive deficits (Gozal et al. 2012). A previous study has shown that the SNP
rs4673 was associated with reduced levels of NOX activity and 8-OH-dG urinary
concentrations, and accounted for part of the discrepant phenotypic expression in
cognitive functioning in the context of pediatric OSA (Gozal et al. 2012). Thus, the
differential cognitive symptoms in the context of OSA could partly be explained by
154 V. M. Pak
the presence of significantly higher levels of oxidative stress among children with
cognitive deficits. Future studies should consider the potential link between oxida-
tive stress and sleepiness symptoms and explore SNPs in the NOX p22phox gene.
Although the mechanism for the initiation of cardiovascular disease in OSA has not
been fully established, the generally accepted theory is the intermittent hypoxia
(IH) produced by frequent respiratory events (Pak et al. 2014). Pre-atherosclerotic
remodeling of large arteries with enlarged intima media thickness under intermittent
hypoxic conditions were found to be inflammatory in nature with biochemical
evidence (greater nuclear factor [NF]-kB expression) (Arnaud et al. 2011). The
link of OSA to cardiovascular sequelae is demonstrated in rodent models, where
intermittent hypoxia produces a moderate BP elevation starting from 5 to 8 days after
onset (Dematteis et al. 2009). Although daytime sleepiness associated with OSA is
the only sleep disturbance symptom associated with total and cardiovascular mor-
tality in adults, the factors involved in this link remain unidentified (Newman et al.
2000). In an exploratory case control study of 36 subjects exploring associations
between subjective sleepiness and metabolite concentrations within the oxidative
and inflammatory pathways, choline was found to be significantly lower in sleepy
subjects (ESS 10) compared with non-sleepy subjects. Other markers with
suggestive differences (P < 0.1) included isovalerylcarnitine, alpha-amino adipic
acid, sphingosine 1 phosphate, aspartic acid, propionylcarnitine, and ceramides
(fatty acids; C14–C16 and C–18) (Pak et al. 2018b). One prior study has specifically
measured sleepiness and objective cardiovascular risk in OSA (Choi et al. 2006;
Empana et al. 2009). Choi et al. found sleepiness independently associated with
decreased cardiac function as assessed by impedance cardiography in 86 patients
with suspected OSA who then underwent confirmatory diagnostic polysomnography
(Choi et al. 2006). However, this study was limited by a small sample size, self-
report of sleepiness, and not controlling for obesity or hypertension. Furthermore,
impedance cardiography is not easily reproducible as the results are highly depen-
dent on the skill of the operator. In another study by Empana et al., investigators
found that elderly people with excessive daytime sleepiness had a 49% increase in
relative risk of cardiovascular death and a 33% increase in relative risk of overall
156 V. M. Pak
Fig. 8.1 Schematic illustration of the proposed role of oxidative stress and inflammation in OSA,
potential molecular targets, and genetic factors in the pathophysiology of sleepiness and cardio-
vascular disease
death (Empana et al. 2009). This study is limited by self-reported patient responses
and the use of ultrasonography of the carotid arteries, which are highly operator
dependent. In addition, this study did not specifically examine OSA. Thus, there may
be a confounding effect of underlying sleep apnea syndrome.
Future studies are needed to extend these study findings using more robust
measures of both sleepiness and cardiovascular risk. The Psychomotor Vigilance
Test (PVT) is a gold standard for the assessment of neurobehavioral impairment
associated with sleep loss (Dinges et al. 1994). This is a simple test (10-min test)
used to classify subjects as sleepy (PVT 2 lapses on systematic
3 trials) vs. non-sleepy (PVT < 2 on systematic 3 trials) by objective criteria. PVT
is a simple, portable reaction time test designed to evaluate the ability to sustain
attention and measures reaction time to signals presented at random intervals over a
brief period of testing (Dinges and Powell 1985). Ambulatory Blood Pressure
Monitoring is increasingly recognized as a valuable tool to refine prediction of
cardiovascular risk related to blood pressure (Verdecchia et al. 2007). The measure-
ment of Pulse Wave Velocity is accepted as a simple and reproducible method that is
a “gold standard” index to measure arterial stiffness along with 24-h ambulatory
blood pressure (Kohler et al. 2011). The use of robust measures of sleepiness and
cardiovascular disease risk will be important in clarifying the cardiovascular
sequelae associated with sleepiness symptoms. See Fig. 8.1 for schematic and
biological pathways.
Fig. 8.2 Schematic illustration of obstructive sleep apnea and the link to atherosclerosis and
cardiovascular disease, including the role of adhesion molecules. CRP C-reactive protein, ICAM-
1 intercellular adhesion molecule 1, IL-6 interleukin-6, IL-8 interleukin-8, MCP-1 monocyte
chemoattractant protein-1, NF-ҡ B nuclear factor-kappa B, TNF-α tumor necrosis factor α,
VCAM-1 vascular cell adhesion molecule-1. Reproduced from Pak et al. (2014)
vascular endothelial cells and migration into the vessel wall is critical in the
development of atherosclerosis (Pak et al. 2014). The repeated episodes of hypoxia
followed by reoxygenation that is characteristic of OSA is proposed to result in
oxidative stress and increased production of reactive oxygen species (Lavie et al.
2004), see Fig. 8.2.
Continuous positive airway pressure treatment has been shown to have a bene-
ficial impact on blood pressure only in patients with OSA who were sleepy and not in
non-sleepy patients (Robinson et al. 2004). These results suggest that daytime
sleepiness may be a significant factor in the pathogenesis of cardiovascular disease.
This finding also reinforces the theory that elevated inflammation and oxidative
stress in sleep apnea patients lead to sleepiness, thus increasing cardiovascular
disease risk.
The logical focus of potential therapies would be to target oxidative stress and
inflammation which causes the increase in cardiovascular disease risk. For instance,
potential therapies for reducing circulating adhesion molecules to diminish cardio-
vascular disease in OSA include CPAP and antioxidant supplementation (Pak et al.
2014). Specific molecular targets include intercellular adhesion molecule-1 (ICAM-
1) and vascular cell adhesion molecule-1 (VCAM-1), which are expressed on cell
surfaces and found in their soluble forms in the plasma (Ballantyne and Entman
2002). Leukocyte adhesion to vascular endothelial cells and migration into the vessel
wall is critical in the development of atherosclerosis, and elevated levels of adhesion
molecules have been demonstrated in subjects with OSA (Ursavas et al. 2007; Ohga
158 V. M. Pak
Fig. 8.3 Predicted least squares mean change from baseline in entire population. The predicted
least squares mean change in ICAM-1 and VCAM-1 levels for each PAP usage group are presented
in the overall population, along with the associated 95% confidence intervals. Results are adjusted
for baseline ICAM-1 or VCAM-1 levels, BMI, change in BMI, hypertension at baseline and follow-
up and statin use at baseline and follow-up (for VCAM-1 change only). Estimates with 95%
confidence intervals not crossing 0 represent significant changes. *P < 0.05. LS least squares.
Reproduced from Pak et al. (2015)
et al. 1999, 2003; El-Solh et al. 2002; Chin et al. 2000; Zamarron et al. 2011;
Zamarron-Sanz et al. 2006). Positive airway pressure (PAP) therapy may reduce
ICAM-1 levels in OSA patients (Chin et al. 2000; Ohga et al. 2003; Zamarron et al.
2011). One prior study has examined VCAM-1, finding no PAP treatment effect
(Chin et al. 2000). However, these studies had small samples and relatively short
duration, and did not directly address the role of obesity on these relationships. See
Fig. 8.3 for an illustration of how in a moderate-to-severe OSA population, adequate
PAP usage limits significant increases in both ICAM-1 and VCAM-1 levels
observed in PAP nonusers after 2 years (Pak et al. 2015). As obesity and OSA
often coexist, limiting these increases with PAP usage may help to decrease the rate
of progression of OSA-related cardiovascular disease. For ICAM-1, full usage
decreased levels, with the largest effect occurring in the most obese subjects (Pak
et al. 2015). For VCAM-1, increased levels over 2 years were observed for all PAP
groups, but nonusers had much larger increases than full users. Although the
VCAM-1 association appeared independent of obesity, larger increases in nonusers
were again seen in the most obese. Obesity is an important risk factor for OSA
(Young et al. 1993), and their shared pathways of oxidative stress and inflammation
make discerning independent roles of obesity and OSA in cardiovascular disease
difficult (Arnardottir et al. 2009). Future studies should consider the role of obesity
when exploring the relationship between sleepiness and cardiovascular disease risk
and the effect of treatment.
The exploration of subgroups to clarify the genetic and biological profile that
ultimately predicts cardiovascular disease risk and explore the effects of intervention
8 Biological and Genetic Mechanisms of Sleepiness in Obstructive Sleep. . . 159
Overall, the preliminary research findings suggest that biological and genetic vari-
ation is involved in the etiology of inflammation and oxidative stress may potentially
impact sleepiness symptoms and cardiovascular disease risk. Replication of these
findings, in combination with exploring how interventions targeting specific mole-
cules within the oxidative stress and inflammatory pathway modulates an individ-
ual’s susceptibility, will be important to guide treatment and management of
symptoms and the prevention of cardiovascular disease. Replication of current
genetic findings remains a challenge, however, with consistent control of con-
founders, consistent phenotype definitions, larger sample sizes, and consideration
of population ethnicity, replication of findings will become more attainable.
References
Arnardottir ES, Mackiewicz M, Gislason T, Teff KL, Pack AI. Molecular signatures of obstructive
sleep apnea in adults: a review and perspective. Sleep. 2009;32(4):447–70.
Arnaud C, Beguin PC, Lantuejoul S, Pepin JL, Guillermet C, Pelli G, et al. The inflammatory
preatherosclerotic remodeling induced by intermittent hypoxia is attenuated by RANTES/CCL5
inhibition. Am J Respir Crit Care Med. 2011;184(6):724–31.
Ballantyne CM, Entman ML. Soluble adhesion molecules and the search for biomarkers for
atherosclerosis. Circulation. 2002;106(7):766–7.
Bast A, Haenen GR, Doelman CJ. Oxidants and antioxidants: state of the art. Am J Med. 1991;91
(3C):2S–13S.
Bedard K, Attar H, Bonnefont J, Jaquet V, Borel C, Plastre O, et al. Three common polymorphisms
in the CYBA gene form a haplotype associated with decreased ROS generation. Hum Mutat.
2009;30(7):1123–33.
Blankenberg S, Rupprecht HJ, Bickel C, Peetz D, Hafner G, Tiret L, et al. Circulating cell adhesion
molecules and death in patients with coronary artery disease. Circulation. 2001;104(12):
1336–42.
Blankenberg S, Barbaux S, Tiret L. Adhesion molecules and atherosclerosis. Atherosclerosis.
2003;170(2):191–203.
160 V. M. Pak
Carpagnano GE, Spanevello A, Sabato R, Depalo A, Palladino GP, Bergantino L, et al. Systemic
and airway inflammation in sleep apnea and obesity: the role of ICAM-1 and IL-8. Transl Res.
2010;155(1):35–43.
Chin K, Nakamura T, Shimizu K, Mishima M, Nakamura T, Miyasaka M, et al. Effects of nasal
continuous positive airway pressure on soluble cell adhesion molecules in patients with obstruc-
tive sleep apnea syndrome. Am J Med. 2000;109(7):562–7.
Choi JB, Nelesen R, Loredo JS, Mills PJ, Ancoli-Israel S, Ziegler MG, et al. Sleepiness in
obstructive sleep apnea: a harbinger of impaired cardiac function? Sleep. 2006;29(12):1531–6.
Das UN. Is obesity an inflammatory condition? Nutrition. 2001;17(11–12):953–66.
Dematteis M, Godin-Ribuot D, Arnaud C, Ribuot C, Stanke-Labesque F, Pepin JL, et al. Cardio-
vascular consequences of sleep-disordered breathing: contribution of animal models to under-
standing the human disease. ILAR J. 2009;50(3):262–81.
Dinges DF, Powell JW. Microcomputer analyses of performance on a portable, simple visual Rt
task during sustained operations. Behav Res Methods Instrum Comput. 1985;17(6):652–5.
Dinges DF, Douglas SD, Zaugg L, Campbell DE, McMann JM, Whitehouse WG, et al. Leukocy-
tosis and natural killer cell function parallel neurobehavioral fatigue induced by 64 hours of
sleep deprivation. J Clin Invest. 1994;93(5):1930–9.
El-Solh AA, Mador MJ, Sikka P, Dhillon RS, Amsterdam D, Grant BJ. Adhesion molecules in
patients with coronary artery disease and moderate-to-severe obstructive sleep apnea. Chest.
2002;121(5):1541–7.
Empana JP, Dauvilliers Y, Dartigues JF, Ritchie K, Gariepy J, Jouven X, et al. Excessive daytime
sleepiness is an independent risk indicator for cardiovascular mortality in community-dwelling
elderly: the three city study. Stroke. 2009;40(4):1219–24.
Engleman HM, Hirst WS, Douglas NJ. Under reporting of sleepiness and driving impairment in
patients with sleep apnoea/hypopnoea syndrome. J Sleep Res. 1997;6(4):272–5.
Findley LJ, Barth JT, Powers DC, Wilhoit SC, Boyd DG, Suratt PM. Cognitive impairment in
patients with obstructive sleep apnea and associated hypoxemia. Chest. 1986;90(5):686–90.
Gottlieb DJ, O’Connor GT, Wilk JB. Genome-wide association of sleep and circadian phenotypes.
BMC Med Genet. 2007;8(Suppl 1(Journal Article)):S9.
Gozal D, Serpero LD, Kheirandish-Gozal L, Capdevila OS, Khalyfa A, Tauman R. Sleep measures
and morning plasma TNF-alpha levels in children with sleep-disordered breathing. Sleep.
2010;33(3):319–25.
Gozal D, Khalyfa A, Capdevila OS, Kheirandish-Gozal L, Khalyfa AA, Kim J. Cognitive function
in prepubertal children with obstructive sleep apnea: a modifying role for NADPH oxidase p22
subunit gene polymorphisms? Antioxid Redox Signal. 2012;16(2):171–7.
Gray SL, Hanlon JT, Landerman LR, Artz M, Schmader KE, Fillenbaum GG. Is antioxidant use
protective of cognitive function in the community-dwelling elderly? Am J Geriatr
Pharmacother. 2003;1(1):3–10.
Halliwell B, Whiteman M. Measuring reactive species and oxidative damage in vivo and in cell
culture: how should you do it and what do the results mean? Br J Pharmacol. 2004;142(2):
231–55.
Khalyfa A, Serpero LD, Kheirandish-Gozal L, Capdevila OS, Gozal D. TNF-alpha gene poly-
morphisms and excessive daytime sleepiness in pediatric obstructive sleep apnea. J Pediatr.
2011;158(1):77–82.
Kim CD, Kim YK, Lee SH, Hong KW. Rebamipide inhibits neutrophil adhesion to hypoxia/
reoxygenation-stimulated endothelial cells via nuclear factor-kappaB-dependent pathway. J
Pharmacol Exp Ther. 2000;294(3):864–9.
Kohler M, Stoewhas AC, Ayers L, Senn O, Bloch KE, Russi EW, et al. Effects of continuous
positive airway pressure therapy withdrawal in patients with obstructive sleep apnea: a random-
ized controlled trial. Am J Respir Crit Care Med. 2011;184(10):1192–9.
Krueger JM. The role of cytokines in sleep regulation. Curr Pharm Des. 2008;14(32):3408–16.
Lavie L, Vishnevsky A, Lavie P. Evidence for lipid peroxidation in obstructive sleep apnea. Sleep.
2004;27(1):123–8.
8 Biological and Genetic Mechanisms of Sleepiness in Obstructive Sleep. . . 161
Morrow JD, Roberts LJ. The isoprostanes: their role as an index of oxidant stress status in human
pulmonary disease. Am J Respir Crit Care Med. 2002;166(12 Pt 2):S25–30.
Newman AB, Spiekerman CF, Enright P, Lefkowitz D, Manolio T, Reynolds CF, et al. Daytime
sleepiness predicts mortality and cardiovascular disease in older adults. The Cardiovascular
Health Study Research Group. J Am Geriatr Soc. 2000;48(2):115–23.
Ohga E, Nagase T, Tomita T, Teramoto S, Matsuse T, Katayama H, et al. Increased levels of
circulating ICAM-1, VCAM-1, and L-selectin in obstructive sleep apnea syndrome. J Appl
Physiol (1985). 1999;87(1):10–4.
Ohga E, Tomita T, Wada H, Yamamoto H, Nagase T, Ouchi Y. Effects of obstructive sleep apnea
on circulating ICAM-1, IL-8, and MCP-1. J Appl Physiol (1985). 2003;94(1):179–84.
Pagel JF. Excessive daytime sleepiness. Am Fam Physician. 2009;79(5):391–6.
Pak VM, Grandner MA, Pack AI. Circulating adhesion molecules in obstructive sleep apnea and
cardiovascular disease. Sleep Med Rev. 2014;18(1):25–34.
Pak VM, Keenan BT, Jackson N, Grandner MA, Maislin G, Teff K, et al. Adhesion molecule
increases in sleep apnea: beneficial effect of positive airway pressure and moderation by obesity.
Int J Obes. 2015;39(3):472–9.
Pak VM, Mazzotti DR, Keenan BT, Hirotsu C, Gehrman P, Bittencourt L, et al. Candidate gene
analysis in the Sao Paulo Epidemiologic Sleep Study (EPISONO) shows an association of
variant in PDE4D and sleepiness. Sleep Med. 2018a;47:106–12.
Pak VM, Dai F, Keenan BT, Gooneratne NS, Pack AI. Lower plasma choline levels are associated
with sleepiness symptoms. Sleep Med. 2018b;44:89–96.
Patel SR, Zhu X, Storfer-Isser A, Mehra R, Jenny NS, Tracy R, et al. Sleep duration and biomarkers
of inflammation. Sleep. 2009;32(2):200–4.
Pierola J, Alemany A, Yanez A, de-la-Pena M, Sanchez-de-la-Torre M, Esquinas C, et al. NADPH
oxidase p22phox polymorphisms and oxidative stress in patients with obstructive sleep apnoea.
Respir Med. 2011;105(11):1748–54.
Robinson GV, Smith DM, Langford BA, Davies RJO, Stradling JR. CPAP does not reduce 24 hour
blood pressure in hypertensive obstructive sleep apnoea patients without daytime sleepiness.
Thorax. 2004;59(1):16.
Sawa Y, Sugimoto Y, Ueki T, Ishikawa H, Sato A, Nagato T, et al. Effects of TNF-alpha on
leukocyte adhesion molecule expressions in cultured human lymphatic endothelium. J
Histochem Cytochem. 2007;55(7):721–33.
Sekut L, Yarnall D, Stimpson SA, Noel LS, Bateman-Fite R, Clark RL, et al. Anti-inflammatory
activity of phosphodiesterase (PDE)-IV inhibitors in acute and chronic models of inflammation.
Clin Exp Immunol. 1995;100(1):126–32.
Selemidis S, Sobey CG, Wingler K, Schmidt HH, Drummond GR. NADPH oxidases in the
vasculature: molecular features, roles in disease and pharmacological inhibition. Pharmacol
Ther. 2008;120(3):254–91.
Singh TD, Patial K, Vijayan VK, Ravi K. Oxidative stress and obstructive sleep apnoea syndrome.
Indian J Chest Dis Allied Sci. 2009;51(4):217–24.
Solfrizzi V, Panza F, Capurso A. The role of diet in cognitive decline. J Neural Transm (Vienna).
2003;110(1):95–110.
Tabner BJ, Turnbull S, Fullwood NJ, German M, Allsop D. The production of hydrogen peroxide
during early-stage protein aggregation: a common pathological mechanism in different neuro-
degenerative diseases? Biochem Soc Trans. 2005;33(Pt 4):548–50.
Tsaluchidu S, Cocchi M, Tonello L, Puri BK. Fatty acids and oxidative stress in psychiatric
disorders. BMC Psychiatry. 2008;8(Suppl 1(Journal Article)):S5.
Ursavas A, Karadag M, Rodoplu E, Yilmaztepe A, Oral HB, Gozu RO. Circulating ICAM-1 and
VCAM-1 levels in patients with obstructive sleep apnea syndrome. Respiration. 2007;74(5):
525–32.
Verdecchia P, Angeli F, Cavallini C. Ambulatory blood pressure for cardiovascular risk stratifica-
tion. Circulation. 2007;115(16):2091–3.
162 V. M. Pak
The diaphragm is the most important respiratory muscle and the diaphragm electro-
myogram (EMG) is useful to assess neural respiratory drive. However, the useful-
ness of the diaphragm EMG is dependent on the accurate recording of the signal
Y.-M. Luo
National Heart and Lung Institute, Imperial College, London, UK
Y.-M. Luo (*)
Chinese National Researcher Center for Respiratory Disease, State Key Laboratory of
Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University,
Guangzhou, China
Sleep Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou,
China
e-mail: y.m.luo@vip.163.com
© Springer Nature B.V. and Shanghai Jiao Tong University Press 2022 163
A. I. Pack, Q. Y. Li (eds.), Sleep and its Disorders, Translational Medicine Research,
https://doi.org/10.1007/978-94-024-2168-2_9
164 Y.-M. Luo and Y.-M. Luo
Fig. 9.1 Combined esophageal electrode and its positioning. (a) Configuration of electrode. (b)
Diaphragmatic EMG recorded from multi-pair esophageal electrode after electrode catheter was
optimally positioned, characterized by large EMG signals from pairs 1 and 5, the smallest EMG was
recorded from pair 3. (c) Photograph of combined balloon electrode catheter [Figure from Luo et al.
(2011)]
without artifact (Laveneziana et al. 2019). Although needle electrodes have been
used in humans in some physiological studies, they are impractical for most clinical
practice, particularly during sleep. Surface electrodes could be an alternative method
for recording the diaphragm EMG, but this technique has disadvantages because of
signal contamination, artifacts, and difficulty in standardization in positioning (Luo
et al. 1998). In contrast, the diaphragm EMG recorded from an esophageal electrode
is less affected by obesity and crosstalk signals (Luo et al. 1998). Consequently,
esophageal diaphragm EMG has become popular in research and clinical practice
(Luo et al. 2008a). One or two balloons are usually attached to an electrode catheter
to simultaneously record esophageal pressure or transdiaphragmatic pressure (Pdi)
while recording diaphragm EMG (Luo et al. 2008a). Although diaphragm EMG is
recorded from esophageal electrodes, mainly from the crus, it is usually considered
to be able to accurately reflect the electric activity of the entire diaphragm (American
Thoracic Society/European Respiratory Society 2002).
For better recording of the diaphragm EMG, the esophageal electrode should be
positioned close to the diaphragm. Luo and co-workers developed a technique to
accurately position an esophageal electrode based on the amplitude and polarity of
the diaphragm compound muscle action potential (CMAP) elicited by bilateral
phrenic nerve stimulation (Luo et al. 1999, 2008a). Esophageal electrodes can also
be positioned based on the spontaneous EMG signals recorded simultaneously from
five pairs of electrodes (Luo et al. 2008b). The optimal position was characterized by
a large EMG from two electrode pairs, which shared the middle electrode, and a
small EMG from the pair straddling the middle electrode (Luo et al. 2011) (Fig. 9.1).
When a catheter is positioned properly, the esophageal balloon records a negative
pressure and the gastric balloon a positive pressure during inspiration for a subject
with normal diaphragm function.
9 Diaphragm EMG Recording and Its Application in Sleep Medicine 165
Fig. 9.2 Peak of root mean 200 RMS of EMG during CO2 rebreathing
square (RMS) increases
during CO2 rebreathing.
There is a good relationship
between end-tidal CO2 and
the amplitude of the RMS. 150
No plateau of EMG is
RMS of EMG(uV)
50
0
5.0 7.0 9.0
End-tidal CO2 %
166 Y.-M. Luo and Y.-M. Luo
Fig. 9.3 Esophageal pressure (Pes; white bar), transdiaphragmatic pressure (Pdi; gray bar), and
diaphragm electromyography (EMG; black bar) signal recorded from the esophageal electrode
during obstructive sleep apnea. Data are presented as mean SD. Pes and Pdi increased gradually
over the course of an apnea, reaching a maximum at the end of the apnea, and decreased
significantly at the beginning of arousal. However, the diaphragm EMG signal was similar at the
beginning and middle of the apnea, increased significantly at the end of the apnea, and increased
further at the beginning of arousal [Figure from Luo et al. (2008b)]
9 Diaphragm EMG Recording and Its Application in Sleep Medicine 167
Sleep apnea includes central sleep apnea and obstructive sleep apnea events.
Because the mechanism responsible for central sleep apnea differs from that for
obstructive sleep apnea and the treatment for obstructive apnea also differs from that
for central sleep apnea, it is important for both clinical practice and research to
accurately distinguish central from obstructive sleep apnea. Chest abdominal wall
movement recorded by respiratory inductance plethysmography is usually taken as
respiratory effort or neural respiratory drive for clinical diagnostic purposes. How-
ever, neural respiratory drive may not be reliably reflected by respiratory inductance
plethysmography (RIP) because chest and abdominal wall motion could be
influenced by lung volume and posture, leading to an overestimation of the
168 Y.-M. Luo and Y.-M. Luo
Fig. 9.5 The mean of the esophageal pressure (a) and diaphragm EMG (b) at the end of apneas that
were associated with arousal from sleep are similar to those without arousal. They are not
significantly different [Figure from Xiao et al. (2015)]
frequency of central apnea events (Luo et al. 2009) (Fig. 9.7). In contrast, Pes is
considered to be an accurate method to assess neural respiratory drive and to
distinguish obstructive from central apneic events. However, it has been documented
that Pes can also be affected by variation of flow and lung volume, which occurs
during apnea, especially during hypopnea. Because central apnea by definition, is
due to cessation of neural respiratory drive, diaphragm EMG which is independent
of changes in lung volume and airflow may be superior to other methods in
9 Diaphragm EMG Recording and Its Application in Sleep Medicine 169
Fig. 9.6 Esophageal pressure (Pes) and diaphragm EMG from multiple electrodes during respira-
tory events and before respiratory events are sometimes larger than those at the end of the events,
although arousal occurs only at the end of events for hypopnea (a) and apnea (b) [Figure from Xiao
et al. (2015)]. Traces from top to bottom are diaphragm electromyography from five pairs of
electrodes (EMGdi 1–5), airflow, Oxygen saturation (SaO2), esophageal pressure (Pes), electroen-
cephalography EEG recorded from C3A2 (C3A2), electrooculography recorded from left side
(LOG), and electromyography recorded from chin (Chin)
Fig. 9.7 An OSA episode is incorrectly diagnosed as CSA based on RIP. Signals (from top to
bottom) are airflow, EMGdi 1-EMGdi 5, chest movement, and abdomen movement. This episode
would have been scored as CSA if the judgment was based on the chest and abdominal movement
recorded from RIP alone. However, this episode is scored as OSA based on EMGdi over the course
of an apneic episode (Luo et al. 2009)
central sleep apnea should be further confirmed by diaphragm EMG when the
diagnosis is in doubt. Although diaphragm EMG has traditionally been unwieldy,
recent technical advances have resolved this and could allow the technique to
potentially be the “gold standard” to differentiate central from obstructive apnea
events.
It has been recognized that patients with COPD are subject to hypoxemia or even
respiratory failure during sleep because of hypoventilation (Luo et al. 2014). How-
ever, it is not clear whether hypoventilation in patients with COPD during sleep is
due to increases in upper airway resistance or reductions in neural drive. It is obvious
that COPD patients with OSA (overlap syndrome) will have a sleep-related increase
in upper airway resistance, which will contribute to the reduction of their ventilation
during sleep. It has been shown that the reduction in ventilation from wakefulness to
non-rapid eye movement (NREM) sleep in laryngectomized patients with constant
9 Diaphragm EMG Recording and Its Application in Sleep Medicine 171
upper airway resistance (breathing through a tracheal stoma) is similar to that when
breathing through an intact upper airway, indicating that changes in upper airway
resistance are not always responsible for reduction of ventilation during sleep
(Morrell et al. 1996). We recently have performed a study to determine whether
neural respiratory drive is the main factor contributing to sleep-related
hypoventilation in patients with COPD (Luo et al. 2014). A study was done on
14 patients with COPD, who were free from obstructive sleep apnea (AHI < 5.0
events/h), confirmed by overnight polysomnography, showed that ventilation
decreased during NREM and further decreased during REM when compared with
wakefulness in patients with COPD. With a decrease in ventilation, there was almost
proportional decrease in neural respiratory drive as assessed by diaphragm EMG
(Luo et al. 2014) (Fig. 9.8). Although there was a reduction of the diaphragm EMG
from wakefulness to NREM sleep in normal subjects, the reduction of neural drive in
patients with COPD was much larger than that in normal subjects (Luo et al. 2014),
probably because there is an additional increase in neural respiratory drive from the
Fig. 9.8 Polysomnography including five-channel diaphragm EMG (EMGdi 1–5) from a multi-
pair esophageal electrode, airflow from pneumotachograph (Flow), oxygen saturation (SaO2),
end-tidal CO2, electroencephalogram (EEG; C3A2 and C4A1), and left and right electrooculograms
(EOGs). Compared with wakefulness, EMGdi and airflow decrease in non-rapid eye movement
(NREM), and further decrease in REM. Data are from a patient with chronic obstructive pulmonary
disease (COPD) [Note, SaO2 scale on REM panel differs from the others, figure from Luo et al.
(2014)]
172 Y.-M. Luo and Y.-M. Luo
It has been previously reported that nocturnal oxygen desaturation is more severe in
patients with both COPD and OSA, a phsenotype termed the “overlap syndrome,”
than in those with COPD alone (Marin et al. 2010; Sanders et al. 2003; Chaouat et al.
1995). However, this view is mainly derived from studies of patients who had
predominantly mild or moderate COPD or who were recruited from OSA patients
with obesity at a sleep center (Marin et al. 2010; Sanders et al. 2003; Chaouat et al.
1995), and thus may not represent a clinical cohort of patients with severe COPD
(He et al. 2017). In contrast to hypoventilation in patients with COPD, OSA is
characterized by increased upper airway resistance associated with an increase in
neural respiratory drive (Luo et al. 2009). If patients with severe COPD develop
OSA, the sleep-related reduction in neural respiratory drive associated with COPD
could be offset by the increase in neural respiratory drive in OSA. Consequently,
ventilation in patients with severe COPD may not further decrease when COPD and
mild or modest OSA occur together (He et al. 2017) (Fig. 9.9). Indeed, a recent
Fig. 9.9 Diaphragm EMG (EMGdi)% (left panel), ventilation (middle panel) and the VT/EMGdi
(right panel) in patients with COPD alone, overlap syndrome, normal subjects and patients with
obstructive sleep apnea (OSA). Ventilation decreases from wakefulness to sleep in all four groups
but only in patients with COPD, overlap syndrome and OSA is the reduction statistically significant.
EMGdi decreases significantly in patients with COPD alone but increases significantly in patients
with OSA from wakefulness to non-rapid eye movement (NREM) sleep, whereas it remains
unchanged in normal subjects and patients with overlap syndrome. VT/EMGdi decreases signifi-
cantly from wakefulness to sleep in patients with overlap syndrome and those with OSA but it
changes little in normal subjects and patients with COPD alone. The decrease in ventilation is
associated with a reduction of EMGdi in patients with COPD alone whereas reduction in ventilation
is associated with decreased VT/EMGdi in patients with overlap syndrome and those with OSA
[Figure from Patout et al. (2015)]
9 Diaphragm EMG Recording and Its Application in Sleep Medicine 173
cohort study on nonobese patients with severe COPD showed that the number of
patients who required oxygen supplementation in patients with COPD alone was
similar to that in those with overlap syndrome. This suggests that the prevalence of
oxygen desaturation may be similar between patients with severe COPD alone and
those with overlap syndrome providing that the impairment of lung function is
similar (Soler et al. 2015). Our recent study also found that mean oxygen saturation
and minimal oxygen saturations during overnight sleep were similar in COPD
patients with or without OSA (He et al. 2017). Moreover, although patients with
coexistent OSA and severe COPD usually have brief periods of desaturation,
prolonged desaturation (SaO2 < 90 for longer than 5 min) occurred actually more
often in patients with severe COPD than in those with overlap syndrome (He et al.
2017). These findings suggest that if patients with severe COPD develop mild or
moderate OSA, oxygen desaturation may not necessarily worsen. This view was
supported by a recent study which showed that the clinical outcome of end-stage
patients with overlap syndrome is not worse than those with COPD alone (Patout
et al. 2015; Du et al. 2018). Inconsistent results in this important area indicate more
research is required.
9.8 Conclusion
References
Du W, Liu J, Zhou J, Ye D, OuYang Y, Deng Q. Obstructive sleep apnea, COPD, the overlap
syndrome, and mortality: results from the 2005–2008 National Health and Nutrition Examina-
tion Survey. Int J Chron Obstruct Pulmon Dis. 2018;13:665–74.
Gleeson K, Zwillich CW, White DP. The influence of increasing ventilatory effort on arousal from
sleep. Am Rev Respir Dis. 1990;142(2):295–300.
He BT, Lu G, Xiao SC, Chen R, Steier J, Moxham J, et al. Coexistence of OSA may compensate for
sleep related reduction in neural respiratory drive in patients with COPD. Thorax. 2017;72(3):
256–62.
Kay A, Trinder J, Kim Y. Individual differences in relationship between upper airway resistance
and ventilation during sleep onset. J Appl Physiol (1985). 1995;79(2):411–9.
Kimoff RJ, Cheong TH, Olha AE, Charbonneau M, Levy RD, Cosio MG, et al. Mechanisms of
apnea termination in obstructive sleep apnea. Role of chemoreceptor and mechanoreceptor
stimuli. Am J Respir Crit Care Med. 1994;149(3 Pt 1):707–14.
Laveneziana P, Albuquerque A, Aliverti A, Babb T, Barreiro E, Dres M, et al. ERS statement on
respiratory muscle testing at rest and during exercise. Eur Respir J. 2019;53(6):1801214.
Luo YM, Moxham J. Measurement of neural respiratory drive in patients with COPD. Respir
Physiol Neurobiol. 2005;146(2–3):165–74.
Luo YM, Polkey MI, Johnson LC, Lyall RA, Harris ML, Green M, et al. Diaphragm EMG
measured by cervical magnetic and electrical phrenic nerve stimulation. J Appl Physiol
(1985). 1998;85(6):2089–99.
Luo YM, Lyall RA, Lou Harris M, Rafferty GF, Polkey MI, Moxham J. Quantification of the
esophageal diaphragm electromyogram with magnetic phrenic nerve stimulation. Am J Respir
Crit Care Med. 1999;160(5 Pt 1):1629–34.
Luo YM, Moxham J, Polkey MI. Diaphragm electromyography using an oesophageal catheter:
current concepts. Clin Sci. 2008a;115(8):233–44.
Luo YM, Wu HD, Tang J, Jolley C, Steier J, Moxham J, et al. Neural respiratory drive during
apnoeic events in obstructive sleep apnoea. Eur Respir J. 2008b;31(3):650–7.
Luo YM, Tang J, Jolley C, Steier J, Zhong NS, Moxham J, et al. Distinguishing obstructive from
central sleep apnea events: diaphragm electromyogram and esophageal pressure compared.
Chest. 2009;135(5):1133–41.
Luo YM, Li RF, Jolley C, Wu HD, Steier J, Moxham J, et al. Neural respiratory drive in patients
with COPD during exercise tests. Respiration. 2011;81(4):294–301.
Luo YM, He BT, Wu YX, Yuan H, Xu J, Moxham J, et al. Neural respiratory drive and ventilation
in patients with chronic obstructive pulmonary disease during sleep. Am J Respir Crit Care Med.
2014;190(2):227–9.
Marin JM, Soriano JB, Carrizo SJ, Boldova A, Celli BR. Outcomes in patients with chronic
obstructive pulmonary disease and obstructive sleep apnea: the overlap syndrome. Am J Respir
Crit Care Med. 2010;182(3):325–31.
Morrell MJ, Harty HR, Adams L, Guz A. Breathing during wakefulness and NREM sleep in
humans without an upper airway. J Appl Physiol (1985). 1996;81(1):274–81.
Patout M, Ramsay M, Mackie M, Lhuillier E, Grey N, Arbane G, et al. Home mechanical
ventilation (HMV): setup and outcome in Europe. Eur Respir J. 2015;46:OA4780.
Sanders MH, Newman AB, Haggerty CL, Redline S, Lebowitz M, Samet J, et al. Sleep and sleep-
disordered breathing in adults with predominantly mild obstructive airway disease. Am J Respir
Crit Care Med. 2003;167(1):7–14.
Sinderby C, Spahija J, Beck J, Kaminski D, Yan S, Comtois N, et al. Diaphragm activation during
exercise in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001;163(7):
1637–41.
9 Diaphragm EMG Recording and Its Application in Sleep Medicine 175
Soler X, Gaio E, Powell FL, Ramsdell JW, Loredo JS, Malhotra A, et al. High prevalence of
obstructive sleep apnea in patients with moderate to severe chronic obstructive pulmonary
disease. Ann Am Thorac Soc. 2015;12(8):1219–25.
Stoohs RA, Blum HC, Knaack L, Butsch-von-der-Heydt B, Guilleminault C. Comparison of pleural
pressure and transcutaneous diaphragmatic electromyogram in obstructive sleep apnea syn-
drome. Sleep. 2005;28(3):321–9.
Xiao SC, Lu YR, Guo HX, Qiu ZH, Luo YM. Effect of expiratory load on neural inspiratory drive.
Chin Med J. 2012;125(20):3629–34.
Xiao SC, He BT, Steier J, Moxham J, Polkey MI, Luo YM. Neural respiratory drive and arousal in
patients with obstructive sleep apnea hypopnea. Sleep. 2015;38(6):941–9.
Chapter 10
Chronic Intermittent Hypoxia in Patients
with OSA
Qing Yun Li, Chen Juan Gu, Ying Ni Lin, and Qiong Wang
Abstract Obstructive sleep apnea (OSA) is closely related to the increasing car-
diovascular, metabolic, and cancer morbidities. Chronic intermittent hypoxia (CIH),
which is marked by cyclic episodes of short duration of oxygen desaturation
followed by resaturation during sleep, is a hallmark feature of OSA, and is regarded
as the main mechanism contributing to the clinical consequences of OSA. The
chapter provides an overview of the association between OSA and its comorbidities
including cardiovascular diseases, metabolic diseases, cancer, and cognitive dys-
function, and the possible underlying mechanisms with an emphasis on the role
of CIH.
Abbreviations
AF Atrial fibrillation
AHI Apnea hypopnea index
ANS Autonomic nervous system
AP-1 Activator protein-1
BMI Body mass index
CAD Coronary artery disease
CH Continuous hypoxia
CIH Chronic intermittent hypoxia
CPAP Continuous positive airway pressure
CSC Cancer stem cell
DHF Diastolic heart failure
DNA Deoxyribonucleic acid
© Springer Nature B.V. and Shanghai Jiao Tong University Press 2022 177
A. I. Pack, Q. Y. Li (eds.), Sleep and its Disorders, Translational Medicine Research,
https://doi.org/10.1007/978-94-024-2168-2_10
178 Q. Y. Li et al.
10.1 Introduction
Fig. 10.1 Patient with severe OSA. (A) oxygen desaturation/resaturation cycle is not sinusoidal.
(B) oxygen desaturation/resaturation has considerable variability across the sleep period in patients
with OSA, including different nadirs and periods of normoxia. [Reproduced with permission from
APS 2015: Lim et al. (2015)]
10.2.1.1 Hypertension
ventricular ectopy are more prevalent in patients with OSA than in non-OSA sub-
jects after adjustment for potential confounders (Mehra et al. 2006). OSA patients
with nadir nocturnal SaO2 lower than 60% are at increased risk of developing
ventricular arrhythmias (Shepard et al. 1985). Untreated OSA patients with AF
recurrence show a greater fall in nocturnal SaO2 (Kanagala et al. 2003). Gami
et al. found that the decrease in nocturnal SaO2 was more important than AHI in
predicting the development of AF in OSA patients <65 years old (Gami et al. 2007).
Cadby et al. showed that both AHI and TS90 (time spent with SaO2 below 90%)
were the independent predictors of AF in OSA patients (Cadby et al. 2015). Neither
of these two studies detected an association between arousal index and AF, indicat-
ing that OSA-related CIH rather than sleep fragmentation contributes to the interac-
tion between OSA and AF (Gami et al. 2007; Cadby et al. 2015). CPAP is effective
in preventing OSA-associated arrhythmias (Abe et al. 2010), lowering the likelihood
of recurrence of AF after cardioversion (Kanagala et al. 2003), and reducing
ventricular premature beats during sleep in OSA patients with heart failure
(HF) (Ryan et al. 2005). CPAP prevents the occurrence rather than the recurrence
of AF after cavotricuspid isthmus radiofrequency ablation, indicating a better effi-
cacy of CPAP in patients postablation without preexisting AF (Bazan et al. 2013).
OSA constitutes a major treatable risk factor for coronary artery disease (CAD). The
prevalence of OSA in patients with CAD is about 30% (Peker et al. 1999), while
among hospitalized male patients with acute myocardial infarction (AMI), the
prevalence rises to nearly 70% (Konecny et al. 2010). In a case control study that
recruited men undergoing coronary angiography because of angina pectoris, patients
with angina pectoris presented a significantly higher ODI or AHI and also had a
lower mean value of the lowest SaO2 than controls. Multiple logistic regression
analysis showed that the risk of CAD increased with an increase of ODI or AHI after
adjustment for age, hypertension, diabetes, smoking habits, and BMI, indicating that
sleep-disordered breathing (SDB) with nocturnal desaturations was an independent
predictor of symptomatic CAD (Mooe et al. 1996). It should also be noted that
obesity was associated with more severe SDB and could confound the association
between SDB and CAD in the study. An ODI or AHI in the highest quartile
(ODI 7 events/h or AHI 12 events/h), a history of hypertension, and a 5-U
increase in BMI were related to comparable ORs for CAD (3.6, 4.5, 4.2, and 4.8,
respectively) (Mooe et al. 1996). In another study, an ODI of 5 events/h and an
AHI of 10 events/h have been shown to independently correlate with cerebrovas-
cular events in CAD patients (Mooe et al. 2001). Marin et al. (2005) reported a
higher incidence of fatal cardiovascular events (1.06 per 100 person-years) and
nonfatal cardiovascular events (2.13 per 100 person-years) in patients with severe
OSA than simple snorers, patients with untreated mild-to-moderate OSA, patients
treated with CPAP, and healthy participants over a follow-up period of 10 years.
Multivariate analysis, adjusted for potential confounders, showed that untreated
10 Chronic Intermittent Hypoxia in Patients with OSA 183
severe OSA significantly increased the risk of fatal (OR 2.87) and nonfatal (OR 3.17)
cardiovascular events compared with healthy participants. Episodes of nocturnal
myocardial ischemia, marked by ST-segment depression, are commonly concomi-
tant with apnea hypopnea or desaturation events and/or rapid eye movement (REM)
sleep in patients with OSA and CAD (Schafer et al. 1997; Mooe et al. 2000). Oxygen
desaturation together with age, sleep efficiency, and severity of ischemic heart
disease, but not AHI, are the main factors associated with nocturnal myocardial
ischemia (Peled et al. 1999). CPAP modestly reduces carotid intima-media thickness
and arterial pulse-wave velocity, the early signs of atherosclerosis, in severe OSA
patients (Drager et al. 2007). A 3-month CPAP treatment in severe OSA patients
with CAD effectively improves left ventricular function (Liu et al. 2014). Impor-
tantly, for CAD patients with OSA, treatment for OSA significantly decreases the
risk of cardiovascular death, recurrent myocardial infarction, hospitalization for
heart failure, and coronary revascularization (Milleron et al. 2004; Garcia-Rio
et al. 2013). The role of OSA in leading to cardiovascular events was evaluated in
a randomized trial. The Sleep Apnea Cardiovascular Endpoints (SAVE) trial
enrolled patients with moderate-to-severe OSA and a history of cardiovascular or
cerebrovascular disease, who were randomly assigned to therapeutic CPAP or usual
care and followed for an average of 3.7 years. The primary analysis gave negative
results that CPAP, with a mean duration of only 3.3 h treatment per night, did not
lower the rate of primary end point (death from cardiovascular causes, myocardial
infarction, stroke, or hospitalization for unstable angina, heart failure, or transient
ischemic attack) (hazard ratio with CPAP, 1.10; 95% CI, 0.91 to 1.32; P ¼ 0.34)
(McEvoy et al. 2016). In contrast, the RICCADSA study involving OSA patients
with established CAD showed a significant reduction in cardiovascular risk in those
who used CPAP for 4 h/night compared with those who used CPAP <4 h/night or
did not receive CPAP treatment after a median follow-up of 57 months. This
indicates that good CPAP adherence is required to achieve cardiovascular benefits
in OSA patients (Peker et al. 2016).
The prevalence of OSA in heart failure (HF) ranges from 12 to 53% (Vazir et al.
2007; Yumino et al. 2009). Cross-sectional data from 6424 men and women showed
a 2.20-fold increased odds of having HF associated with OSA after adjustment for
confounders including age, race, sex, smoking status, number of cigarettes smoked
per day, self-reported diabetes, self-reported hypertension, use of antihypertension
medications, systolic blood pressure, BMI, total cholesterol, and high-density lipo-
protein cholesterol (Shahar et al. 2001). SHHS demonstrated that OSA was a
predictor for incident HF in men (adjusted hazard ratio 1.13 per 10-unit increase in
AHI) but not in women. The reason why the authors had less power to detect the
association of OSA with HF in women could be the low prevalence of severe OSA in
women (Gottlieb et al. 2010). Moreover, there is a high prevalence of SDB,
184 Q. Y. Li et al.
including both OSA and CSA, in patients with heart failure with preserved left
ventricular ejection fraction (HFpEF) (Bitter et al. 2009).
CIH is the main contributor to HF in OSA patients. Mean nocturnal SaO2
differentiates between OSA patients with and without right heart failure (RHF).
Additionally, persistent hypoxemia and/or hypercapnia over a 24-h period is a novel
mediator of RHF in OSA patients (Bradley et al. 1985). In a small cohort study with
symptomatic diastolic heart failure (DHF), overnight minimum percentage SaO2, but
not AHI, was associated with more severe diastolic dysfunction (Chan et al. 1997).
Similar results were obtained in a clinic-based study of patients without known heart
diseases, which also identified worsening nocturnal hypoxemia as an independent
indicator for isolated septal growth as well as decreased nitric oxide-mediated
dilation in large arteries (Kraiczi et al. 2001).
Several randomized controlled trials have reported that CPAP treatment signifi-
cantly reverses the decline of left ventricular ejection fraction (LVEF) in HF patients
combined with OSA (Usui et al. 2005; Mansfield et al. 2004). The efficacy of CPAP
has been further confirmed by another study that showed an improvement in stroke
volume and cardiac output after 1-month of CPAP therapy (Kasai et al. 2015).
Additionally, adaptive servo-ventilation (ASV) improves the prognosis of HFpEF
patients with SDB with favorable effects such as improvement of cardiac diastolic
function, and arterial stiffness after a 6-month follow-up (Yoshihisa et al. 2013).
Positive airway pressure (PAP), including both ASV and CPAP, also improves right
heart and pulmonary function, and may reduce all-cause mortality in HFpEF patients
with SDB (Yoshihisa et al. 2015). However, in the Treatment of Sleep-Disordered
Breathing with Predominant Central Sleep Apnea by Adaptive Servo-Ventilation in
Patients with Heart Failure (SERVE-HF) trial, the incidence of the primary end point
[the first event of death from any cause, lifesaving cardiovascular intervention
(cardiac transplantation, implantation of a ventricular assist device, resuscitation
after sudden cardiac arrest, or appropriate lifesaving shock), or unplanned hospital-
ization for worsening heart failure] did not differ significantly between the ASV
group, with a low adherence of average 3.4 h per night, and the control group.
However, all-cause and cardiovascular mortality were both increased with the use of
ASV (Cowie et al. 2015). The explanation of the negative results may be that
attenuating CSA, which is a compensatory mechanism in HF patients, with ASV
may be detrimental. But compared with CSA, OSA causes more adverse cardiac
loading by increasing the left ventricular afterload, which can be reversed by PAP.
Therefore, the SERVE-HF results cannot be simply extrapolated to HF patients
with OSA.
10.2.1.6 Stroke
OSA prevalence in stroke patients ranges from 50 to 70% (Hermann and Bassetti
2009). Cross-sectional analysis revealed that patients with AHI 20 events/h had an
increased risk for stroke (OR ¼ 4.33) compared with those without OSA after
adjustment for known confounding factors (Arzt et al. 2005). In a prospective
10 Chronic Intermittent Hypoxia in Patients with OSA 185
study of patients with CAD over a 10-year follow-up, sleep apnea was presented in
54% of these patients. Patients with an ODI 5 events/h were at greater risk for
stroke (Valham et al. 2008). Wessendorf et al. observed higher plasma levels of
fibrinogen, and average minimal SaO2 was identified as an independent predictor of
fibrinogen levels in stroke patients (Wessendorf et al. 2000). This indicates that
OSA-related hypoxia may mediate the interaction between fibrinogen and OSA in
promoting stroke. Regarding the impact of CPAP on stroke, most studies focus on
CPAP treatment for OSA patients after a stroke. In outpatients with OSA, 2–4 weeks
after a stroke, CPAP treatment resulted in a greater improvement in the depression
score, but not in cognitive or physical function over 4 weeks (Sandberg et al. 2001).
As opposed to the controversial effects of CPAP on neurocognitive outcomes in
stroke patients with OSA (Hsu et al. 2006), several long-term follow-up studies have
shown that CPAP reduces the incidence of cardiovascular events (Martinez-Garcia
et al. 2012), and more importantly, improves the long-term survival in stroke patients
with moderate-to-severe OSA (Parra et al. 2015; Martinez-Garcia et al. 2009).
10.2.2 Mechanisms
The intermediary pathways linking OSA to cardiovascular disease have been eluci-
dated over the last two decades. The main trigger is CIH, which leads to an array of
injurious effects on the heart and vessels through several maladaptive mechanisms,
including oxidative stress, inflammation, vascular endothelial dysfunction, and
IH-induced sympathetic surges.
episodes of intermittent hypoxia provide a substrate that increases the potential for
cardiac arrhythmogenesis. Apneic episodes concomitant with hypoxia stimulate the
carotid body and increase vagal tone, which can lead to bradyarrhythmias and heart
block (Zwillich et al. 1982). The following resumption of breathing with or without
arousals increases sympathetic tone characterized by hyperpnea and may cause
tachyarrhythmias in OSA patients (Rossi et al. 2013).
OSA increases the risk of type 2 diabetes mellitus (T2DM) and a great proportion of
T2DM patients suffer from concomitant OSA (Shaw et al. 2008; Tahrani et al.
2012). In a meta-analysis of six prospective cohort studies including 5953 partici-
pants with follow-up periods of 2.7–16 years, moderate-to-severe OSA was associ-
ated with an increased risk of T2DM (Wang et al. 2013a). An ODI > 5 events/h was
a predictor of developing diabetes (OR ¼ 4.4) during a mean period of 11-year
follow-up (Lindberg et al. 2012). OSA exacerbates the progression of T2DM, as
manifested by difficulty in controlling blood glucose and increased occurrence of
diabetic complications. The adjusted mean glycosylated hemoglobin (HbA1c) was
increased by 1.49%, 1.93%, and 3.69% in patients with mild, moderate, and severe
OSA, respectively (Aronsohn et al. 2010). Concomitant OSA was independently
associated with diabetic peripheral neuropathy and nephropathy (OR ¼ 2.64), and
baseline AHI was an independent predictor of estimated glomerular filtration rate
(eGFR) (Tahrani et al. 2012, 2013).
In addition to T2DM, OSA is reported to be associated with prediabetic state,
including impaired fasting glucose (IFG) and impaired glucose tolerance (IGT). The
prevalence of IFG and IGT is higher in OSA patients (20–67%) (Pamidi and Tasali
2012). Data from the SHHS revealed that in 2588 participants without known
diabetes, OSA subjects had significantly higher adjusted prevalence and adjusted
odds of IFG and IFG plus IGT (Seicean et al. 2008). Insulin resistance (IR), another
important marker of prediabetes, is also found to be independently associated with
OSA. In 118 subjects without diabetes, those with mild, moderate, and severe OSA
displayed a 26.7%, 36.5%, and 43.7% reduction in insulin sensitivity, respectively,
compared with normal subjects, independent of age, sex, race, and percent body fat
(Punjabi and Beamer 2009). In young, lean, and healthy men, who were free of
cardio-metabolic diseases, the presence of mild OSA was associated with a 27%
decrease in insulin sensitivity (Pamidi et al. 2012).
Studies have examined the efficacy of CPAP on glucose metabolism in OSA.
Dawson et al. (2008) reported that an average 41-day CPAP treatment decreased and
stabilized sleeping glucose levels, when they used a continuous glucose monitoring
system to measure interstitial glucose every 5 min during polysomnography (PSG)
in 20 T2DM patients with newly diagnosed OSA. Babu et al. (2005) showed that
188 Q. Y. Li et al.
CPAP treatment (83 50 days) significantly reduced the mean 1-h postprandial
glucose values and HbA1c level. A parallel RCT enrolling OSA patients with T2DM
with HbA1c levels 6.5% showed a decrease in HbA1c level and an improvement
of insulin resistance in the CPAP group compared with those without CPAP. In
CPAP-treated patients, the 6-month change in HbA1c levels was associated with
mean nocturnal oxygen saturation (Martinez-Ceron et al. 2016). Moreover, CPAP
improved insulin sensitivity in severe OSA as assessed by IGT (Weinstock et al.
2012) and nondiabetic patients with moderate-to-severe OSA without changing BMI
(Yang et al. 2013). These findings suggest that OSA is related to impaired glucose
homeostasis independent of obesity, and that CPAP can be an important therapeutic
approach for patients with OSA and impaired glucose metabolism.
However, two RCTs showed that PAP did not influence glycemic control,
including the HbA1c level, fasting plasma glucose, and insulin resistance, but
improved blood pressure (Shaw et al. 2016; Myhill et al. 2012). One of the
explanations could be that the enrolled patients had a mean baseline HbA1c level
of about 7%, which means that the T2DM patients were already relatively well
controlled, and thus, there was limited scope for further improvement with CPAP.
Insulin resistance (IR) and impaired pancreatic β-cell function are the two main
features involved in the pathogenesis of T2DM. Evidence from human and animal
models showed that CIH causes IR in the absence of obesity, while data regarding
the effects of CIH on pancreatic β-cell function is relatively limited.
Several human and animal studies have shown that CIH induces IR (Iiyori et al.
2007; Chen et al. 2010; Louis and Punjabi 2009; Carreras et al. 2012; Polak et al.
2013). However, the potential molecular mechanisms underlying CIH-induced IR
are not fully elucidated. CIH-related activation of sympathetic nerve system, possi-
bly through increased release of catecholamine in adrenal medulla and dysregulation
of insulin pathway in liver, may be involved (Shin et al. 2014a; Mesarwi et al. 2015).
Some studies have shown, however, that CIH decreases the whole-body insulin
sensitivity independent of sympathetic activation (Iiyori et al. 2007; Shin et al.
2014b). Further, CIH may also activate the hypothalamic–pituitary–adrenal axis
and facilitate the release of corticosteroids. Yokoe et al. (2008) revealed that
intermittent hypoxia reversed the normal diurnal blood glucose rhythm and exacer-
bated diurnal peak corticosterone, which was temporally associated with the peak in
blood glucose. Moreover, CIH-induced oxidative stress and inflammation also
partially contribute to IR in OSA patients (Tasali and Ip 2008).
10 Chronic Intermittent Hypoxia in Patients with OSA 189
The effect of CIH on pancreatic β-cells is not well known. Current evidence suggests
the coexistence of protective and injurious pathways in pancreas as a result of
intermittent hypoxia. Intermittent hypoxia promotes both apoptosis and proliferation
of β-cells in mice. The net effect of exposure is increased turnover or increased
replication of pancreatic β-cells (Yokoe et al. 2008; Xu et al. 2009). Similarly, our
group also found that 8-week intermittent hypoxia exposure increased β-cell mass in
lean C57BL/6J mice (Gu et al. 2013).
Regarding the effects of CIH on the function of pancreatic β-cells, a rodent model
of CIH demonstrated that 6-week exposure to intermittent hypoxia impaired the
function of pancreatic β-cells, marked by augmented basal insulin secretion, defective
proinsulin processing, and impaired glucose-stimulated insulin secretion (Shin et al.
2014a). Mitochondrial ROS levels increase in pancreatic β-cells with intermittent
hypoxia, and administration of ROS scavenger reversed the basal insulin secretion
and proinsulin processing (Wang et al. 2013b). Thus, oxidative stress in CIH plays a
role in pancreatic β-cell dysfunction (Wang et al. 2013b). Moreover, in a mouse model
of diabetes mellitus, 14-day exposure to intermittent hypoxia-induced pancreatic
apoptosis and exacerbated dysfunction of pancreatic β-cells, possibly mediated by
the intermittent hypoxia-induced increase in free fatty acids and a shift in composition
of fatty acids toward long-chain saturated fatty acid species (Sherwani et al. 2013).
the severity of OSA (Newman et al. 2001). Another cross-sectional study revealed
that nocturnal hypoxia and OSA severity were independent indicators for higher TG
and lower high-density lipoprotein (HDL) levels after adjustment for confounding
factors (Trzepizur et al. 2013). Furthermore, there is a relationship between OSA and
the progression of non-alcoholic fatty liver disease (NAFLD). In obese adult patients
with NAFLD, OSA is related to elevated alanine aminotransferase (ALT) levels and
a trend toward histologic evidence of progressive liver diseases (Kallwitz et al.
2007). In pediatric NAFLD, OSA is also associated with biochemical, immunohis-
tochemical, and histological features of nonalcoholic steatohepatitis (NASH) and
fibrosis (Nobili et al. 2014). CPAP may be beneficial to improve lipid profile in OSA
patients. A randomized controlled trial including 220 OSA patients demonstrated
that 1-month nasal CPAP produced a decrease in plasma TC by 10.8 mg/dl (Nobili
et al. 2014). However, another study showed that CPAP decreased serum TG levels
only when it was combined with weight loss (Chirinos et al. 2014). A study
examined the effect of CPAP on postprandial lipids and reported a beneficial effect
of CPAP on decreasing TG and TC, which consequently reduced the risk for
cardiovascular events (Phillips et al. 2011). More recently, a meta-regression anal-
ysis of 29 studies including a total of 1958 subjects revealed that CPAP treatment for
OSA seems to improve dyslipidemia, manifested by the decrease in the levels of TC
and low-density lipoprotein (LDL), and the increase in the levels of HDL, without
affecting TG levels (Nadeem et al. 2014).
acid synthesis, suggesting that peripheral lipolysis is the main source of free fatty
acids (FFA) in TG and VLDL (Li et al. 2005). A study in apolipoprotein E-deficient
mice showed an increase in FFA levels, indicating that intermittent hypoxia induces
adipose tissue lipolysis (Jun et al. 2010). The activation of sympathetic nervous
system in response to intermittent hypoxia exposure could potentially induce lipol-
ysis (Lam et al. 2008; Lafontan and Langin 2009; Zechner et al. 2009). The
confluence of increased FFA delivery and impaired β-oxidation may also underlie
the association between OSA and accumulation of fat in the liver and liver injury
(Tanne et al. 2005; Polotsky et al. 2009; Savransky et al. 2007). Thus, CIH may
disrupt lipid metabolism through the interactions of upregulated hepatic lipids
biosynthesis, increased adipose tissue lipolysis, and FFA flux to the liver.
Furthermore, CIH exposure in mice activates NADPH oxidase in liver and
induces oxidative stress. Lipid peroxidation and inflammation may play a role in
intermittent hypoxia-induced progression of NAFLD (Jun et al. 2008). In a mouse
model of diet-induced fatty liver, intermittent hypoxia-induced lobular inflammation
and fibrosis in the liver, converting hepatic steatosis to steatohepatitis. Significant
increases in lipid peroxidation and myeloperoxidase have been observed in liver
with significant increased hepatic levels of pro-inflammatory cytokines IL-1β, IL-6,
and CXC chemokine MIP-2 (Li et al. 2007b).
10.4.2 Mechanisms
The relationship between OSA and cancer has been established by several large
cohort studies (Campos-Rodriguez et al. 2013; Martinez-Garcia et al. 2014a; Nieto
et al. 2012; Kendzerska et al. 2014). Data from the Wisconsin Sleep Cohort showed
that increased cancer mortality correlated with percent nighttime with oxygen
saturation below 90% (TSat90) (Nieto et al. 2012). Two Spanish cohort studies
further confirmed the association between OSA and cancer mortality, and consis-
tently identified TSat90, rather than AHI, as an independent predictor for increased
cancer incident, particularly in patients younger than 65 years (Campos-Rodriguez
et al. 2013; Martinez-Garcia et al. 2014a). In the Canadian cohort study of more than
10,000 patients who had suspected OSA, Kendzerska et al. (2014) found that oxygen
desaturation, but not AHI, was associated with smoking-related cancers, although
they failed to detect a significant relationship between OSA severity and the overall
prevalent or incident cancer in the whole cohort. Another Denmark cohort study also
showed no association between symptoms of SDB and incident cancer, but only
found higher cancer incidence in patients younger than 50 years with high daytime
sleepiness (Christensen et al. 2013). With regard to the cancer locations, studies from
Spain and Canada showed that colorectal, prostate, lung, and breast cancers are most
common in OSA patients (Campos-Rodriguez et al. 2013; Kendzerska et al. 2014).
The American insurance database, however, in nearly 5.6 million individuals
showed the adjusted risks of pancreatic and kidney cancer and melanoma were
significantly higher in patients with OSA, while the risks of colorectal, breast, and
prostate cancers appeared to be lower (Gozal et al. 2016). OSA patients, especially
with insomnia, have been shown to have higher risk of primary brain cancers (Chen
and Hwang 2014). In patients with cutaneous malignant melanoma, both AHI and
ODI are independently associated with an increased melanoma growth rate and other
aggressiveness factors including increased maximum tumor thickness in millimeters
(Breslow index), presence of ulceration and mitotic index (Martinez-Garcia et al.
2014b). Of note, a recent study reported that OSA increased expression of a
disproportionate number of genes mapping to neoplastic pathways in circulating
leukocytes, and more importantly, expression of these genes could be modestly
reduced by effective CPAP treatment (Gharib et al. 2014).
10.5.2 Mechanisms
studies have been conducted to explore the underlying mechanisms of how OSA-like
intermittent hypoxia pattern affects tumor behavior. The proposed hypotheses include
intermittent hypoxia-induced oxidative stress, genomic instability, angiogenesis, and
immunological microenvironment alterations (Kukwa et al. 2015).
Rapid proliferation of cancer cells and abnormal vasculature network results in
heterogeneous hypoxic microenvironments within tumors, which may be responsi-
ble for tumor aggressive behavior and resistance to therapeutic intervention. Cycles
of hypoxia and reoxygenation leads to ROS production and hence may directly
cause DNA damage. On the other hand, hypoxia inhibits multiple DNA repair
pathways, consequently resulting in incomplete or inaccurate DNA replication
(Klein and Glazer 2010). Additionally, increased ROS modifies proteins or lipids
and regulates critical transcription factors in redox-sensitive signaling pathways,
such as HIF-1, NF-κB, and AP-1 (Lavie 2015), leading to self-renewal and apoptotic
imbalance, angiogenesis, and platelet aggregation in tumors. The pattern of
OSA-like intermittent hypoxia in tumor is generally different from that in OSA. In
particular, cycles of hypoxia and reoxygenation in tumors vary from minutes to
hours (Toffoli and Michiels 2008), while intermittent hypoxia in OSA is high-
frequency and could result in one or more hypoxic event per minute. Despite the
difference in the pattern of intermittent hypoxia, nocturnal repetitive blood oxygen
desaturation in OSA could result in relapsing swings in tumor oxygen supply and
exaggerate ROS-related changes in tumors, thereby leading to poor clinical
prognosis.
Studies have shown that OSA-like intermittent hypoxia promotes tumor angio-
genesis in rodent models (Almendros et al. 2012b; Vilaseca et al. 2017; Zhang et al.
2018), which alters hypoxic microenvironment in tumors. Serum levels of vascular
endothelial growth factor (VEGF), a potent angiogenic cytokine, have been found to
be elevated in OSA patients and are strongly associated with the degree of nocturnal
hypoxemia (Schulz et al. 2002; Teramoto et al. 2003). Consistently, increased VEGF
levels were observed in intermittent hypoxia-exposed mice (Almendros et al. 2012b;
Vilaseca et al. 2017; Zhang et al. 2018). In an OSA mouse model bearing kidney
cancer, exposure to intermittent hypoxia increased the endothelial cell contents,
which was positively correlated with serum VEGF levels (Vilaseca et al. 2017).
Additionally, intermittent hypoxia promoted VEGF expression in macrophages, but
not in the kidney adenocarcinoma cells (RENCA cells) or endothelial cells. This
indicates that increased tumor angiogenesis upon exposure to intermittent hypoxia
was more likely due to an increased expression of VEGF by cell populations such as
macrophages rather than directly in tumor cells or endothelial cells (Vilaseca et al.
2017). VEGF expression is regulated by HIF-1, which is a heterodimer composed of
an alpha subunit (HIF-1α) and a beta subunit (HIF-1β). Under hypoxic conditions,
HIF-1α rapidly transfers to the nucleus and binds to HIF-1β, and then the complex
binds to hypoxia response elements, which induces transcription of HIF-1 target
genes. Monocytes from OSA patients have higher levels of HIF-1α and VEGF
compared with those of control subjects and CPAP-treated patients. The mRNA
levels of HIF-1α and VEGF are positively correlated with nighttime oxygen satura-
tion levels <90% (CT90) (Cubillos-Zapata et al. 2018). Knockdown of HIF-1α
10 Chronic Intermittent Hypoxia in Patients with OSA 195
References
Almendros I, Montserrat JM, Torres M, Bonsignore MR, Chimenti L, Navajas D, Farre R. Obesity
and intermittent hypoxia increase tumor growth in a mouse model of sleep apnea. Sleep Med.
2012b;13(10):1254–60.
Almendros I, Montserrat JM, Torres M, Dalmases M, Cabanas ML, Campos-Rodriguez F,
Navajas D, Farre R. Intermittent hypoxia increases melanoma metastasis to the lung in a
mouse model of sleep apnea. Respir Physiol Neurobiol. 2013;186(3):303–7.
Almendros I, Wang Y, Becker L, Lennon FE, Zheng J, Coats BR, Schoenfelt KS, Carreras A,
Hakim F, Zhang SX, et al. Intermittent hypoxia-induced changes in tumor-associated macro-
phages and tumor malignancy in a mouse model of sleep apnea. Am J Respir Crit Care Med.
2014;189(5):593–601.
Almendros I, Gileles-Hillel A, Khalyfa A, Wang Y, Zhang SX, Carreras A, Farre R, Gozal
D. Adipose tissue macrophage polarization by intermittent hypoxia in a mouse model
of OSA: effect of tumor microenvironment. Cancer Lett. 2015;361(2):233–9.
Almendros I, Khalyfa A, Trzepizur W, Gileles-Hillel A, Huang L, Akbarpour M, Andrade J,
Farre R, Gozal D. Tumor cell malignant properties are enhanced by circulating exosomes in
sleep apnea. Chest. 2016;150(5):1030–41.
Antic NA, Catcheside P, Buchan C, Hensley M, Naughton MT, Rowland S, Williamson B,
Windler S, McEvoy RD. The effect of CPAP in normalizing daytime sleepiness, quality of
life, and neurocognitive function in patients with moderate to severe OSA. Sleep. 2011;34(1):
111–9.
Arias MA, Garcia-Rio F, Alonso-Fernandez A, Martinez I, Villamor J. Pulmonary hypertension in
obstructive sleep apnoea: effects of continuous positive airway pressure: a randomized, con-
trolled cross-over study. Eur Heart J. 2006;27(9):1106–13.
Aronsohn RS, Whitmore H, Van Cauter E, Tasali E. Impact of untreated obstructive sleep apnea on
glucose control in type 2 diabetes. Am J Respir Crit Care Med. 2010;181(5):507–13.
Arzt M, Young T, Finn L, Skatrud JB, Bradley TD. Association of sleep-disordered breathing and
the occurrence of stroke. Am J Respir Crit Care Med. 2005;172(11):1447–51.
Azmi AS, Bao B, Sarkar FH. Exosomes in cancer development, metastasis, and drug resistance: a
comprehensive review. Cancer Metastasis Rev. 2013;32(3–4):623–42.
Babu AR, Herdegen J, Fogelfeld L, Shott S, Mazzone T. Type 2 diabetes, glycemic control, and
continuous positive airway pressure in obstructive sleep apnea. Arch Intern Med. 2005;165(4):
447–52.
Badran M, Ayas N, Laher I. Cardiovascular complications of sleep apnea: role of oxidative stress.
Oxidative Med Cell Longev. 2014;2014:985258.
Bazan V, Grau N, Valles E, Felez M, Sanjuas C, Cainzos-Achirica M, Benito B, Jauregui-
Abularach M, Gea J, Bruguera-Cortada J, et al. Obstructive sleep apnea in patients with typical
atrial flutter: prevalence and impact on arrhythmia control outcome. Chest. 2013;143(5):
1277–83.
Bedard MA, Montplaisir J, Malo J, Richer F, Rouleau I. Persistent neuropsychological deficits and
vigilance impairment in sleep apnea syndrome after treatment with continuous positive airways
pressure (CPAP). J Clin Exp Neuropsychol. 1993;15(2):330–41.
Beebe DW, Groesz L, Wells C, Nichols A, McGee K. The neuropsychological effects of obstructive
sleep apnea: a meta-analysis of norm-referenced and case-controlled data. Sleep. 2003;26(3):
298–307.
Bitter T, Faber L, Hering D, Langer C, Horstkotte D, Oldenburg O. Sleep-disordered breathing in
heart failure with normal left ventricular ejection fraction. Eur J Heart Fail. 2009;11(6):602–8.
Bradley TD, Floras JS. Obstructive sleep apnoea and its cardiovascular consequences. Lancet.
2009;373(9657):82–93.
Bradley TD, Rutherford R, Grossman RF, Lue F, Zamel N, Moldofsky H, Phillipson EA. Role of
daytime hypoxemia in the pathogenesis of right heart failure in the obstructive sleep apnea
syndrome. Am Rev Respir Dis. 1985;131(6):835–9.
Bucks RS, Olaithe M, Eastwood P. Neurocognitive function in obstructive sleep apnoea: a meta-
review. Respirology. 2013;18(1):61–70.
198 Q. Y. Li et al.
Cadby G, McArdle N, Briffa T, Hillman DR, Simpson L, Knuiman M, Hung J. Severity of OSA is
an independent predictor of incident atrial fibrillation hospitalization in a large sleep-clinic
cohort. Chest. 2015;148(4):945–52.
Campen MJ, Shimoda LA, O’Donnell CP. Acute and chronic cardiovascular effects of intermittent
hypoxia in C57BL/6J mice. J Appl Physiol (1985). 2005;99(5):2028–35.
Campillo N, Torres M, Vilaseca A, Nonaka PN, Gozal D, Roca-Ferrer J, Picado C, Montserrat JM,
Farre R, Navajas D, et al. Role of Cyclooxygenase-2 on intermittent hypoxia-induced lung
tumor malignancy in a mouse model of sleep apnea. Sci Rep. 2017;7(44693):44693.
Campos-Rodriguez F, Martinez-Garcia MA, Martinez M, Duran-Cantolla J, Pena Mde L, Masdeu
MJ, Gonzalez M, Campo F, Gallego I, Marin JM, et al. Association between obstructive sleep
apnea and cancer incidence in a large multicenter Spanish cohort. Am J Respir Crit Care Med.
2013;187(1):99–105.
Canessa N, Castronovo V, Cappa SF, Aloia MS, Marelli S, Falini A, Alemanno F, Ferini-Strambi
L. Obstructive sleep apnea: brain structural changes and neurocognitive function before and
after treatment. Am J Respir Crit Care Med. 2011;183(10):1419–26.
Capone C, Faraco G, Coleman C, Young CN, Pickel VM, Anrather J, Davisson RL, Iadecola
C. Endothelin 1-dependent neurovascular dysfunction in chronic intermittent hypoxia. Hyper-
tension. 2012;60(1):106–13.
Carreras A, Kayali F, Zhang J, Hirotsu C, Wang Y, Gozal D. Metabolic effects of intermittent
hypoxia in mice: steady versus high-frequency applied hypoxia daily during the rest period. Am
J Physiol Regul Integr Comp Physiol. 2012;303(7):R700–9.
Chan J, Sanderson J, Chan W, Lai C, Choy D, Ho A, Leung R. Prevalence of sleep-disordered
breathing in diastolic heart failure. Chest. 1997;111(6):1488–93.
Chen JC, Hwang JH. Sleep apnea increased incidence of primary central nervous system cancers: a
nationwide cohort study. Sleep Med. 2014;15(7):749–54.
Chen EP, Smyth EM. COX-2 and PGE2-dependent immunomodulation in breast cancer. Prosta-
glandins Other Lipid Mediat. 2011;96(1–4):14–20.
Chen L, Einbinder E, Zhang Q, Hasday J, Balke CW, Scharf SM. Oxidative stress and left
ventricular function with chronic intermittent hypoxia in rats. Am J Respir Crit Care Med.
2005;172(7):915–20.
Chen L, Cao ZL, Han F, Gao ZC, He QY. Chronic intermittent hypoxia from pedo-stage decreases
glucose transporter 4 expression in adipose tissue and causes insulin resistance. Chin Med
J. 2010;123(4):463–70.
Chirinos JA, Gurubhagavatula I, Teff K, Rader DJ, Wadden TA, Townsend R, Foster GD,
Maislin G, Saif H, Broderick P, et al. CPAP, weight loss, or both for obstructive sleep apnea.
N Engl J Med. 2014;370(24):2265–75.
Christensen AS, Clark A, Salo P, Nymann P, Lange P, Prescott E, Rod NH. Symptoms of sleep
disordered breathing and risk of cancer: a prospective cohort study. Sleep. 2013;36(10):
1429–35.
Cowie MR, Woehrle H, Wegscheider K, Angermann C, d’Ortho MP, Erdmann E, Levy P, Simonds
AK, Somers VK, Zannad F, et al. Adaptive servo-ventilation for central sleep apnea in systolic
heart failure. N Engl J Med. 2015;373(12):1095–105.
Cubillos-Zapata C, Hernandez-Jimenez E, Avendano-Ortiz J, Toledano V, Varela-Serrano A,
Fernandez-Navarro I, Casitas R, Carpio C, Aguirre LA, Garcia-Rio F, et al. Obstructive sleep
apnea monocytes exhibit high levels of vascular endothelial growth factor secretion,
augmenting tumor progression. Mediat Inflamm. 2018;2018(7373921):7373921.
Dalmases M, Sole-Padulles C, Torres M, Embid C, Nunez MD, Martinez-Garcia MA, Farre R,
Bargallo N, Bartres-Faz D, Montserrat JM. Effect of CPAP on cognition, brain function, and
structure among elderly patients with OSA: a randomized pilot study. Chest. 2015;148(5):
1214–23.
Dawson A, Abel SL, Loving RT, Dailey G, Shadan FF, Cronin JW, Kripke DF, Kline LE. CPAP
therapy of obstructive sleep apnea in type 2 diabetics improves glycemic control during sleep. J
Clin Sleep Med. 2008;4(6):538–42.
10 Chronic Intermittent Hypoxia in Patients with OSA 199
Dewan NA, Nieto FJ, Somers VK. Intermittent hypoxemia and OSA: implications for
comorbidities. Chest. 2015;147(1):266–74.
Drager LF, Bortolotto LA, Figueiredo AC, Krieger EM, Lorenzi GF. Effects of continuous positive
airway pressure on early signs of atherosclerosis in obstructive sleep apnea. Am J Respir Crit
Care Med. 2007;176(7):706–12.
Drager LF, Polotsky VY, Lorenzi-Filho G. Obstructive sleep apnea: an emerging risk factor for
atherosclerosis. Chest. 2011;140(2):534–42.
Ferini-Strambi L, Baietto C, Di Gioia MR, Castaldi P, Castronovo C, Zucconi M, Cappa
SF. Cognitive dysfunction in patients with obstructive sleep apnea (OSA): partial reversibility
after continuous positive airway pressure (CPAP). Brain Res Bull. 2003;61(1):87–92.
Findley LJ, Barth JT, Powers DC, Wilhoit SC, Boyd DG, Suratt PM. Cognitive impairment in
patients with obstructive sleep apnea and associated hypoxemia. Chest. 1986;90(5):686–90.
Gale SD, Hopkins RO. Effects of hypoxia on the brain: neuroimaging and neuropsychological
findings following carbon monoxide poisoning and obstructive sleep apnea. J Int Neuropsychol
Soc. 2004;10(1):60–71.
Gallego-Martin T, Farre R, Almendros I, Gonzalez-Obeso E, Obeso A. Chronic intermittent
hypoxia mimicking sleep apnoea increases spontaneous tumorigenesis in mice. Eur Respir
J. 2017;49:1602111.
Gami AS, Hodge DO, Herges RM, Olson EJ, Nykodym J, Kara T, Somers VK. Obstructive sleep
apnea, obesity, and the risk of incident atrial fibrillation. J Am Coll Cardiol. 2007;49(5):565–71.
Garcia-Rio F, Alonso-Fernandez A, Armada E, Mediano O, Lores V, Rojo B, Fernandez-Lahera J,
Fernandez-Navarro I, Carpio C, Ramirez T. CPAP effect on recurrent episodes in patients with
sleep apnea and myocardial infarction. Int J Cardiol. 2013;168(2):1328–35.
Gharib SA, Seiger AN, Hayes AL, Mehra R, Patel SR. Treatment of obstructive sleep apnea alters
cancer-associated transcriptional signatures in circulating leukocytes. Sleep. 2014;37(4):
709–714, 714A–T.
Gottlieb DJ, Yenokyan G, Newman AB, O’Connor GT, Punjabi NM, Quan SF, Redline S, Resnick
HE, Tong EK, Diener-West M, et al. Prospective study of obstructive sleep apnea and incident
coronary heart disease and heart failure: the sleep heart health study. Circulation. 2010;122(4):
352–60.
Gozal D, Daniel JM, Dohanich GP. Behavioral and anatomical correlates of chronic episodic
hypoxia during sleep in the rat. J Neurosci. 2001;21(7):2442–50.
Gozal D, Ham SA, Mokhlesi B. Sleep apnea and cancer: analysis of a nationwide population
sample. Sleep. 2016;39(8):1493–500.
Gu CJ, Li M, Li QY, Li N. Chronic intermittent hypoxia increases beta cell mass and activates the
mammalian target of rapamycin/hypoxia inducible factor 1/vascular endothelial growth factor A
pathway in mice pancreatic islet. Chin Med J. 2013;126(12):2368–73.
Guo C, Buranych A, Sarkar D, Fisher PB, Wang XY. The role of tumor-associated macrophages in
tumor vascularization. Vasc Cell. 2013;5(1):20.
Haas DC, Foster GL, Nieto FJ, Redline S, Resnick HE, Robbins JA, Young T, Pickering
TG. Age-dependent associations between sleep-disordered breathing and hypertension: impor-
tance of discriminating between systolic/diastolic hypertension and isolated systolic hyperten-
sion in the Sleep Heart Health Study. Circulation. 2005;111(5):614–21.
Hayashi T, Yamashita C, Matsumoto C, Kwak CJ, Fujii K, Hirata T, Miyamura M, Mori T,
Ukimura A, Okada Y, et al. Role of gp91phox-containing NADPH oxidase in left ventricular
remodeling induced by intermittent hypoxic stress. Am J Phys Heart Circ Phys. 2008;294(5):
H2197–203.
Hedner JA, Wilcox I, Laks L, Grunstein RR, Sullivan CE. A specific and potent pressor effect of
hypoxia in patients with sleep apnea. Am Rev Respir Dis. 1992;146(5 Pt 1):1240–5.
Hermann DM, Bassetti CL. Sleep-related breathing and sleep-wake disturbances in ischemic stroke.
Neurology. 2009;73(16):1313–22.
200 Q. Y. Li et al.
Hsu CY, Vennelle M, Li HY, Engleman HM, Dennis MS, Douglas NJ. Sleep-disordered breathing
after stroke: a randomised controlled trial of continuous positive airway pressure. J Neurol
Neurosurg Psychiatry. 2006;77(10):1143–9.
Iiyori N, Alonso LC, Li J, Sanders MH, Garcia-Ocana A, O’Doherty RM, Polotsky VY, O’Donnell
CP. Intermittent hypoxia causes insulin resistance in lean mice independent of autonomic
activity. Am J Respir Crit Care Med. 2007;175(8):851–7.
Ip MS, Lam B, Chan LY, Zheng L, Tsang KW, Fung PC, Lam WK. Circulating nitric oxide is
suppressed in obstructive sleep apnea and is reversed by nasal continuous positive airway
pressure. Am J Respir Crit Care Med. 2000;162(6):2166–71.
Jackman KA, Zhou P, Faraco G, Peixoto PM, Coleman C, Voss HU, Pickel V, Manfredi G,
Iadecola C. Dichotomous effects of chronic intermittent hypoxia on focal cerebral ischemic
injury. Stroke. 2014;45(5):1460–7.
Jagadapillai R, Mellen NM, Sachleben LR Jr, Gozal E. Ceftriaxone preserves glutamate trans-
porters and prevents intermittent hypoxia-induced vulnerability to brain excitotoxic injury.
PLoS One. 2014;9(7):e100230.
Jun J, Savransky V, Nanayakkara A, Bevans S, Li J, Smith PL, Polotsky VY. Intermittent hypoxia
has organ-specific effects on oxidative stress. Am J Physiol Regul Integr Comp Physiol.
2008;295(4):R1274–81.
Jun J, Reinke C, Bedja D, Berkowitz D, Bevans-Fonti S, Li J, Barouch LA, Gabrielson K, Polotsky
VY. Effect of intermittent hypoxia on atherosclerosis in apolipoprotein E-deficient mice.
Atherosclerosis. 2010;209(2):381–6.
Kallwitz ER, Herdegen J, Madura J, Jakate S, Cotler SJ. Liver enzymes and histology in obese
patients with obstructive sleep apnea. J Clin Gastroenterol. 2007;41(10):918–21.
Kanagala R, Murali NS, Friedman PA, Ammash NM, Gersh BJ, Ballman KV, Shamsuzzaman AS,
Somers VK. Obstructive sleep apnea and the recurrence of atrial fibrillation. Circulation.
2003;107(20):2589–94.
Kanagy NL, Walker BR, Nelin LD. Role of endothelin in intermittent hypoxia-induced hyperten-
sion. Hypertension. 2001;37(2 Pt 2):511–5.
Kasai T, Floras JS, Bradley TD. Sleep apnea and cardiovascular disease: a bidirectional relation-
ship. Circulation. 2012;126(12):1495–510.
Kasai T, Yumino D, Redolfi S, Su MC, Ruttanaumpawan P, Mak S, Newton GE, Floras JS, Bradley
TD. Overnight effects of obstructive sleep apnea and its treatment on stroke volume in patients
with heart failure. Can J Cardiol. 2015;31(7):832–8.
Kato M, Roberts-Thomson P, Phillips BG, Haynes WG, Winnicki M, Accurso V, Somers
VK. Impairment of endothelium-dependent vasodilation of resistance vessels in patients with
obstructive sleep apnea. Circulation. 2000;102(21):2607–10.
Kendzerska T, Leung RS, Hawker G, Tomlinson G, Gershon AS. Obstructive sleep apnea and the
prevalence and incidence of cancer. CMAJ. 2014;186(13):985–92.
Kent BD, Grote L, Ryan S, Pepin JL, Bonsignore MR, Tkacova R, Saaresranta T, Verbraecken J,
Levy P, Hedner J, et al. Diabetes mellitus prevalence and control in sleep-disordered breathing:
the European Sleep Apnea Cohort (ESADA) study. Chest. 2014;146(4):982–90.
Klein TJ, Glazer PM. The tumor microenvironment and DNA repair. Semin Radiat Oncol. 2010;20
(4):282–7.
Konecny T, Kuniyoshi FH, Orban M, Pressman GS, Kara T, Gami A, Caples SM, Lopez-Jimenez F,
Somers VK. Under-diagnosis of sleep apnea in patients after acute myocardial infarction. J Am
Coll Cardiol. 2010;56(9):742–3.
Kotterba S, Rasche K, Widdig W, Duscha C, Blombach S, Schultze-Werninghaus G, Malin
JP. Neuropsychological investigations and event-related potentials in obstructive sleep apnea
syndrome before and during CPAP-therapy. J Neurol Sci. 1998;159(1):45–50.
Kraiczi H, Caidahl K, Samuelsson A, Peker Y, Hedner J. Impairment of vascular endothelial
function and left ventricular filling: association with the severity of apnea-induced hypoxemia
during sleep. Chest. 2001;119(4):1085–91.
10 Chronic Intermittent Hypoxia in Patients with OSA 201
Kukwa W, Migacz E, Druc K, Grzesiuk E, Czarnecka AM. Obstructive sleep apnea and cancer:
effects of intermittent hypoxia? Future Oncol. 2015;11(24):3285–98.
Kushida CA, Nichols DA, Holmes TH, Quan SF, Walsh JK, Gottlieb DJ, Simon RD Jr,
Guilleminault C, White DP, Goodwin JL, et al. Effects of continuous positive airway pressure
on neurocognitive function in obstructive sleep apnea patients: the Apnea Positive Pressure
Long-term Efficacy Study (APPLES). Sleep. 2012;35(12):1593–602.
Lafontan M, Langin D. Lipolysis and lipid mobilization in human adipose tissue. Prog Lipid Res.
2009;48(5):275–97.
Lam JC, Xu A, Tam S, Khong PI, Yao TJ, Lam DC, Lai AY, Lam B, Lam KS, Mary
SM. Hypoadiponectinemia is related to sympathetic activation and severity of obstructive
sleep apnea. Sleep. 2008;31(12):1721–7.
Lavie L. Oxidative stress in obstructive sleep apnea and intermittent hypoxia—revisited—the bad
ugly and good: implications to the heart and brain. Sleep Med Rev. 2015;20:27–45.
Lavie L, Dyugovskaya L, Polyakov A. Biology of peripheral blood cells in obstructive sleep
apnea—the tip of the iceberg. Arch Physiol Biochem. 2008;114(4):244–54.
Lefebvre B, Godin-Ribuot D, Joyeux-Faure M, Caron F, Bessard G, Levy P, Stanke-Labesque
F. Functional assessment of vascular reactivity after chronic intermittent hypoxia in the rat.
Respir Physiol Neurobiol. 2006;150(2–3):278–86.
Leuenberger U, Jacob E, Sweer L, Waravdekar N, Zwillich C, Sinoway L. Surges of muscle
sympathetic nerve activity during obstructive apnea are linked to hypoxemia. J Appl Physiol
(1985). 1995;79(2):581–8.
Levy P, Pepin JL, Arnaud C, Baguet JP, Dematteis M, Mach F. Obstructive sleep apnea and
atherosclerosis. Prog Cardiovasc Dis. 2009;51(5):400–10.
Li J, Thorne LN, Punjabi NM, Sun CK, Schwartz AR, Smith PL, Marino RL, Rodriguez A,
Hubbard WC, O’Donnell CP, et al. Intermittent hypoxia induces hyperlipidemia in lean mice.
Circ Res. 2005;97(7):698–706.
Li J, Bosch-Marce M, Nanayakkara A, Savransky V, Fried SK, Semenza GL, Polotsky VY. Altered
metabolic responses to intermittent hypoxia in mice with partial deficiency of hypoxia-inducible
factor-1alpha. Physiol Genomics. 2006;25(3):450–7.
Li J, Savransky V, Nanayakkara A, Smith PL, O’Donnell CP, Polotsky VY. Hyperlipidemia and
lipid peroxidation are dependent on the severity of chronic intermittent hypoxia. J Appl Physiol
(1985). 2007a;102(2):557–63.
Li J, Nanayakkara A, Jun J, Savransky V, Polotsky VY. Effect of deficiency in SREBP cleavage-
activating protein on lipid metabolism during intermittent hypoxia. Physiol Genomics.
2007b;31(2):273–80.
Lim DC, Pack AI. Obstructive sleep apnea and cognitive impairment: addressing the blood-brain
barrier. Sleep Med Rev. 2014;18(1):35–48.
Lim DC, Brady DC, Po P, Chuang LP, Marcondes L, Kim EY, Keenan BT, Guo X, Maislin G,
Galante RJ, et al. Simulating obstructive sleep apnea patients’ oxygenation characteristics into a
mouse model of cyclical intermittent hypoxia. J Appl Physiol (1985). 2015;118(5):544–57.
Lindberg E, Theorell-Haglow J, Svensson M, Gislason T, Berne C, Janson C. Sleep apnea and
glucose metabolism: a long-term follow-up in a community-based sample. Chest. 2012;142(4):
935–42.
Liu JN, Zhang JX, Lu G, Qiu Y, Yang D, Yin GY, Zhang XL. The effect of oxidative stress in
myocardial cell injury in mice exposed to chronic intermittent hypoxia. Chin Med J. 2010;123
(1):74–8.
Liu X, Feng L, Cao G, Huang H, Xu Q, Yu J, Zhang S, Zhou M. Cardiac structure and function
improvements in coronary artery disease combined with severe obstructive sleep apnea/
hypopnea syndrome patients via noninvasive positive pressure ventilation therapy. Coron
Artery Dis. 2014;25(6):516–20.
Logan AG, Tkacova R, Perlikowski SM, Leung RS, Tisler A, Floras JS, Bradley TD. Refractory
hypertension and sleep apnoea: effect of CPAP on blood pressure and baroreflex. Eur Respir
J. 2003;21(2):241–7.
202 Q. Y. Li et al.
Louis M, Punjabi NM. Effects of acute intermittent hypoxia on glucose metabolism in awake
healthy volunteers. J Appl Physiol (1985). 2009;106(5):1538–44.
Magalang UJ, Cruff JP, Rajappan R, Hunter MG, Patel T, Marsh CB, Raman SV, Parinandi
NL. Intermittent hypoxia suppresses adiponectin secretion by adipocytes. Experimental Clin
Endocrinol Diabetes. 2009;117(3):129–34.
Mansfield DR, Gollogly NC, Kaye DM, Richardson M, Bergin P, Naughton MT. Controlled trial of
continuous positive airway pressure in obstructive sleep apnea and heart failure. Am J Respir
Crit Care Med. 2004;169(3):361–6.
Marin JM, Carrizo SJ, Vicente E, Agusti AG. Long-term cardiovascular outcomes in men with
obstructive sleep apnoea-hypopnoea with or without treatment with continuous positive airway
pressure: an observational study. Lancet. 2005;365(9464):1046–53.
Martinez-Ceron E, Barquiel B, Bezos AM, Casitas R, Galera R, Garcia-Benito C, Hernanz A,
Alonso-Fernandez A, Garcia-Rio F. Effect of continuous positive airway pressure on glycemic
control in patients with obstructive sleep apnea and type 2 diabetes. A Randomized Clinical
Trial. Am J Respir Crit Care Med. 2016;194(4):476–85.
Martinez-Garcia MA, Soler-Cataluna JJ, Ejarque-Martinez L, Soriano Y, Roman-Sanchez P, Illa
FB, Canal JM, Duran-Cantolla J. Continuous positive airway pressure treatment reduces
mortality in patients with ischemic stroke and obstructive sleep apnea: a 5-year follow-up
study. Am J Respir Crit Care Med. 2009;180(1):36–41.
Martinez-Garcia MA, Campos-Rodriguez F, Soler-Cataluna JJ, Catalan-Serra P, Roman-Sanchez P,
Montserrat JM. Increased incidence of nonfatal cardiovascular events in stroke patients with
sleep apnoea: effect of CPAP treatment. Eur Respir J. 2012;39(4):906–12.
Martinez-Garcia MA, Campos-Rodriguez F, Duran-Cantolla J, de la Pena M, Masdeu MJ,
Gonzalez M, Del Campo F, Serra PC, Valero-Sanchez I, Ferrer MJ, et al. Obstructive sleep
apnea is associated with cancer mortality in younger patients. Sleep Med. 2014a;15(7):742–8.
Martinez-Garcia MA, Martorell-Calatayud A, Nagore E, Valero I, Selma MJ, Chiner E, Landete P,
Montserrat JM, Carrera C, Perez-Gil A, et al. Association between sleep disordered breathing
and aggressiveness markers of malignant cutaneous melanoma. Eur Respir J. 2014b;43(6):
1661–8.
McEvoy RD, Antic NA, Heeley E, Luo Y, Ou Q, Zhang X, Mediano O, Chen R, Drager LF, Liu Z,
et al. CPAP for prevention of cardiovascular events in obstructive sleep apnea. N Engl J Med.
2016;375(10):919–31.
Mehra R, Benjamin EJ, Shahar E, Gottlieb DJ, Nawabit R, Kirchner HL, Sahadevan J, Redline
S. Association of nocturnal arrhythmias with sleep-disordered breathing: the Sleep Heart Health
Study. Am J Respir Crit Care Med. 2006;173(8):910–6.
Mesarwi OA, Sharma EV, Jun JC, Polotsky VY. Metabolic dysfunction in obstructive sleep apnea:
a critical examination of underlying mechanisms. Sleep Biol Rhythms. 2015;13(1):2–17.
Milleron O, Pilliere R, Foucher A, de Roquefeuil F, Aegerter P, Jondeau G, Raffestin BG, Dubourg
O. Benefits of obstructive sleep apnoea treatment in coronary artery disease: a long-term follow-
up study. Eur Heart J. 2004;25(9):728–34.
Mooe T, Rabben T, Wiklund U, Franklin KA, Eriksson P. Sleep-disordered breathing in men with
coronary artery disease. Chest. 1996;109(3):659–63.
Mooe T, Franklin KA, Wiklund U, Rabben T, Holmstrom K. Sleep-disordered breathing and
myocardial ischemia in patients with coronary artery disease. Chest. 2000;117(6):1597–602.
Mooe T, Franklin KA, Holmstrom K, Rabben T, Wiklund U. Sleep-disordered breathing and
coronary artery disease: long-term prognosis. Am J Respir Crit Care Med. 2001;164(10 Pt 1):
1910–3.
Myhill PC, Davis WA, Peters KE, Chubb SA, Hillman D, Davis TM. Effect of continuous positive
airway pressure therapy on cardiovascular risk factors in patients with type 2 diabetes and
obstructive sleep apnea. J Clin Endocrinol Metab. 2012;97(11):4212–8.
Nadeem R, Singh M, Nida M, Kwon S, Sajid H, Witkowski J, Pahomov E, Shah K, Park W,
Champeau D. Effect of CPAP treatment for obstructive sleep apnea hypopnea syndrome on lipid
profile: a meta-regression analysis. J Clin Sleep Med. 2014;10(12):1295–302.
10 Chronic Intermittent Hypoxia in Patients with OSA 203
Naegele B, Pepin JL, Levy P, Bonnet C, Pellat J, Feuerstein C. Cognitive executive dysfunction in
patients with obstructive sleep apnea syndrome (OSAS) after CPAP treatment. Sleep. 1998;21
(4):392–7.
Nahmias J, Lao R, Karetzky M. Right ventricular dysfunction in obstructive sleep apnoea: reversal
with nasal continuous positive airway pressure. Eur Respir J. 1996;9(5):945–51.
Nanduri J, Yuan G, Kumar GK, Semenza GL, Prabhakar NR. Transcriptional responses to
intermittent hypoxia. Respir Physiol Neurobiol. 2008;164(1–2):277–81.
Newman AB, Nieto FJ, Guidry U, Lind BK, Redline S, Pickering TG, Quan SF. Relation of sleep-
disordered breathing to cardiovascular disease risk factors: the Sleep Heart Health Study. Am J
Epidemiol. 2001;154(1):50–9.
Nieto FJ, Young TB, Lind BK, Shahar E, Samet JM, Redline S, D’Agostino RB, Newman AB,
Lebowitz MD, Pickering TG. Association of sleep-disordered breathing, sleep apnea, and
hypertension in a large community-based study. Sleep Heart Health Study. JAMA. 2000;283
(14):1829–36.
Nieto FJ, Peppard PE, Young T, Finn L, Hla KM, Farre R. Sleep-disordered breathing and cancer
mortality: results from the Wisconsin Sleep Cohort Study. Am J Respir Crit Care Med.
2012;186(2):190–4.
Nisbet RE, Graves AS, Kleinhenz DJ, Rupnow HL, Reed AL, Fan TH, Mitchell PO, Sutliff RL,
Hart CM. The role of NADPH oxidase in chronic intermittent hypoxia-induced pulmonary
hypertension in mice. Am J Respir Cell Mol Biol. 2009;40(5):601–9.
Nobili V, Cutrera R, Liccardo D, Pavone M, Devito R, Giorgio V, Verrillo E, Baviera G, Musso
G. Obstructive sleep apnea syndrome affects liver histology and inflammatory cell activation in
pediatric nonalcoholic fatty liver disease, regardless of obesity/insulin resistance. Am J Respir
Crit Care Med. 2014;189(1):66–76.
Pack AI, Gislason T. Obstructive sleep apnea and cardiovascular disease: a perspective and future
directions. Prog Cardiovasc Dis. 2009;51(5):434–51.
Pamidi S, Tasali E. Obstructive sleep apnea and type 2 diabetes: is there a link? Front Neurol.
2012;3:126.
Pamidi S, Wroblewski K, Broussard J, Day A, Hanlon EC, Abraham V, Tasali E. Obstructive sleep
apnea in young lean men: impact on insulin sensitivity and secretion. Diabetes Care. 2012;35
(11):2384–9.
Parra O, Sanchez-Armengol A, Capote F, Bonnin M, Arboix A, Campos-Rodriguez F, Perez-
Ronchel J, Duran-Cantolla J, Martinez-Null C, de la Pena M, et al. Efficacy of continuous
positive airway pressure treatment on 5-year survival in patients with ischaemic stroke and
obstructive sleep apnea: a randomized controlled trial. J Sleep Res. 2015;24(1):47–53.
Payne RS, Goldbart A, Gozal D, Schurr A. Effect of intermittent hypoxia on long-term potentiation
in rat hippocampal slices. Brain Res. 2004;1029(2):195–9.
Peker Y, Kraiczi H, Hedner J, Loth S, Johansson A, Bende M. An independent association between
obstructive sleep apnoea and coronary artery disease. Eur Respir J. 1999;14(1):179–84.
Peker Y, Glantz H, Eulenburg C, Wegscheider K, Herlitz J, Thunstrom E. Effect of positive airway
pressure on cardiovascular outcomes in coronary artery disease patients with nonsleepy obstruc-
tive sleep apnea. The RICCADSA Randomized Controlled Trial. Am J Respir Crit Care Med.
2016;194(5):613–20.
Peled N, Abinader EG, Pillar G, Sharif D, Lavie P. Nocturnal ischemic events in patients with
obstructive sleep apnea syndrome and ischemic heart disease: effects of continuous positive air
pressure treatment. J Am Coll Cardiol. 1999;34(6):1744–9.
Peng YJ, Yuan G, Ramakrishnan D, Sharma SD, Bosch-Marce M, Kumar GK, Semenza GL,
Prabhakar NR. Heterozygous HIF-1alpha deficiency impairs carotid body-mediated systemic
responses and reactive oxygen species generation in mice exposed to intermittent hypoxia. J
Physiol. 2006;577(Pt 2):705–16.
Peppard PE, Young T, Palta M, Skatrud J. Prospective study of the association between sleep-
disordered breathing and hypertension. N Engl J Med. 2000;342(19):1378–84.
204 Q. Y. Li et al.
Phillips BG, Narkiewicz K, Pesek CA, Haynes WG, Dyken ME, Somers VK. Effects of obstructive
sleep apnea on endothelin-1 and blood pressure. J Hypertens. 1999;17(1):61–6.
Phillips CL, Yee BJ, Marshall NS, Liu PY, Sullivan DR, Grunstein RR. Continuous positive airway
pressure reduces postprandial lipidemia in obstructive sleep apnea: a randomized, placebo-
controlled crossover trial. Am J Respir Crit Care Med. 2011;184(3):355–61.
Pichiule P, Chavez JC, Boero J, Arregui A. Chronic hypoxia induces modification of the N-methyl-
D-aspartate receptor in rat brain. Neurosci Lett. 1996;218(2):83–6.
Polak J, Shimoda LA, Drager LF, Undem C, McHugh H, Polotsky VY, Punjabi NM. Intermittent
hypoxia impairs glucose homeostasis in C57BL6/J mice: partial improvement with cessation of
the exposure. Sleep. 2013;36(10):1483–1490; 1490a–b.
Polotsky VY, Patil SP, Savransky V, Laffan A, Fonti S, Frame LA, Steele KE, Schweizter MA,
Clark JM, Torbenson MS, et al. Obstructive sleep apnea, insulin resistance, and steatohepatitis
in severe obesity. Am J Respir Crit Care Med. 2009;179(3):228–34.
Prabhakar NR, Kumar GK. Oxidative stress in the systemic and cellular responses to intermittent
hypoxia. Biol Chem. 2004;385(3–4):217–21.
Prabhakar NR, Kumar GK. Mechanisms of sympathetic activation and blood pressure elevation by
intermittent hypoxia. Respir Physiol Neurobiol. 2010;174(1–2):156–61.
Punjabi NM, Beamer BA. Alterations in glucose disposal in sleep-disordered breathing. Am J
Respir Crit Care Med. 2009;179(3):235–40.
Quan SF, Chan CS, Dement WC, Gevins A, Goodwin JL, Gottlieb DJ, Green S, Guilleminault C,
Hirshkowitz M, Hyde PR, et al. The association between obstructive sleep apnea and
neurocognitive performance—the Apnea Positive Pressure Long-term Efficacy Study
(APPLES). Sleep. 2011;34(3):303–314b.
Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol.
2013;200(4):373–83.
Reinke C, Bevans-Fonti S, Drager LF, Shin MK, Polotsky VY. Effects of different acute hypoxic
regimens on tissue oxygen profiles and metabolic outcomes. J Appl Physiol (1985). 2011;111
(3):881–90.
Rossi VA, Stradling JR, Kohler M. Effects of obstructive sleep apnoea on heart rhythm. Eur Respir
J. 2013;41(6):1439–51.
Ryan CM, Usui K, Floras JS, Bradley TD. Effect of continuous positive airway pressure on
ventricular ectopy in heart failure patients with obstructive sleep apnoea. Thorax. 2005;60(9):
781–5.
Sajkov D, Cowie RJ, Thornton AT, Espinoza HA, McEvoy RD. Pulmonary hypertension and
hypoxemia in obstructive sleep apnea syndrome. Am J Respir Crit Care Med. 1994;149(2 Pt 1):
416–22.
Sajkov D, Wang T, Saunders NA, Bune AJ, Neill AM, Douglas Mcevoy R. Daytime pulmonary
hemodynamics in patients with obstructive sleep apnea without lung disease. Am J Respir Crit
Care Med. 1999;159(5 Pt 1):1518–26.
Sajkov D, Wang T, Saunders NA, Bune AJ, McEvoy RD. Continuous positive airway pressure
treatment improves pulmonary hemodynamics in patients with obstructive sleep apnea. Am J
Respir Crit Care Med. 2002;165(2):152–8.
Sandberg O, Franklin KA, Bucht G, Eriksson S, Gustafson Y. Nasal continuous positive airway
pressure in stroke patients with sleep apnoea: a randomized treatment study. Eur Respir
J. 2001;18(4):630–4.
Sanner BM, Doberauer C, Konermann M, Sturm A, Zidek W. Pulmonary hypertension in patients
with obstructive sleep apnea syndrome. Arch Intern Med. 1997;157(21):2483–7.
Sapin E, Peyron C, Roche F, Gay N, Carcenac C, Savasta M, Levy P, Dematteis M. Chronic
intermittent hypoxia induces chronic low-grade neuroinflammation in the dorsal hippocampus
of mice. Sleep. 2015;38(10):1537–46.
Savransky V, Nanayakkara A, Vivero A, Li J, Bevans S, Smith PL, Torbenson MS, Polotsky
VY. Chronic intermittent hypoxia predisposes to liver injury. Hepatology. 2007;45(4):1007–13.
10 Chronic Intermittent Hypoxia in Patients with OSA 205
Savransky V, Jun J, Li J, Nanayakkara A, Fonti S, Moser AB, Steele KE, Schweitzer MA, Patil SP,
Bhanot S, et al. Dyslipidemia and atherosclerosis induced by chronic intermittent hypoxia are
attenuated by deficiency of stearoyl coenzyme A desaturase. Circ Res. 2008;103(10):1173–80.
Schafer H, Koehler U, Ploch T, Peter JH. Sleep-related myocardial ischemia and sleep structure in
patients with obstructive sleep apnea and coronary heart disease. Chest. 1997;111(2):387–93.
Schulz R, Hummel C, Heinemann S, Seeger W, Grimminger F. Serum levels of vascular endothelial
growth factor are elevated in patients with obstructive sleep apnea and severe nighttime hypoxia.
Am J Respir Crit Care Med. 2002;165(1):67–70.
Seicean S, Kirchner HL, Gottlieb DJ, Punjabi NM, Resnick H, Sanders M, Budhiraja R, Singer M,
Redline S. Sleep-disordered breathing and impaired glucose metabolism in normal-weight and
overweight/obese individuals: the Sleep Heart Health Study. Diabetes Care. 2008;31(5):
1001–6.
Sforza E, Krieger J, Weitzenblum E, Apprill M, Lampert E, Ratamaharo J. Long-term effects of
treatment with nasal continuous positive airway pressure on daytime lung function and pulmo-
nary hemodynamics in patients with obstructive sleep apnea. Am Rev Respir Dis. 1990;141(4 Pt
1):866–70.
Shahar E, Whitney CW, Redline S, Lee ET, Newman AB, Nieto FJ, O’Connor GT, Boland LL,
Schwartz JE, Samet JM. Sleep-disordered breathing and cardiovascular disease: cross-sectional
results of the Sleep Heart Health Study. Am J Respir Crit Care Med. 2001;163(1):19–25.
Shamsuzzaman AS, Winnicki M, Lanfranchi P, Wolk R, Kara T, Accurso V, Somers VK. Elevated
C-reactive protein in patients with obstructive sleep apnea. Circulation. 2002;105(21):2462–4.
Shaw JE, Punjabi NM, Wilding JP, Alberti KG, Zimmet PZ. Sleep-disordered breathing and type
2 diabetes: a report from the International Diabetes Federation Taskforce on Epidemiology and
Prevention. Diabetes Res Clin Pract. 2008;81(1):2–12.
Shaw JE, Punjabi NM, Naughton MT, Willes L, Bergenstal RM, Cistulli PA, Fulcher GR, Richards
GN, Zimmet PZ. The effect of treatment of obstructive sleep apnea on glycemic control in type
2 diabetes. Am J Respir Crit Care Med. 2016;194(4):486–92.
Shepard JW Jr, Garrison MW, Grither DA, Dolan GF. Relationship of ventricular ectopy to
oxyhemoglobin desaturation in patients with obstructive sleep apnea. Chest. 1985;88(3):
335–40.
Sherwani SI, Aldana C, Usmani S, Adin C, Kotha S, Khan M, Eubank T, Scherer PE, Parinandi N,
Magalang UJ. Intermittent hypoxia exacerbates pancreatic beta-cell dysfunction in a mouse
model of diabetes mellitus. Sleep. 2013;36(12):1849–58.
Shin MK, Han W, Bevans-Fonti S, Jun JC, Punjabi NM, Polotsky VY. The effect of adrenal
medullectomy on metabolic responses to chronic intermittent hypoxia. Respir Physiol
Neurobiol. 2014a;203:60–7.
Shin MK, Yao Q, Jun JC, Bevans-Fonti S, Yoo DY, Han W, Mesarwi O, Richardson R, Fu YY,
Pasricha PJ, et al. Carotid body denervation prevents fasting hyperglycemia during chronic
intermittent hypoxia. J Appl Physiol (1985). 2014b;117(7):765–76.
Shpirer I, Elizur A, Shorer R, Peretz RB, Rabey JM, Khaigrekht M. Hypoxemia correlates with
attentional dysfunction in patients with obstructive sleep apnea. Sleep Breath ¼ Schlaf Atmung.
2012;16(3):821–7.
Somers VK, Dyken ME, Clary MP, Abboud FM. Sympathetic neural mechanisms in obstructive
sleep apnea. J Clin Invest. 1995;96(4):1897–904.
Somers VK, White DP, Amin R, Abraham WT, Costa F, Culebras A, Daniels S, Floras JS, Hunt
CE, Olson LJ, et al. Sleep apnea and cardiovascular disease: an American Heart Association/
American College of Cardiology Foundation Scientific Statement from the American Heart
Association Council for High Blood Pressure Research Professional Education Committee,
Council on Clinical Cardiology, Stroke Council, and Council on Cardiovascular Nursing. J Am
Coll Cardiol. 2008;52(8):686–717.
Tahrani AA, Ali A, Raymond NT, Begum S, Dubb K, Mughal S, Jose B, Piya MK, Barnett AH,
Stevens MJ. Obstructive sleep apnea and diabetic neuropathy: a novel association in patients
with type 2 diabetes. Am J Respir Crit Care Med. 2012;186(5):434–41.
206 Q. Y. Li et al.
Tahrani AA, Ali A, Raymond NT, Begum S, Dubb K, Altaf QA, Piya MK, Barnett AH, Stevens
MJ. Obstructive sleep apnea and diabetic nephropathy: a cohort study. Diabetes Care. 2013;36
(11):3718–25.
Tang X, Mo C, Wang Y, Wei D, Xiao H. Anti-tumour strategies aiming to target tumour-associated
macrophages. Immunology. 2013;138(2):93–104.
Tanne F, Gagnadoux F, Chazouilleres O, Fleury B, Wendum D, Lasnier E, Lebeau B, Poupon R,
Serfaty L. Chronic liver injury during obstructive sleep apnea. Hepatology. 2005;41(6):1290–6.
Tasali E, Ip MS. Obstructive sleep apnea and metabolic syndrome: alterations in glucose metabo-
lism and inflammation. Proc Am Thorac Soc. 2008;5(2):207–17.
Teramoto S, Kume H, Yamamoto H, Ishii T, Miyashita A, Matsuse T, Akishita M, Toba K, Ouchi
Y. Effects of oxygen administration on the circulating vascular endothelial growth factor
(VEGF) levels in patients with obstructive sleep apnea syndrome. Int Med. 2003;42(8):681–5.
Thunstrom E, Glantz H, Fu M, Yucel-Lindberg T, Petzold M, Lindberg K, Peker Y. Increased
inflammatory activity in nonobese patients with coronary artery disease and obstructive sleep
apnea. Sleep. 2015;38(3):463–71.
Tkacova R, McNicholas WT, Javorsky M, Fietze I, Sliwinski P, Parati G, Grote L, Hedner
J. Nocturnal intermittent hypoxia predicts prevalent hypertension in the European Sleep Apnoea
Database cohort study. Eur Respir J. 2014;44(4):931–41.
Toffoli S, Michiels C. Intermittent hypoxia is a key regulator of cancer cell and endothelial cell
interplay in tumours. FEBS J. 2008;275(12):2991–3002.
Tonon C, Vetrugno R, Lodi R, Gallassi R, Provini F, Iotti S, Plazzi G, Montagna P, Lugaresi E,
Barbiroli B. Proton magnetic resonance spectroscopy study of brain metabolism in obstructive
sleep apnoea syndrome before and after continuous positive airway pressure treatment. Sleep.
2007;30(3):305–11.
Trzepizur W, Le Vaillant M, Meslier N, Pigeanne T, Masson P, Humeau MP, Bizieux-Thaminy A,
Goupil F, Chollet S, Ducluzeau PH, et al. Independent association between nocturnal intermit-
tent hypoxemia and metabolic dyslipidemia. Chest. 2013;143(6):1584–9.
Usui K, Bradley TD, Spaak J, Ryan CM, Kubo T, Kaneko Y, Floras JS. Inhibition of awake
sympathetic nerve activity of heart failure patients with obstructive sleep apnea by nocturnal
continuous positive airway pressure. J Am Coll Cardiol. 2005;45(12):2008–11.
Valham F, Mooe T, Rabben T, Stenlund H, Wiklund U, Franklin KA. Increased risk of stroke in
patients with coronary artery disease and sleep apnea: a 10-year follow-up. Circulation.
2008;118(9):955–60.
Vazir A, Hastings PC, Dayer M, McIntyre HF, Henein MY, Poole-Wilson PA, Cowie MR, Morrell
MJ, Simonds AK. A high prevalence of sleep disordered breathing in men with mild symptom-
atic chronic heart failure due to left ventricular systolic dysfunction. Eur J Heart Fail. 2007;9(3):
243–50.
Vilaseca A, Campillo N, Torres M, Musquera M, Gozal D, Montserrat JM, Alcaraz A, Touijer KA,
Farre R, Almendros I. Intermittent hypoxia increases kidney tumor vascularization in a murine
model of sleep apnea. PLoS One. 2017;12(6):e0179444.
Wang X, Bi Y, Zhang Q, Pan F. Obstructive sleep apnoea and the risk of type 2 diabetes: a meta-
analysis of prospective cohort studies. Respirology. 2013a;18(1):140–6.
Wang N, Khan SA, Prabhakar NR, Nanduri J. Impairment of pancreatic beta-cell function by
chronic intermittent hypoxia. Exp Physiol. 2013b;98(9):1376–85.
Weinstock TG, Wang X, Rueschman M, Ismail-Beigi F, Aylor J, Babineau DC, Mehra R, Redline
S. A controlled trial of CPAP therapy on metabolic control in individuals with impaired glucose
tolerance and sleep apnea. Sleep. 2012;35(5):617–625b.
Weiss JW, Liu MD, Huang J. Physiological basis for a causal relationship of obstructive sleep
apnoea to hypertension. Exp Physiol. 2007;92(1):21–6.
Wessendorf TE, Thilmann AF, Wang YM, Schreiber A, Konietzko N, Teschler H. Fibrinogen
levels and obstructive sleep apnea in ischemic stroke. Am J Respir Crit Care Med. 2000;162(6):
2039–42.
10 Chronic Intermittent Hypoxia in Patients with OSA 207
Xu J, Long YS, Gozal D, Epstein PN. Beta-cell death and proliferation after intermittent hypoxia:
role of oxidative stress. Free Radic Biol Med. 2009;46(6):783–90.
Yang D, Liu Z, Yang H, Luo Q. Effects of continuous positive airway pressure on glycemic control
and insulin resistance in patients with obstructive sleep apnea: a meta-analysis. Sleep Breath ¼
Schlaf Atmung. 2013;17(1):33–8.
Ye L, Ma GH, Chen L, Li M, Liu JL, Yang K, Li QY, Li N, Wan HY. Quantification of circulating
cell-free DNA in the serum of patients with obstructive sleep apnea-hypopnea syndrome. Lung.
2010;188(6):469–74.
Yokoe T, Alonso LC, Romano LC, Rosa TC, O’Doherty RM, Garcia-Ocana A, Minoguchi K,
O’Donnell CP. Intermittent hypoxia reverses the diurnal glucose rhythm and causes pancreatic
beta-cell replication in mice. J Physiol. 2008;586(3):899–911.
Yoshihisa A, Suzuki S, Yamaki T, Sugimoto K, Kunii H, Nakazato K, Suzuki H, Saitoh S, Takeishi
Y. Impact of adaptive servo-ventilation on cardiovascular function and prognosis in heart failure
patients with preserved left ventricular ejection fraction and sleep-disordered breathing. Eur J
Heart Fail. 2013;15(5):543–50.
Yoshihisa A, Suzuki S, Yamauchi H, Sato T, Oikawa M, Kobayashi A, Yamaki T, Sugimoto K,
Kunii H, Nakazato K, et al. Beneficial effects of positive airway pressure therapy for sleep-
disordered breathing in heart failure patients with preserved left ventricular ejection fraction.
Clin Cardiol. 2015;38(7):413–21.
Yumino D, Wang H, Floras JS, Newton GE, Mak S, Ruttanaumpawan P, Parker JD, Bradley
TD. Prevalence and physiological predictors of sleep apnea in patients with heart failure and
systolic dysfunction. J Card Fail. 2009;15(4):279–85.
Zechner R, Kienesberger PC, Haemmerle G, Zimmermann R, Lass A. Adipose triglyceride lipase
and the lipolytic catabolism of cellular fat stores. J Lipid Res. 2009;50(1):3–21.
Zhang X, Ma L, Li S, Wang Y, Wang L. A functional MRI evaluation of frontal dysfunction in
patients with severe obstructive sleep apnea. Sleep Med. 2011;12(4):335–40.
Zhang XB, Yang YY, Zeng Y, Zeng HQ, Fu BB, Ko CY, Luo X, Du YP, Chen LD, Lai YT, et al.
Anti-tumor effect of endostatin in a sleep-apnea mouse model with tumor. Clin Transl Oncol.
2018;6(10):018–1955.
Zhu Y, Fenik P, Zhan G, Mazza E, Kelz M, Aston-Jones G, Veasey SC. Selective loss of
catecholaminergic wake active neurons in a murine sleep apnea model. J Neurosci. 2007;27
(37):10060–71.
Zwillich C, Devlin T, White D, Douglas N, Weil J, Martin R. Bradycardia during sleep apnea.
Characteristics and mechanism. J Clin Invest. 1982;69(6):1286–92.
Chapter 11
Neural Injury in Models of Intermittent
Hypoxia
Sigrid C. Veasey
S. C. Veasey (*)
Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University
of Pennsylvania, Philadelphia, PA, USA
e-mail: veasey@mail.med.upenn.edu
© Springer Nature B.V. and Shanghai Jiao Tong University Press 2022 209
A. I. Pack, Q. Y. Li (eds.), Sleep and its Disorders, Translational Medicine Research,
https://doi.org/10.1007/978-94-024-2168-2_11
210 S. C. Veasey
well as arousal and sleep disruption (Remmers et al. 1978; Tolle et al. 1983; Shiomi
et al. 1991). IH can have sustained effects on sympathetic drive (Katragadda et al.
1997). Specifically, sympathetic drive is lower in sleep overall, relative to wakeful-
ness. With sufficiently frequent and severe oxyhemoglobin desaturations, sympa-
thetic activity remains high across the nighttime sleep period, thereby preventing the
normal “nocturnal dipping” of blood pressure, and with greater changes in oxyhe-
moglobin saturation, sympathetic drive will remain heightened even across the day
(Somers et al. 1988; Gilmartin et al. 2010). Sustained increases in sympathetic drive,
in turn, raise blood pressure, promote thrombosis, and may induce vascular
remodeling, narrowing, and stiffening of arterial vessels [for review, see Austin
et al. (2013)]. All these changes have the potential to negatively impact neural
health, including the promotion of cerebral microvascular disease and stroke. As
we consider OSA as typically a disease present for years before diagnosis and
treatment, it is important to appreciate that the consequences of IH in OSA extend
beyond acute direct neuronal changes to include the above-mentioned pathophysi-
ological changes that, in turn, may also impact neural function.
Patterns and severity of hypoxemia in OSA are influenced by numerous factors.
Within a given obstructive sleep-disordered breathing event, the oxyhemoglobin
saturation nadir is determined by (1) the effect of the obstruction on ventilation
(apnea or hypopnea), (2) the functional residual capacity of the lungs in the particular
sleep stage in which an event occurs, and (3) central ventilatory control and
responsiveness in that sleep stage for that individual. For example, patients with
underlying impaired respiratory control at baseline may have a higher arousal
threshold for carbon dioxide (CO2) and/or hypoxemia severity and therefore longer
events with lower oxygen and higher CO2 arterial values. Progesterone, and poten-
tially estrogen, have stimulatory effects on ventilation (Behan and Wenninger 2008),
so that premenopausal females typically have higher oxygen values for a similar
level of obstruction as males or postmenopausal females. Additionally, individuals
with significant underlying restrictive lung disease or severe obesity, who have
reduced functional residual capacity in the lungs in sleep, will have more rapid
decline in oxygen values and thus may have more severe oxyhemoglobin
desaturations across an obstructive sleep-disordered breathing event of a given
length (Dempsey et al. 2010). The oxyhemoglobin patterns will also vary with
stage of sleep. For example, in rapid eye movement (REM) sleep, most accessory
muscles are paralyzed (further reducing the functional residual capacity) (Douglas
et al. 1982). Obstructive sleep-disordered breathing events may be limited to REM
sleep (when upper airway muscle tone is most reduced and ventilatory responsive-
ness is also reduced). In this case, IH occurs sparsely across the night but in 3–10 min
periods. There are also individuals who have IH for reasons of OSA combined with a
pulmonary or neuromuscular abnormality (e.g., severe chronic obstructive lung
disease or unilateral diaphragmatic paralysis). These individuals may have not
only IH but also sustained hypoxemia, particularly across REM sleep (Anderson
et al. 1996).
At present, the risk of cardiovascular morbidities and mortality, and cognitive
impairments in OSA have been largely related to the apnea/hypopnea index, which
11 Neural Injury in Models of Intermittent Hypoxia 211
largely relates to oxygen desaturation frequency (Nieto et al. 2000; Young and
Peppard 2000; Shahar et al. 2001; Stone et al. 2016). While clinical studies have
not parceled out in humans the relative risk of the particular patterns described
above, work in animal models (described below) suggests that very different out-
comes are possible with the variations in oxyhemoglobin patterns. In clinical studies,
it is quite difficult to distinguish effects of IH from the interconnected pathophysi-
ological effects of obstructive sleep-disordered breathing events described above.
Moreover, because obesity is a major risk factor for OSA, it is very difficult in
clinical studies to delineate what the consequences of OSA are from the conse-
quences of diabetes, hypertension, cardiovascular disease, etc. In light of these
challenges, animal studies have been helpful in elucidating potential detrimental
and beneficial effects of IH on the brain and the mechanisms by which IH influences
neural function and injury.
The first published account of IH exposures in animals dates back to 1948, well
before the clinical recognition of OSA (Altland 1948). The work was presented in
abstract form describing rats exposed to hypoxia for 4 h/day for 3 months. Remark-
ably, this particular pattern of IH exposure was shown to have effects on hematocrit,
body growth and spermatogenesis that persisted for weeks into the recovery period
(Altland 1948). Neuronal function and health, however, were not examined (Altland
1948). One of the first descriptions of IH effects on the brain came from Alberghina
and Giuffrida in 1981 (Alberghina and Giuffrida 1981), with a demonstration that
intermittent sustained hypoxia (hypoxia for 17 h/day for 6 days) reduces lipid
biogenesis in the brain. Serra et al. (1981), using the same protocol, also in 1981,
showed reduced labeled precursor uptake for deoxyribonucleic and ribonucleic acids
and proteins, particularly in mitochondrial fractions. In both studies, effects were
observed in the cortex and hippocampus without an effect observed in the striatum,
212 S. C. Veasey
suggesting regional or cell type differential susceptibility to IH. As with the first
paradigm, effects lasted beyond exposure and in this case were evident for at least
12 h after the last exposure. In another model of sustained IH (10 h/day) for 10 days,
Meyer and Doelle (1988) found that even 3 months after IH exposure, there were
alterations in the endoplasmic reticulum and ribosomes, with more free polysomes,
in the cortices. Hermans et al. (1992) found that sustained (4 h once daily for 5 days)
IH in utero across the last third of gestation, resulted in enduring behavioral
disturbances (Hermans et al. 1992). Specifically, the awake righting reflex was
delayed on postnatal days 6 and 7. Open-field locomotor activity was reduced in
females but not males at 19 days, supporting a gender effect of IH. Intriguingly, even
later as young adults (postnatal days 60–65), mice exposed in utero to IH had less
locomotor activity for the first half of the lights off (active) period.
The body has important adaptive responses to chronic sustained hypoxia. Adap-
tive responses include increasing red blood cells to carry more oxygen, expanding
the vasculature also to deliver more oxygen, and a shift in metabolism toward
glycolysis. The molecular response to these physiological changes includes increas-
ing erythropoietin to increase red blood cell mass, increasing vascular growth
factors, and activating specific transcriptional pathways to promote glycolysis over
aerobic metabolism [for reviews, see Jelkmann (2007) and Prabhakar and Semenza
(2015)]. Perhaps the most established molecular response to sustained hypoxia was
discovered by Wang and Semenza, who discovered hypoxia-inducible factor-1 alpha
(HIF1a) as a major transcriptional activator (Wang and Semenza 1993), essential for
the erythropoietin response to hypoxia (Heinicke et al. 2003). For the past three
decades, some athletes have taken advantage of brief intermittent sustained mild
hypoxia (2–5 exposures of 3–5 min) exposures to increase erythropoietin and
physical endurance (Heinicke et al. 2003; Piehl Aulin et al. 1998).
the ventral lateral medulla) in response to LTIH. The regional pattern of c-fos
expression was remarkably consistent across all rats exposed to LTIH. The
brainstem c-fos activation induced by IH was evident 12–18 h after the last IH
exposure. The persistence in c-fos activation many hours after the last LTIH
exposure supports the concept that LTIH can have lasting effects on brain responses
and function. Additionally, this work supports the concept that cardiovascular
responses to OSA may at least in part be secondary to neurogenic changes in
response to LTIH. In a follow-up study, the same researchers examined the cortical
c-fos response to LTIH and found that here, too, only very specific neurons were
activated in response to LTIH (Sica et al. 2000). These groups included layers II/III
of the cortex, particularly the cingulate and piriform cortices (Sica et al. 2000), areas
also implicated in sympathetic responses. Collectively, this early body of work
provides evidence that the brain responds to LTIH; the response can last longer
than the IH exposure, and that the response within the brain at least for this
immediate early gene activation is regionally, and potentially neuronal group
specific.
One of the most remarkable IH findings is that even very brief exposures to IH (three
5 min exposures) can have lasting effects on a respiratory neuronal function beyond
the termination of the stimulus. This prolonged effect on neuronal activity or
function is termed plasticity. Respiratory plasticity was originally described by
Millhorn et al. with repeated carotid sinus stimulation (Millhorn et al. 1980).
Understanding that the carotid sinus can be stimulated by hypoxia, IH (using the
same temporal pattern as carotid nerve stimulation) was tested for effects on phrenic
nerve activity and found to promote phrenic nerve long-term facilitation in anesthe-
tized rats (Hayashi et al. 1993). Similarly in anaesthetized rats, IH, administered as
three bursts of 3 min of hypoxia (arterial oxygen 35–45 mmHg) while holding
carbon dioxide constant (acute IH), results in a sustained (at least 60 min) increase in
phrenic nerve activity (Dwinell et al. 1997). Intriguingly, phrenic plasticity was not
observed in response to sustained hypoxia (Baker and Mitchell 2000). There is, at
least in rats, a genetic basis for acute IH in that only specific strains and substrains of
rats show evidence of the phrenic nerve long-term facilitation (LTF) response
(Janssen and Fregosi 2000; Fuller et al. 2001). Genetics underlying the strain
differences in phrenic LTF responses to acute IH have not been elucidated. LTF
response to short-term IH is also age-dependent in males, in that it is only observed
in very young adult male rats; in middle-aged male rats, the response is completely
blunted (Zabka et al. 2001). There are gender differences in this age-dependency of
effect in that while older male rats do not show acute IH phrenic LTF, older female
rats do (Behan et al. 2002).
The molecular basis of the phrenic LTF response to acute IH has been extensively
evaluated and involves several complex molecular signaling pathways. Central to
phrenic LTF from acute IH is the activation of caudal serotoninergic raphe neurons
(Ling et al. 2001). These activated neurons send increased serotonin (5-HT) to
phrenic motoneurons, which is essential for acute IH plasticity (Baker-Herman and
Mitchell 2002). Repeated activation of 5-HT receptors (subtypes 2A, 2C, and/or 7)
initiate several cell signaling pathways that ultimately increase the production and
activation of brain-derived neurotrophic factor (BDNF) (Leal et al. 2014). BDNF
then activates the troponin receptor kinase B (Trk-B) receptor on the motoneurons to
phosphorylate and activate an extracellular signal kinase (ERK) or mitogen-
activated protein kinase (MAPK) (Wilkerson and Mitchell 2009; Dougherty et al.
2015). Each of these components is critical to acute IH phrenic LTF (Ling et al.
2001). A second distinct pathway has been identified that activates protein kinase A
that in turn activates TRK to activate AKT which augments glutamatergic signaling,
leading to LTF (Nichols et al. 2012). The system is not redundant in that the former
PKC pathway is activated in moderate acute IH while the latter pathway is activated
in severe hypoxia (paO2 < 35 mmHg) (Nichols et al. 2012). Terada and Mitchell
examined the role of sleep in the phrenic LTF response to acute IH (Terada and
Mitchell 2011). Acute IH increases diaphragmatic LTF in both wakefulness and
11 Neural Injury in Models of Intermittent Hypoxia 215
NREM sleep but does not affect phrenic activity in REM sleep. Daily acute IH does
not influence either baseline diaphragmatic activity or induce LTF lasting hours and,
therefore, cannot induce a metaplasticity of the phrenic response that would last
across longer periods of sleep to protect sleep-disordered breathing. Nonetheless,
now understanding the molecular mechanisms of acute IH, pharmacologic targets
along the acute IH LTF pathways may be tested for effectiveness in improving
ventilation during sleep.
In light of the above responses, it would seem that IH is beneficial and should help
prevent patients with OSA from having apneic events in sleep, at least across
portions of the night. In this section below, research is summarized to support the
concept that the above-described effects with acute IH are not always observed in
chronic IH (CIH). Indeed, acute IH to mild levels of hypoxemia can augment
respiratory nerve activity, while longer more severe CIH has detrimental effects on
neural function.
Peng and Prabhakar examined whether moderate 1-week CIH, modeling OSA,
induced phrenic LTF (Peng and Prabhakar 2003). Importantly, the investigators
found that CIH for 1 week prior to acute IH enhanced phrenic LTF, while sustained
hypoxia actually prevented the LTF, and the CIH-enhanced LTF effect could be
blunted by giving a reactive oxygen species scavenger (Peng and Prabhakar 2003).
Thus, CIH can influence the above acute IH phrenic LTF and this effect appears to
involve reactive oxygen species.
Appreciating that the carotid body is the gatekeeper for cardiorespiratory
responses to hypoxia of all forms, including IH, mechanisms whereby CIH activates
carotid body glomus cells were recently addressed. One of the critical steps toward
glomus cell activation is an increase in intracellular calcium (Kumar and Prabhakar
2012). A rise in intracellular calcium may emanate from endoplasmic reticulum or
mitochondrial stores within or through influx via voltage-gated calcium channels.
However, Makarenko et al., found that blocking T-type voltage-gated calcium
channels (VGCCs) prevented the CIH effect on intracellular calcium in glomus
cells (Makarenko et al. 2012). A reactive oxygen species scavenger prevented the
CIH-induced increase in calcium as well, suggesting that ROS activates the Type 3.2
VGCC to increase intracellular calcium (Makarenko et al. 2012). Mechanisms by
which calcium influx into the glomus cells leads to long-term facilitation of
216 S. C. Veasey
One of the most significant advances in IH neurobiology has been the incredible
detailing of oxygen sensing mechanisms in the carotid body, and while it is quite
complicated, it is worthwhile considering, as this is certainly one of the first chains of
events in IH neural cardiopulmonary responses. The overall picture is that fluctua-
tions in oxygen in IH increase the availability of reactive oxygen species that sets up
a complex signaling pathway leading to sympathetic activity through gaseous trans-
mitters in the carotid body. In greater detail, seated just at the bifurcation of the
carotid artery into internal and external carotid arteries, carotid bodies are well-
poised to almost immediately detect fluctuations in oxygen delivery to the brain. The
actual oxygen sensing cells are the glomus cells described above. But just how do
glomus cells translate low oxygen tension into calcium influx? The reoxygenation
phase in IH increases reactive oxygen species within the carotid body, which then
oxidatively modify and inactivate hemoxygenase-2 (Yuan et al. 2004). In normoxia,
hemoxygenase-2 is constitutively active, catalyzing the synthesis of carbon monox-
ide. Carbon monoxide then, in normoxia, activates protein kinase G, which phos-
phorylates and inactivates cystathionine g-ligase (CSE) an enzyme critical for
hydrogen sulfide (H2S) production (Yuan et al. 2015a). In CIH with
hemoxygenase-2 inactivated carbon monoxide is not present to suppress ultimately
CSE from making H2S. H2S excites carotid glomus cells by inhibiting background
(TASK) potassium channels, and increasing intracellular calcium (Buckler 2012; Li
et al. 2010; Telezhkin et al. 2010). Inhibition of CSE and transgenic absence of CSE
both prevent IH activation of sympathetic drive (Yuan et al. 2016). It is exciting to
consider that drugs targeting carotid body CSE might provide an important thera-
peutic avenue to reduce sympathetic drive in OSA.
As described above, CIH results in chronic activation of carotid sinus neurons. How
this translates into increased blood pressure has been carefully examined and
recently reviewed (Kumar et al. 2015). The adrenal medulla is essential for
CIH-induced hypertension in that rats subjected to bilateral adrenal medullectomy
confer resistance to hypertension produced by CIH (Bao et al. 1997). It is quite
interesting that several of the same mediators in CIH responses in adrenal medullary
11 Neural Injury in Models of Intermittent Hypoxia 217
chromaffin cells are the same players in the carotid glomus cellular response,
including increased intracellular calcium and increased reactive oxygen species.
CIH seems to disturb the balance of hypoxia-inducible factors (HIFs) 1 and
2. HIF-1α promotes glycolysis over oxidative phosphorylation and a pro-oxidant
state (increased NADPH oxidase, which is critical to the CIH-increased ROS, even
in glomus cells), while HIF-2α promotes the transcription of many antioxidant
enzymes (superoxide dismutases 1 and 2 and catalase) (Semenza and Prabhakar
2018). Overall, CIH-induced carotid sinus nerve activation leads to sympathetic
nerve activation that in turn somehow alters the balance of HIF-1α and -2α to favor a
pro-oxidant HIF-1α state (Peng et al. 2006, 2014; Nanduri et al. 2009).
Overall, these well-delineated pathways provide several promising targets for the
prevention of OSA-related hypertension, including targeting NADPH oxidase inhib-
itors and drugs that influence H2S production.
The hippocampus is particularly sensitive to hypoxia (Kirino and Sano 1984), and
within the hippocampus neurons in the CA1 region are the most vulnerable to
constant hypoxia (Schmidt-Kastner and Freund 1991). David Gozal and colleagues
began examining the mechanisms of this differential vulnerability of CA1 compared
to CA3 neurons to longer-term severe CIH, finding increased hypoxic depolarization
in CA1 vs. CA3 pyramidal neurons (Kreisman et al. 2000). Then, in considering
OSA, his group utilized a longer and more frequent exposure of IH (40 events/h to
FIO2 concentrations around 10% continuously for 1–14 days) in young adult rats
(2 months old) (Gozal et al. 2001). This long-term IH (LTIH) did not alter total
hourly amounts of wakefulness, NREM or REM sleep, but markedly impaired
performance in spatial memory (hippocampal-dependent) task, the Morris water
maze. Not only was performance acutely impaired but even after 14 days into
recovery performance remained impaired. Histological examination of the hippo-
campus revealed apoptosis in the CA1, supporting heightened vulnerability for the
CA1 also with LTIH. Additionally, 14 days of IH (LTIH) resulted in an astrocytosis
within the hippocampus. This landmark study provided the first evidence that LTIH
could impart lasting functional impairments and neural injury. A similar immediate
effect of LTIH on hippocampal-dependent memory was observed in developing rats
(age 15–30 days) (Row et al. 2002). Here, the effect was more pronounced in male
rats than in females. Shortly thereafter, Michael Decker with David Rye confirmed
spatial memory impairments in developing rats exposed to LTIH and found that
impairments in performance persisted at least until the age of young adults (2mos)
(Decker et al. 2003). Additionally, these rats had sustained reductions in wakeful-
ness and increases in REM sleep (Decker et al. 2003). In an effort to begin to
elucidate major molecular pathways involved in a relatively non-biased manner,
218 S. C. Veasey
Evelyn Gozal and colleagues analyzed proteomic differences within the CA1 (LTIH
vulnerable hippocampus) and CA3 (relatively LTIH resistant) regions of the hippo-
campus (Gozal et al. 2002). Overall, the proteomic changes in CA1 not evident in
CA3 included alterations in cytoskeletal, metabolic/energetic, and chaperone pro-
teins. Intriguingly, there is a strong age-dependency to the vulnerability to IH, in that
rats pups exposed at very early stages (postnatal day 2–5) develop less hippocampal
neuronal apoptosis relative to older pups. The most vulnerable group spanned rat
pups of 10–25 days, so that rats older at the time of exposure (up to 120 days of age
studied) had less apoptosis. It is important to recognize that the duration of LTIH
may also be important as 2 vs. 4 or more weeks may also have very different effects
on hippocampal responses to LTIH. Specifically, apoptosis of CA1 pyramidal
neurons peaks at day 3 of LTIH exposure, and phosphorylation of CREB peaks at
day 14 and then declines gradually to normoxic levels by day 30 of LTIH (Goldbart
et al. 2003a). Similarly, phosphorylated protein kinase B (AKT) and phosphorylated
glycogen synthase kinase B (GSK-B) both decline across the first day of exposure
and then gradually return to baseline levels over 2 week (Goldbart et al. 2003b). A
temporal pattern in hippocampal function was also observed in that long-term
potentiation in hippocampal brain slices was markedly reduced (34% of baseline)
at day 3 IH and then improved to 54% of baseline by day 7 IH (Payne et al. 2004).
Clinical imaging studies of the hippocampus in patients with OSA demonstrate
reduced volume, less gray matter, and potentially some reversal with treatment.
Clearly, understanding the molecular mechanisms by which OSA and IH can disturb
hippocampal function is of utmost importance. It is quite interesting then that some
of the same molecular pathways as described above for acute IH phrenic LTF and for
carotid body and adrenal medullary responses to LTIH, are highlighted here as well.
Specifically, LTIH increases oxidative stress in the hippocampus, increasing
peroxynitrite and reducing nitric oxide (NO) availability. In animals exposed to
LTIH, this reduction in NO availability impairs calcium-activated potassium chan-
nels on CA1 pyramidal neurons (Tjong et al. 2008a). Melatonin supplementation
can, interestingly prevent the LTIH-induced impaired potassium channel responses
(Tjong et al. 2008b), potentially by reducing oxidative stress. Additionally,
increased physical exercise which increases both BDNF and antioxidant defense in
brain is also protective against LTIH spatial memory impairments (Gozal et al.
2010). An important source of oxidative stress seems to be NADPH oxidase, in
that mice deficient in one of the key subunits for NADPH oxidase are resistant to
both the LTIH-induced oxidative stress and spatial memory impairments (Nair et al.
2011). Of interest NADPH oxidase activity in the hippocampi of wild-type mice
gradually climbs over the first week of life and then plateaus (Nair et al. 2011).
Additional sustained sources of oxidative stress in LTIH within the hippocampus
include HIF-1α and endoplasmic reticulum oxidoreductin-1 like (ERO-1L) that are
both elevated on a sustained basis in the hippocampus (Chou et al. 2013). Angio-
tensin II inhibition can prevent oxidative stress, lipid peroxidation, and inflammatory
responses (Yuan et al. 2015b). Of interest, angiotensin II activation in the carotid
sinus also potentiates NADPH oxidase activation, and is therefore responsible in
large part for the NADPH oxidase ROS. Overall, the above research supports
11 Neural Injury in Models of Intermittent Hypoxia 219
Residual sleepiness persists in over 10% of patients in which OSA and other sleep
disorders have been treated (Gasa et al. 2013). This finding raises the possibility that
OSA through IH or sleep fragmentation might injure neurons essential for alertness
and optimal wakefulness. Veasey et al. (2004a) examined the effects of 8 week of
severe LTIH (40 events/h to oxyhemoglobin saturation nadirs of 75%) on sponta-
neous wakefulness in a 24-h period and sleepiness as measured with a mouse version
of the multiple sleep latency test (Veasey et al. 2004b). To determine whether
sleepiness was affected and whether the effect could persist, wake parameters were
assessed for 2 and 24 week into recovery in normoxia after LTIH. Total sleep time
was increased for a 24-h period in LTIH mice by 2 and ½ h. Both NREM and REM
sleep times increased, and effects were present at least 6 mos into recovery (Veasey
et al. 2004a). LTIH also significantly reduced sleep latency (Veasey et al. 2004a).
Lipid peroxidation, which signifies both oxidative and nitrative stress, was increased
in the lateral hypothalamus and basal forebrain (Veasey et al. 2004a). In a follow-up
study, the role of NO in wake impairments was addressed by studying the effects of
LTIH on wakefulness in mice with and without inducible NOS (iNOS) (Zhan et al.
2005a). That transgenic absence of iNOS prevented the wake impairments, LTIH
oxidative and inflammatory responses strongly support a critical role for ROS and
NO in both behavioral and molecular responses to LTIH (Zhan et al. 2005a).
Importantly, NADPH oxidase is central to the iNOS activation, as well as the
downstream oxidative stress, inflammatory response, and wake impairments, in
that all can be prevented in mice with either transgenic absence or pharmacologic
inhibition of NADPH oxidase (Zhan et al. 2005b). As with acute IH, female mice
conferred some resistance to IH injury to wake impairments (Sanfilippo-Cohn et al.
2006). The most important finding in this series of studies was that catecholaminer-
gic wake-activate neurons, the noradrenergic locus coeruleus and the dopaminergic
ventral periacqueductal gray neurons were not only less responsive but lost with
LTIH (Zhu et al. 2007). This injury effect as well was an NADPH oxidase-
dependent effect.
Residual sleepiness impacts the well-being and safety of patients with OSA and
injury to wake-activated neurons may also contribute to depression, cognitive
impairments, and in the case of the noradrenergic locus coeruleus neurons, injury
can accelerate cognitive decline, and amyloid plaques in Alzheimer’s disease
(Weinshenker 2008). Thus, identifying therapies that protect wake-activate neurons
and wakefulness in persons with OSA should be a high priority in sleep medicine. It
220 S. C. Veasey
is important to note that there have been intermittent hypoxia trials in humans to
assess effects on cognition. When limited to one 90-min episode of hypoxia with
oxyhemoglobin saturation nadirs of 85% three times 1 week and three times at 80%
the next, older humans improved in cognitive performances (executive function)
using a within-subject assessment (Schega et al. 2016). Weiss et al. (2009) examined
the effects of an IH pattern that modeled moderate OSA with 20 events/h and
desaturations into the high to mid-80th %-ile. Exposures were given 9 h/night for
28 days. Using a within-study design the researchers did not observe any differences
in cognitive performance. However, the sample size was small (n ¼ 8), and all were
healthy young adults. Additionally, there was no control group to exclude learning
effects with some of the tests over time. Thus, it remains unknown whether humans
are resistant to IH effects on cognition and sleepiness; whether LTIH effects are
evident only with severe hypoxia and more frequent events, or whether
comorbidities influence cognitive and sleepiness effects of LTIH.
Another group of neurons that may be injured by LTIH are upper airway dilator
motoneurons that are critical for pharyngeal patency. Several studies have shown
injury to the nerves in upper airway dilator muscles and changes in the muscles
consistent with nerve injury (Saboisky et al. 2012; Ramchandren et al. 2010; Boyd
et al. 2004). LTIH has been shown to influence upper airway motoneurons activity.
Specifically, the effects of LTIH on hypoglossal nerve responses to excitatory
neurochemicals serotonin and glutamate have been examined and shown to be
markedly reduced in animals after LTIH (Veasey et al. 2004c). This supports the
view that severe LTIH leads to major disruption in functional responsiveness of
upper airway motoneurons. Endoplasmic reticulum stress has been shown in the
LTIH exposed motoneurons (Zhu et al. 2008). The molecular basis of this hypo-
glossal neuron injury involves endoplasmic reticulum dyshomeostasis as evidenced
by increased C/EBP homologous binding protein (CHOP) in these motoneurons that
could be prevented by enhancing an adaptive ER stress response (eIF-2a) (Zhu et al.
2008). (For further discussion of the role of CHOP, see Chap. 7) Importantly, CHOP
plays a major role in both the NADPH oxidase and inflammatory responses. The
transgenic absence of CHOP prevents oxidative, nitrative, and inflammatory injuries
in upper airway motoneurons, including hypoglossal neurons. Additionally, CHOP
is essential for the HIF1a response to LTIH at motoneurons, and thus may be the
most upstream pathway component identified to date (Chou et al. 2013).
11 Neural Injury in Models of Intermittent Hypoxia 221
Glial cells within the central nervous system include astrocytes, microglia, and
oligodendrocytes. Recent reports suggest that these cells, too, maybe affected by
IH. Specifically, very severe LTIH (60 events/h to a fraction of inspired oxygen of
5%) causes a subtle increase in a number of astrocytes in the dorsal hippocampus in
adult mice (Sapin et al. 2015). Whether this gliosis contributes to injury or is
protective is not known. As with neuronal responses to IH, microglia responses
may be beneficial with mild exposures and harmful with more severe exposures
(Kiernan et al. 2016). The clearest evidence of glial injury from IH involves
oligodendrocytes. Severe LTIH disturbs the ultrastructure of myelin and reduces
myelin proteins (Veasey et al. 2013; Kim et al. 2015). Importantly, this is true in
developing rats as well upon exposure to LTIH (Cai et al. 2012). Individuals with
OSA have increased white matter lesions, and the prevalence increases with OSA
severity (Kim et al. 2013).
IH in humans with OSA occurs across a vast spectrum of severity, defined by the
frequency and severity of oxyhemoglobin desaturation. Several mild IH events can
augment ventilation and upper airway patency, while the more severe events have
lasting effects on neuronal function and survival in selectively vulnerable groups of
neurons and may impact glial function. NADPH oxidase plays a vital role in both
LTF plasticity and neuronal injury and degradation. Because NADPH oxidase may
also contribute to metabolic disturbances in OSA and to cardiovascular pathophys-
iology, this oxidase should be a major target for pharmacotherapies to prevent the
major comorbidities in OSA. As CHOP is upstream to NADPH oxidase, this too
may be a promising target for preventative therapies. Collectively, animal studies
have successfully provided a translational platform, ripe for implementation science.
References
Alberghina M, Giuffrida AM. Effect of hypoxia on the incorporation of [2-3H] glycerol and
[1-14C]-palmitate into lipids of various brain regions. J Neurosci Res. 1981;6(3):403–19.
Altland PD. Recovery rate from some of the effects of chronic intermittent hypoxia in rats. Anat
Rec. 1948;101(4):668.
Anderson CA, Dick TE, Orem J. Respiratory responses to tracheobronchial stimulation during sleep
and wakefulness in the adult cat. Sleep. 1996;19(6):472–8.
222 S. C. Veasey
Austin AW, Wissmann T, von Kanel R. Stress and hemostasis: an update. Semin Thromb Hemost.
2013;39(8):902–12.
Baker TL, Mitchell GS. Episodic but not continuous hypoxia elicits long-term facilitation of
phrenic motor output in rats. J Physiol. 2000;529(Pt 1):215–9.
Baker-Herman TL, Mitchell GS. Phrenic long-term facilitation requires spinal serotonin receptor
activation and protein synthesis. J Neurosci. 2002;22(14):6239–46.
Bao G, Metreveli N, Li R, Taylor A, Fletcher EC. Blood pressure response to chronic episodic
hypoxia: role of the sympathetic nervous system. J Appl Physiol (1985). 1997;83(1):95–101.
Behan M, Wenninger JM. Sex steroidal hormones and respiratory control. Respir Physiol
Neurobiol. 2008;164(1–2):213–21.
Behan M, Zabka AG, Mitchell GS. Age and gender effects on serotonin-dependent plasticity in
respiratory motor control. Respir Physiol Neurobiol. 2002;131(1–2):65–77.
Boyd JH, Petrof BJ, Hamid Q, Fraser R, Kimoff RJ. Upper airway muscle inflammation and
denervation changes in obstructive sleep apnea. Am J Respir Crit Care Med. 2004;170(5):
541–6.
Buckler KJ. Effects of exogenous hydrogen sulphide on calcium signalling, background (TASK) K
channel activity and mitochondrial function in chemoreceptor cells. Pflugers Arch. 2012;463(5):
743–54.
Cai J, Tuong CM, Zhang Y, Shields CB, Guo G, Fu H, et al. Mouse intermittent hypoxia mimicking
apnoea of prematurity: effects on myelinogenesis and axonal maturation. J Pathol. 2012;226(3):
495–508.
Chou YT, Zhan G, Zhu Y, Fenik P, Panossian L, Li Y, et al. C/EBP homologous binding protein
(CHOP) underlies neural injury in sleep apnea model. Sleep. 2013;36(4):481–92.
Decker MJ, Hue GE, Caudle WM, Miller GW, Keating GL, Rye DB. Episodic neonatal hypoxia
evokes executive dysfunction and regionally specific alterations in markers of dopamine
signaling. Neuroscience. 2003;117(2):417–25.
Dempsey JA, Veasey SC, Morgan BJ, O’Donnell CP. Pathophysiology of sleep apnea. Physiol
Rev. 2010;90(1):47–112.
Dougherty BJ, Fields DP, Mitchell GS. Mammalian target of rapamycin is required for phrenic
long-term facilitation following severe but not moderate acute intermittent hypoxia. J
Neurophysiol. 2015;114(3):1784–91.
Douglas NJ, White DP, Pickett CK, Weil JV, Zwillich CW. Respiration during sleep in normal
man. Thorax. 1982;37(11):840–4.
Douglas RM, Miyasaka N, Takahashi K, Latuszek-Barrantes A, Haddad GG, Hetherington
HP. Chronic intermittent but not constant hypoxia decreases NAA/Cr ratios in neonatal
mouse hippocampus and thalamus. Am J Physiol Regul Integr Comp Physiol. 2007;292(3):
R1254–9.
Dwinell MR, Janssen PL, Bisgard GE. Lack of long-term facilitation of ventilation after exposure to
hypoxia in goats. Respir Physiol. 1997;108(1):1–9.
Fletcher EC. An animal model of the relationship between systemic hypertension and repetitive
episodic hypoxia as seen in sleep apnoea. J Sleep Res. 1995;4(S1):71–7.
Fletcher EC, Lesske J, Qian W, Miller CC 3rd, Unger T. Repetitive, episodic hypoxia causes diurnal
elevation of blood pressure in rats. Hypertension. 1992;19(6 Pt 1):555–61.
Fletcher EC, Bao G, Miller CC 3rd. Effect of recurrent episodic hypocapnic, eucapnic, and
hypercapnic hypoxia on systemic blood pressure. J Appl Physiol (1985). 1995;78(4):1516–21.
Fuller DD, Baker TL, Behan M, Mitchell GS. Expression of hypoglossal long-term facilitation
differs between substrains of Sprague-Dawley rat. Physiol Genomics. 2001;4(3):175–81.
Gasa M, Tamisier R, Launois SH, Sapene M, Martin F, Stach B, et al. Residual sleepiness in sleep
apnea patients treated by continuous positive airway pressure. J Sleep Res. 2013;22(4):389–97.
Gilmartin GS, Lynch M, Tamisier R, Weiss JW. Chronic intermittent hypoxia in humans during
28 nights results in blood pressure elevation and increased muscle sympathetic nerve activity.
Am J Physiol Heart Circ Physiol. 2010;299(3):H925–31.
11 Neural Injury in Models of Intermittent Hypoxia 223
Goldbart A, Cheng ZJ, Brittian KR, Gozal D. Intermittent hypoxia induces time-dependent changes
in the protein kinase B signaling pathway in the hippocampal CA1 region of the rat. Neurobiol
Dis. 2003a;14(3):440–6.
Goldbart A, Row BW, Kheirandish L, Schurr A, Gozal E, Guo SZ, et al. Intermittent hypoxic
exposure during light phase induces changes in cAMP response element binding protein activity
in the rat CA1 hippocampal region: water maze performance correlates. Neuroscience.
2003b;122(3):585–90.
Gozal D, Daniel JM, Dohanich GP. Behavioral and anatomical correlates of chronic episodic
hypoxia during sleep in the rat. J Neurosci. 2001;21(7):2442–50.
Gozal E, Gozal D, Pierce WM, Thongboonkerd V, Scherzer JA, Sachleben LR Jr, et al. Proteomic
analysis of CA1 and CA3 regions of rat hippocampus and differential susceptibility to inter-
mittent hypoxia. J Neurochem. 2002;83(2):331–45.
Gozal D, Nair D, Goldbart AD. Physical activity attenuates intermittent hypoxia-induced spatial
learning deficits and oxidative stress. Am J Respir Crit Care Med. 2010;182(1):104–12.
Greenberg HE, Sica AL, Scharf SM, Ruggiero DA. Expression of c-fos in the rat brainstem after
chronic intermittent hypoxia. Brain Res. 1999;816(2):638–45.
Hayashi F, Coles SK, Bach KB, Mitchell GS, McCrimmon DR. Time-dependent phrenic nerve
responses to carotid afferent activation: intact vs. decerebellate rats. Am J Phys. 1993;265(4 Pt
2):R811–9.
Heinicke K, Prommer N, Cajigal J, Viola T, Behn C, Schmidt W. Long-term exposure to
intermittent hypoxia results in increased hemoglobin mass, reduced plasma volume, and
elevated erythropoietin plasma levels in man. Eur J Appl Physiol. 2003;88(6):535–43.
Hermans RH, Hunter DE, McGivern RF, Cain CD, Longo LD. Behavioral sequelae in young rats of
acute intermittent antenatal hypoxia. Neurotoxicol Teratol. 1992;14(2):119–29.
Janssen PL, Fregosi RF. No evidence for long-term facilitation after episodic hypoxia in sponta-
neously breathing, anesthetized rats. J Appl Physiol (1985). 2000;89(4):1345–51.
Jelkmann W. Erythropoietin after a century of research: younger than ever. Eur J Haematol.
2007;78(3):183–205.
Katragadda S, Xie A, Puleo D, Skatrud JB, Morgan BJ. Neural mechanism of the pressor response
to obstructive and nonobstructive apnea. J Appl Physiol (1985). 1997;83(6):2048–54.
Kiernan EA, Smith SM, Mitchell GS, Watters JJ. Mechanisms of microglial activation in models of
inflammation and hypoxia: implications for chronic intermittent hypoxia. J Physiol. 2016;594
(6):1563–77.
Kim H, Yun CH, Thomas RJ, Lee SH, Seo HS, Cho ER, et al. Obstructive sleep apnea as a risk
factor for cerebral white matter change in a middle-aged and older general population. Sleep.
2013;36(5):709–15B.
Kim LJ, Martinez D, Fiori CZ, Baronio D, Kretzmann NA, Barros HM. Hypomyelination, memory
impairment, and blood-brain barrier permeability in a model of sleep apnea. Brain Res.
2015;1597:28–36.
Kirino T, Sano K. Selective vulnerability in the gerbil hippocampus following transient ischemia.
Acta Neuropathol. 1984;62(3):201–8.
Kreisman NR, Soliman S, Gozal D. Regional differences in hypoxic depolarization and swelling in
hippocampal slices. J Neurophysiol. 2000;83(2):1031–8.
Kumar P, Prabhakar NR. Peripheral chemoreceptors: function and plasticity of the carotid body.
Compr Physiol. 2012;2(1):141–219.
Kumar GK, Nanduri J, Peng YJ, Prabhakar NR. Neuromolecular mechanisms mediating the effects
of chronic intermittent hypoxia on adrenal medulla. Respir Physiol Neurobiol. 2015;209:115–9.
Leal G, Comprido D, Duarte CB. BDNF-induced local protein synthesis and synaptic plasticity.
Neuropharmacology. 2014;76(Pt C):639–56.
Li Q, Sun B, Wang X, Jin Z, Zhou Y, Dong L, et al. A crucial role for hydrogen sulfide in oxygen
sensing via modulating large conductance calcium-activated potassium channels. Antioxid
Redox Signal. 2010;12(10):1179–89.
224 S. C. Veasey
Ling L, Fuller DD, Bach KB, Kinkead R, Olson EB Jr, Mitchell GS. Chronic intermittent hypoxia
elicits serotonin-dependent plasticity in the central neural control of breathing. J Neurosci.
2001;21(14):5381–8.
Liu Y, Ji ES, Xiang S, Tamisier R, Tong J, Huang J, et al. Exposure to cyclic intermittent hypoxia
increases expression of functional NMDA receptors in the rat carotid body. J Appl Physiol
(1985). 2009;106(1):259–67.
Makarenko VV, Nanduri J, Raghuraman G, Fox AP, Gadalla MM, Kumar GK, et al. Endogenous
H2S is required for hypoxic sensing by carotid body glomus cells. Am J Physiol Cell Physiol.
2012;303(9):C916–23.
Meyer U, Doelle B. Long-lasting shifts in ribosomal systems of hippocampal granular neurons due
to early postnatal hypoxia. J Hirnforsch. 1988;29(3):237–42.
Millhorn DE, Eldridge FL, Waldrop TG. Prolonged stimulation of respiration by a new central
neural mechanism. Respir Physiol. 1980;41(1):87–103.
Nagata N, Saji M, Ito T, Ikeno S, Takahashi H, Terakawa N. Repetitive intermittent hypoxia-
ischemia and brain damage in neonatal rats. Brain Dev. 2000;22(5):315–20.
Nair D, Dayyat EA, Zhang SX, Wang Y, Gozal D. Intermittent hypoxia-induced cognitive deficits
are mediated by NADPH oxidase activity in a murine model of sleep apnea. PLoS One. 2011;6
(5):e19847.
Nanduri J, Wang N, Yuan G, Khan SA, Souvannakitti D, Peng YJ, et al. Intermittent hypoxia
degrades HIF-2alpha via calpains resulting in oxidative stress: implications for recurrent apnea-
induced morbidities. Proc Natl Acad Sci U S A. 2009;106(4):1199–204.
Nichols NL, Dale EA, Mitchell GS. Severe acute intermittent hypoxia elicits phrenic long-term
facilitation by a novel adenosine-dependent mechanism. J Appl Physiol (1985). 2012;112(10):
1678–88.
Nieto FJ, Young TB, Lind BK, Shahar E, Samet JM, Redline S, et al. Association of sleep-
disordered breathing, sleep apnea, and hypertension in a large community-based study. Sleep
Heart Health Study. JAMA. 2000;283(14):1829–36.
Payne RS, Goldbart A, Gozal D, Schurr A. Effect of intermittent hypoxia on long-term potentiation
in rat hippocampal slices. Brain Res. 2004;1029(2):195–9.
Peng YJ, Prabhakar NR. Reactive oxygen species in the plasticity of respiratory behavior elicited by
chronic intermittent hypoxia. J Appl Physiol (1985). 2003;94(6):2342–9.
Peng YJ, Yuan G, Ramakrishnan D, Sharma SD, Bosch-Marce M, Kumar GK, et al. Heterozygous
HIF-1alpha deficiency impairs carotid body-mediated systemic responses and reactive oxygen
species generation in mice exposed to intermittent hypoxia. J Physiol. 2006;577(Pt 2):705–16.
Peng YJ, Yuan G, Khan S, Nanduri J, Makarenko VV, Reddy VD, et al. Regulation of hypoxia-
inducible factor-alpha isoforms and redox state by carotid body neural activity in rats. J Physiol.
2014;592(17):3841–58.
Piehl Aulin K, Svedenhag J, Wide L, Berglund B, Saltin B. Short-term intermittent normobaric
hypoxia—haematological, physiological and mental effects. Scand J Med Sci Sports. 1998;8(3):
132–7.
Pokala P, Llanera M, Sherwood J, Scharf S, Steinberg H. Erythropoietin response in subjects with
obstructive sleep apnea. Am J Respir Crit Care Med. 1995;151(6):1862–5.
Prabhakar NR, Semenza GL. Oxygen sensing and homeostasis. Physiology (Bethesda). 2015;30(5):
340–8.
Ramchandren S, Gruis KL, Chervin RD, Lisabeth LD, Concannon M, Wolfe J, et al. Hypoglossal
nerve conduction findings in obstructive sleep apnea. Muscle Nerve. 2010;42(2):257–61.
Remmers JE, deGroot WJ, Sauerland EK, Anch AM. Pathogenesis of upper airway occlusion
during sleep. J Appl Physiol Respir Environ Exerc Physiol. 1978;44(6):931–8.
Row BW, Kheirandish L, Neville JJ, Gozal D. Impaired spatial learning and hyperactivity in
developing rats exposed to intermittent hypoxia. Pediatr Res. 2002;52(3):449–53.
Saboisky JP, Stashuk DW, Hamilton-Wright A, Carusona AL, Campana LM, Trinder J, et al.
Neurogenic changes in the upper airway of patients with obstructive sleep apnea. Am J Respir
Crit Care Med. 2012;185(3):322–9.
11 Neural Injury in Models of Intermittent Hypoxia 225
Abbreviations
© Springer Nature B.V. and Shanghai Jiao Tong University Press 2022 229
A. I. Pack, Q. Y. Li (eds.), Sleep and its Disorders, Translational Medicine Research,
https://doi.org/10.1007/978-94-024-2168-2_12
230 F. L. Xiao et al.
GHB Gamma-hydroxybutyrate
HLA Human leukocyte antigen
LC Locus coeruleus
LDT Laterodorsal tegmental nucleus
MAOI Monoamine oxidase inhibitor
Orx1 Orexin-1
Orx2 Orexin-2
OSAHS Obstructive sleep apnea-hypopnea syndrome
PPT Pedunculopontine nucleus
SNRI Serotonin norepinephrine reuptake inhibitor
SSRI Selective serotonin reuptake inhibitor
TCA Tricyclic antidepressant
TMN Tuberomammillary nucleus
VLPO Ventrolateral preoptic nucleus
VTA Ventral tegmental area
The orexin system is conserved across many mammalian species. The orexin system
consists of two neuropeptides, orexin-A (or orexin-1) and orexin-B (or orexin-2),
and two G-protein-coupled receptors, orexin-1 (Orx1) and orexin-2 (Orx2) receptor.
In 1996, a set of neuropeptides associated with the secretin of hormones were
isolated from the rat lateral hypothalamus by the process of directional tag PCR
subtraction cloning (Gautvik et al. 1996). The cloning of the neuropeptides gene
from rat and mouse, the location of the peptide-generating cell bodies and a
description of their neuro-connections were first presented in 1997 (Ebrahim et al.
2002). In 1998, the discovery of hypocretin/orexin was reported independently by
two groups using different techniques from rat brain: de Lecea et al. (1998) identified
the pro-hormone pre-prohypocretin and its peptide products hypocretin-1 and -2 by
nucleotide sequencing (de Lecea et al. 1998); at the same time, Sakurai et al. (1998)
also reported orexin-1 and -2 by orphan receptor cloning. However, hypocretin and
orexin are proven to be synonymous. In this chapter we will use the term orexin.
pyroglutamyl residue and C-terminal amidation (Sakurai et al. 1998). The primary
structure of Orx1 predicted from the cDNA sequences is completely conserved
among different mammalian species (rat, cow, dog, pig, and human). Orx2 from
rat is a 28-amino acid, C-terminally amidated linear peptide of 2937 Da, which is
46% (13/28) same to the sequence of Orx1. The C-terminal half of Orx2 is very
similar to that of Orx1 (73%; 11/15), whereas the N-terminal half is variable. Orx2
also has a high degree of sequence conservation among species.
The two orexin receptors, Orx1 receptor (Orx1R) and Orx2 receptor (Orx2R)
from human brain, have an identical amino acid sequence of 64% (Tsujino and
Sakurai 2009). Amino acid identities between human and rat counterparts of each of
these receptors are 94% for Orx1R and 95% for Orx2R, suggesting that both receptor
genes are highly conserved among species (Sakurai et al. 1998). Orx1R has a greater
affinity for Orx1 than Orx2, whereas Orx2R has a similar affinity for both kinds of
Orexins (Sakurai et al. 1998) (Fig. 12.1). Studies in animal brain tissues indicate a
complexity in the signal transduction of Orexins (Kukkonen et al. 2002) including
the following: direct coupling to nonselective cationic channels (Brown et al. 2002;
Yang and Ferguson 2003); transient receptor potential channels (Sergeeva et al.
2003); electrogenic sodium–calcium exchangers (Eriksson et al. 2001; Wu et al.
2002); inhibition of GIRK channels that have previously been activated by somato-
statin; nociception or the mu-opioid agonist DAMGO (Hoang et al. 2003); and
unusual Ca2+-dependent signaling pathways associated with activation of mitogen-
activated protein kinase (Kukkonen et al. 2002; Lund et al. 2000), and/or
thapsigargin- and cAMP-PKA-sensitive pathways (Korotkova et al. 2002, 2003;
Yamanaka et al. 2003a). Furthermore, Orx1R is coupled to the Gq/11 class of G
protein, which lead to activation of phospholipase C with subsequent triggering of
232 F. L. Xiao et al.
In both human and rat brains, orexin-producing neurons are exclusively localized in
the perifornical area and the lateral and posterior hypothalamic area (Date et al.
1999). These cells diffusely project to the supra-tentorium structure (Fig. 12.2),
excluding the cerebellum (Peyron et al. 1998), which suggests that brain areas are
widely influenced by orexin neuronal activity. Moreover, the heaviest staining of
orexin-immunoreactive neuronal innervation was detected in the paraventricular
thalamus nucleus, arcuate nucleus of the hypothalamus, raphe nuclei, TMN, and
LC (Tsujino and Sakurai 2009).
In rat, the orexin-producing neurons are specific to the hypothalamus and have
widespread neuronal projections with the densest extra-hypothalamic projection to
the noradrenergic LC and lesser projections to the basal ganglia, thalamic regions,
the medullary reticular formation, and the nucleus of the solitary tract within the
central nervous system. Whereas, there are minor projections to cortical regions,
central and anterior amygdaloid nuclei, and the olfactory bulb (Gautvik et al. 1996;
Mignot 2000; Peyron et al. 1998). In humans, the distribution of orexin-producing
neurons is restricted to the dorsolateral hypothalamus with extensive dense pro-
jections to the LC, dorsal raphe nuclei, amygdala, suprachiasmatic nucleus, basal
12 Narcolepsy and Orexin/Hypocretin 233
Fig. 12.2 Projections of orexin-producing neurons in the brain, including the main monoaminergic
and cholinergic regions where orexin receptors are enriched. BF Basal forebrain, DR Dorsal raphe,
LC Locus coeruleus; LDT Laterodorsal tegmental nucleus, PPT Pedunculopontine tegmental
nucleus, SN Substantia nigra, TMN Tuberomamillary nucleus, VTA Ventral tegmental area
forebrain, brainstem, and spinal cord (Peyron et al. 2000; Thannickal et al. 2000; van
den Pol 1999). Orexin neurons are innervated by many neurons, including from the
lateral parabrachial nucleus, amygdala, BST, lateral septum, ventrolateral preoptic
nucleus (VLPO), medial and lateral preoptic areas, basal forebrain, posterior/
dorsomedial hypothalamus, VTA, and median raphe nuclei (Tsujino and Sakurai
2009). These neurons that project to orexin neurons are located in regions associated
with emotion (Tsujino and Sakurai 2009). Also, the orexin neurons are innervated
from the arcuate nucleus associated with energy homeostasis, namely by agouti-
related peptide and α-melanin-stimulating hormone-immunoreactive fibers
(Broberger et al. 1998).
Regulation of the sleep–wake cycle is a primary role of orexin but orexin also has
been identified as playing a role in various other functions including feeding and
energy homeostasis, neuroendocrine regulation, gastrointestinal and cardiovascular
system management, respiratory control, water equilibrium, and autonomic regula-
tion (Edwards et al. 1999; Haynes et al. 1999; Ida et al. 2000; Kirchgessner and Liu
1999; Kuru et al. 2000; Moriguchi et al. 1999; Sakurai et al. 1998; van den Pol et al.
1998). Orexin is involved in the control of other behaviors, such as reward-
associated behaviors and emotional processing (de Lecea and Huerta 2014; Ida
et al. 1999; Li et al. 2014). Metabolic rate can be increased by administration of
orexin in rats and hypoglycemia as a result of administration of insulin can excite up
to one-third of orexin-containing neurons. This infers that there is a participation of
orexin neurons in regulation of metabolism (Moriguchi et al. 1999; Samson and
Resch 2000; Sutcliffe and de Lecea 2000). With respect to neuroendocrine mecha-
nisms, orexin results in a decreased level of prolactin and growth hormone in plasma,
combined with an increased level of corticotropin, cortisol, insulin, and luteinizing
hormone (Ida et al. 2000; Sutcliffe and de Lecea 2000; van den Pol et al. 1998).
Administration of orexin in the central nervous system can improve water
12 Narcolepsy and Orexin/Hypocretin 235
Gélineáu first described the term “narcolepsy” in 1880 in a patient with excessive
daytime sleepiness, sleep attacks, and episodes of muscle weakness induced by
emotions (see Nishino 2007a). Narcolepsy is a chronic neurological condition but
is not a progressive disorder (Nishino 2007a). It affects approximately 1 in 2000
individuals in the United States (Mignot 1998). Males and females are both affected.
The onset of the disease usually occurs during adolescence. Most cases of human
narcolepsy are sporadic; while genetic and environmental factors are also significant
for the pathogenesis of narcolepsy, with a few familial cases of human narcolepsy
reported (Guilleminault et al. 1989).
12.2.1 Epidemiology
has been reported among Chinese cases (Dauvilliers et al. 2001). Although it was
thought originally that there was no obvious gender disparity in the prevalence of
narcolepsy, newer information indicate that men are more commonly affected, with
narcolepsy occurring 1.6 times more frequently than in women (Han et al. 2011;
Longstreth et al. 2007). Most cases of narcolepsy in humans are sporadic. Up to 5%
are familial cases, and the risk of a first-degree relative suffering narcolepsy-
cataplexy is 1–2%, which is 10–40 times higher than that for the general population
(Mignot 1998).
The classic tetrad of core symptoms for narcolepsy are excessive daytime sleepiness
(EDS), cataplexy, hypnagogic, and/or hypnopompic hallucination and sleep paral-
ysis, with disturbed nocturnal sleep. Not all symptoms may be present in an
individual patient, and the symptoms may vary in frequency and intensity over
time. EDS and cataplexy are the main symptoms of narcolepsy, with the main
complaint being EDS.
After disease onset, EDS persists throughout the patient’s life. It results in unwanted
episodes of sleep during monotonous sedentary activities and also under circum-
stances when the patients are involved in a task, such as eating, walking, talking,
cycling, or driving. As a result, patients with narcolepsy have a threefold increased
risk of motor vehicle accidents due to lapses in attention and lack of alertness
(Sakurai 2013). The EDS can be relieved by short naps (from a few seconds to
several minutes), but in most situations the sensation of waking up refreshed only
lasts from 1 h to several hours before the next feeling of sleepiness. That short naps
are refreshing is considered to be diagnostic (Nishino 2007a) and can be helpful in
differentiating narcolepsy from other causes of EDS. Sleepiness also happens in
irresistible waves in patients with narcolepsy, which is best called “sleep attacks.”
Sleep attacks may occur in very common situations, such as in the middle of a meal,
a conversation, or a bicycle ride. Patients may continue their activities in a semi-
conscious status with blurred writing or speaking, which is also described as
“automatic behavior” (Nishino 2007a). Abnormalities in learning and impaired
attention are frequently associated with narcolepsy, but objective psychophysiolog-
ical testing is generally normal (Rogers and Rosenberg 1990).
EDS is usually the first symptom of narcolepsy to appear, followed by the other
three symptoms (Nishino 2007a). The onset of cataplexy typically happens within
5 years following the presentation of EDS in about two-thirds of patients (Ohayon
et al. 2002). The mean duration of onset of hypnagogic and/or hypnopompic
12 Narcolepsy and Orexin/Hypocretin 237
hallucination and sleep paralysis is also 2–7 years after that of EDS (Kales et al.
1982).
12.2.2.2 Cataplexy
Sleep paralysis presents in 20–50% of all narcolepsy patients (Hublin et al. 1994),
and it is often associated with hypnagogic hallucination. Sleep paralysis is described
as an inability to carry out voluntary movements at the onset of sleep, or upon
awakening from sleep. The patients are unable to perform any movement, even a
simple action such as lifting a finger or breathing deeply. The patient is aware of this
condition and can recall it completely later. Sleep paralysis may continue for a few
minutes and is often interrupted by noise or other external stimulation. The symptom
is often troublesome in narcolepsy cases, particularly when accompanied by
238 F. L. Xiao et al.
Abnormal visual or auditory perceptions which happen at the time of falling asleep
(hypnagogic) or on waking up (hypnopompic) are often reported in cases with
narcolepsy (Goode 1962). These hallucinations are often unpleasant and are related
to an emotion of fear or threat (Dahlitz and Parkes 1993). Polysomnography studies
suggest that these hallucinations most often occur during REM sleep (Chetrit et al.
1994). The episodes are often difficult to distinguish from nightmares or unpleasant
dreams, which can also occur in narcolepsy.
Hypnagogic hallucinations are often related to sleep attacks, and the patients can
recall the content of the hallucinations. The hallucinations are usually complex,
vivid, and dream-mimic experiences, and may be followed by an attack of cataplexy
or sleep paralysis. Other common hallucinations observed at sleep onset include
primary feelings (i.e., light touching and rubbing), changes in location of body parts
(e.g., arm or leg). The hallucinations described in narcolepsy should be distinguished
from hallucinations reported in schizophrenia or other psychotic disorders. Patients
with narcolepsy reporting these hallucinations may be misdiagnosed.
however, that these metabolic disorders could be the result of orexin deficiency
(Nishino et al. 2001).
Narcolepsy interferes with every aspect of daily life. The negative social impact,
working disability, depression in mood have also been extensively described
(Aldrich 1989).
12.2.3 Diagnosis
one-third of mean normal values. Definite type-1 narcolepsy requires meeting the
items of A+B+C or A+B+D. Low levels of CSF orexin-1 are specific for type-1
narcolepsy and can confirm the diagnosis. Nocturnal polysomnography SOREMPs
can also be included in the total for SOREMPs.
Type-2 narcolepsy requires the followings: (A) excessive daytime sleepiness
lasting at least 3 months; (B) no history of cataplexy; (C) abnormal MSLT (mean
sleep latency 8 min and 2 or more SOREMPs); (D) CSF orexin-1 concentrations
>110 pg/mL, or greater than one-third of mean normal values; (E) excluding
hypersomnia due to other disorders and/or diseases resulting in abnormal MSLT.
The diagnosis of type-2 narcolepsy is controversial and requires meeting all of the
items discussed above (see Table 12.1).
Mignot and colleagues described that dogs with a mutation in the orexin-2
receptor are similar to humans with narcolepsy (Lin et al. 1999). As in human
narcolepsy, canine narcolepsy shows cataplexy, sleepiness (such as decreased
sleep latency) and SOREMPs (Nishino and Mignot 1997). The narcoleptic dogs
have fragmented sleep/wake patterns and they do not maintain long bouts of
wakefulness or sleep (Kaitin et al. 1986a, b; Nishino et al. 2000a). Narcoleptic
Dobermans showed decreased sleep latency and reduced latency to REM sleep
(Nishino et al. 2000a). Canine cataplexy is typically induced by playing with other
dogs or with human caretakers; presentation of food (especially favored food) is a
powerful precipitant. This has led to the use of food elicited cataplexy as a test for
narcolepsy in canine models. Sexual activity will often elicit cataplexy in male
narcoleptic canines (Boehmer et al. 2004; Nishino and Mignot 1997). Canines
with narcolepsy usually have about two episodes of cataplexy/hour during the day,
but in the Food Elicited Cataplexy Test, a narcoleptic dog can have five or more
events of cataplexy in just 1 or 2 min (Nishino et al. 1989, 2000a). Canine cataplexy
is not triggered by startling loud noises.
Cataplexy in dogs typically shows a progressive inhibition of muscle tone, with
the hindlimbs involved first (Fujiki et al. 2002). Hindlimb weakness often resolves in
a few seconds in the case of partial cataplexy, or it may spread to the forelimbs and
somatic muscles, resulting in generalized weakness (Scammell et al. 2009). In
narcoleptic dogs, all the limbs may collapse at the same time (Fujiki et al. 2002).
Even though the dog collapses to the ground, it may continue to eat, suggesting the
tone of the cranial muscles can be preserved. But, these cranial muscles might also
be fully atonic in cataplexy (Scammell et al. 2009). The eyes are open during the
cataplectic attacks, with eyes following objects of interest, indicating the preserva-
tion of consciousness during cataplexy in both human and dogs (Nishino et al.
1995). The duration of cataplexy can range from a few seconds to several minutes in
dogs (Nishino et al. 1989, 2000a). Longer durations may be reported in a situation
like REM sleep with closure of the eyes, rapid eye movements and distal muscle
twitching (Siegel et al. 1991).
Typical murine models of narcolepsy with cataplexy have been produced with
removal of the prepro-orexin gene or transgenic expression of a toxic protein which
selectively kills orexin-producing neurons (Chemelli et al. 1999; Hara et al. 2001).
As in human narcolepsy, murine narcolepsy shows cataplexy, impaired maintenance
of wakefulness and fragmented sleep (Chemelli et al. 1999; Hara et al. 2001). As a
mouse changes from normal activity into cataplexy, the EEG shifts from a waking
pattern to that for REM sleep or pre-REM sleep (Chemelli et al. 1999; Hara et al.
2001; Willie et al. 2003). Cataplexy in rodent is similar to human cataplexy
(Chemelli et al. 1999; Hara et al. 2001; Willie et al. 2003). Cataplexy is more
frequent with emotional stimulation, such as social interaction, enriched environ-
ment (novel toys, fresh bedding, etc.) and in the open field (Chemelli et al. 1999;
Espana et al. 2007). Mild fasting, restricted feeding programs, and presentation of
favored food can make the cataplectic attacks more frequent, but these situations are
not as effective as the Food Elicited Cataplexy Test, which is employed in identifi-
cation of canine cataplexy (Nishino and Mignot 1997; Scammell et al. 2009). Also,
12 Narcolepsy and Orexin/Hypocretin 243
In contrast to animal models, most human cases of narcolepsy are not due to
mutations in the orexin or orexin receptor genes (Han 2012b). The first link between
orexin dysfunction and narcolepsy in humans came from a clinical report of nine
human narcolepsy patients who were found to have very low levels of orexin-1 in the
cerebrospinal fluid (CSF) as compared with healthy subjects (Nishino et al. 2000b).
Seven of these narcoleptic subjects were reported to have undetectable orexin-1
levels, while two narcoleptic patients had normal levels of orexin-1. All the eight
healthy control subjects had normal levels of orexin-1. This result indicated that
human narcolepsy may be caused by a deficiency in orexin (Nishino et al. 2000b).
Subsequently, autopsy studies of human narcolepsy patients indicated a loss of
orexin peptides in the cortex and pons, with an 80–100% reduction in the number
of orexin-producing neurons in the hypothalamus (Peyron et al. 2000; Thannickal
et al. 2000). A possible explanation is that the orexin-producing neurons may be
destroyed by an autoimmune mechanism associated with the specific HLA genotype
in narcolepsy patients (Mignot 2014). There are just a hundred thousand orexin-
producing neurons located in the hypothalamus and a specific lesion in these cells
has not been reported in most other neurological disorders (Mignot 2014).
More studies have supported the concept of human narcolepsy is the direct result
of loss of orexin-producing neurons (Han 2012b). Peyron et al. reported a total loss
of orexin in the brains of six narcoleptic cases using in situ hybridization and
radioimmunoassay of the perifornical hypothalamus (Peyron et al. 2000). There
was no evidence of inflammatory pathology in all the six brains, albeit many years
after the initial presentation (Peyron et al. 2000). Another study demonstrated an
85–95% decline in the number of orexin-producing neurons in four narcoleptic
brains by using immunocytochemistry (Thannickal et al. 2000). Moreover, in both
studies, it was suggested that the melanin-concentrating hormone neurons, which are
situated close to the orexin-producing neurons in hypothalamus, were intact in the
narcoleptic brains. This suggested that the putative autoimmune process was rela-
tively specific for orexin-producing neurons (Peyron et al. 2000; Thannickal et al.
2000). Additional studies also found that loss of orexin cells in the hypothalamus,
rather than failure of orexin expression, was the main pathological mechanism
(Blouin et al. 2005; Crocker et al. 2005). Some studies also found an increased
244 F. L. Xiao et al.
A low CSF level of orexin-1 is now one of the diagnostic criteria for narcolepsy-
cataplexy according to the 3rd edition of the International Classification of Sleep
Disorders (ICSD-3) (2014). Narcolepsy-cataplexy is considered to be more closely
associated with orexin deficiency as compared with narcolepsy without cataplexy.
12 Narcolepsy and Orexin/Hypocretin 245
The HLA genes, also called major histocompatibility (MHC) genes, are located on
human chromosome 6. The HLA locus encompasses genes encoding for HLA class I
molecules (HLA-A, HLA-B, and HLA-C), which present antigenic peptides to the
T-cell receptors (TCR) at the surface of CD8+T cells. HLA class II molecules
(HLA-DR, HLA-DQ, and HLA-DP) present antigenic peptides to TCR on CD4+T
cells. HLA polymorphisms contribute to genetic variations in the immune response
(McDevitt and Tyan 1968). HLA polymorphisms regulate immune responses to
infections, but they are also associated with autoimmune disorders (Dausset 1972).
Over the past 30 years, a better understanding of the genetic basis of narcolepsy has
shown that narcolepsy is strongly associated with a specific HLA allele,
DQB1*0602. This polymorphism is consistently present in 90–100% of patients
across different ethnic groups (Mignot et al. 2001). As a result, it has long been
speculated that the pathogenesis of narcolepsy results from an autoimmune-
mediated mechanism (Kadotani et al. 1998). The identification of the Tribbles
homolog 2 (Trib2, an antigen abundantly expressed on orexin-producing neurons)
reactive antibodies (Cvetkovic-Lopes et al. 2010); the association of polymorphisms
in the T-cell receptor alpha locus and the purinergic receptor subtype 2Y11
(P2RY11) loci found in genome-wide association studies (Hallmayer et al. 2009;
Han et al. 2012a; Kornum et al. 2011b) all add further support for the proposed
autoimmune mechanism.
The first report about HLA relationship with narcolepsy was from Juli and Honda,
they reported that all Japanese narcolepsy-cataplexy cases (22 male and 18 female
patients) included in the study carried HLA-DR2 gene, while only 49.1% control
subjects carried HLA-DR2 gene, indicating an absolute association with HLA-DR2
in the Japanese narcolepsy patients (Juji et al. 1984). Guilleminault and his
246 F. L. Xiao et al.
colleagues, however, found that very rare DR2-negative narcolepsy patients were
found in American (Guilleminault and Grumet 1986). This is in conflict with
Japanese research described above (Juji et al. 1984). The possible explanation for
this conflict may be the ethnic difference in linkage disequilibrium between DR2 and
other narcolepsy-associated genes. On the other hand, diagnostic criteria also
affected the findings of association between HLA-DR2 and narcolepsy (Matsuki
et al. 1987). In German Caucasoids narcolepsy patients, Gertrud et al. found that
57 out of 58 unrelated patients (98.3%) carried HLA-DR2 and DQw1 (Mueller-
Eckhardt et al. 1986), which was demonstrated as the subtype of HLADQ0602 by
high resolution analysis. While the HLA-DR and HLA-DQ region is compact,
containing in sequence the DRA gene (practically monomorphic), accessory DRB
genes (DRB3,4,5 genes present in some but not all haplotypes), the DRB1 gene
(a very polymorphic gene), and finally the polymorphic DQA1 and DQB1 loci. In
Caucasians and Japanese, linkage disequilibrium between DQ and DR is remarkable
such that almost all DQB1*06:02 alleles are linked with DQA1*01:02 and
DRB1*15:01 (DR2), making it difficult to distinguish whether the effect is caused
by DR or DQ (Mignot 2014). In the early 1990s, studies in African Americans
showed additional diversity in DR-DQ haplotypes, such that DQB1*06:02 was
detected not only in linkage with DRB1*15:01, but also in linkage with DRB1*11:
01 and more rarely DRB1*12:02 (Behar et al. 1995; Fernandez-Vina et al. 1991;
Mignot et al. 1997). There are less DR2 carriers in African American narcoleptic
cases (Neely et al. 1986), and the detection of HLA-DR and HLA-DQ associations
in this ethnic group seem to be significant (Neely et al. 1986).
Mignot and his colleagues reported that all African American patients with
narcolepsy carried both DQA1*01:02 and DQB1*06:02 (Matsuki et al. 1992; Mignot
et al. 1994); while in many instances, DRB1*15 (DR2) was not detected. This
demonstrates that the primary association is with HLA-DQ, not DR, and more
particularly with the DQ heterodimer DQ0602 that encodes DQA1*01:02 and
DQB1*06:02. This was replicated in Chinese patients with narcolepsy, we found
that DRB1*15:01 alone does not predispose to narcolepsy in the context of the
DRB1*15:01, DQA1*01:02, and DQB1*06:01 in South China (Han et al. 2012b).
The data illustrated the extraordinary conservation of HLA class II effects in
narcolepsy across populations and show that DRB1*15:01 has no effect on narco-
lepsy susceptibility in the absence of DQB1*06:02 (Han et al. 2012b). In follow-up
studies, Mignot and his colleagues found a number of very rare cases of DQB1*06:
02-positive cases that carried various DRB1 alleles by screening hundreds of
patients. Alleles such as DRB1*03:01, DRB1*08:01, DRB1*08:06, and DRB1*16:
01 are only exceptionally detected in control subjects (Mignot et al. 1993, 1997),
confirming the abundance of these rare DQ0602 haplotypes in narcolepsy. Other
HLA alleles genes encoding MHC I molecules can also predispose individuals to
narcolepsy, including HLA-A*11:01, HLA-B*35:01, HLA-B*51:01, and
HLA-C*04:01 (Tafti et al. 2016). Current findings suggest antigen presentation by
the heterodimer DQ0602 (MHC II molecule) to T cells may be central to the
pathogenesis of narcolepsy (Latorre et al. 2018; Luo et al. 2018; Irukayama-Tomobe
et al. 2017).
12 Narcolepsy and Orexin/Hypocretin 247
As described above, studies have demonstrated that the best genetic marker for
narcolepsy is the closely linked HLA-DQB1*06:01 and DQA1*01:02 loci (encoding
the DQ0602 heterodimer) rather than HLA-DR2.
The hypothalamus, in which orexin-producing neurons are located, has long been
considered as being involved in respiratory regulation (Kuwaki 2010). Recent
studies indicate that this effect is partially mediated by the orexin system. Orexin-
producing neurons may participate in breathing control with the changing of con-
sciousness. Neuronal projections from orexin-producing neurons include ascending
pathways to arousal regulation regions such as the thalamus and cortex, and
descending pathways to brainstem regulating nuclei, such as the raphe nuclei,
nucleus tractus solitarius, the rostral ventrolateral medulla as well as to phrenic
and hypoglossal nuclei (Kuwaki 2010; Williams and Burdakov 2008). Both the
Orx2 receptor and the Orx1 receptor are expressed in brainstem respiratory centers,
and the orexin-producing neurons also serve as acid and CO2 sensors, as well as
receiving afferent signals from amygdala and the bed nucleus of the stria terminals
(Williams and Burdakov 2008). Activation of orexin receptors at different locations
in the brainstem and spinal cord can influence the ventilatory rate and depth, as well
as the coordination between upper airway and thoracic pump muscles.
Microperfusion of orexin either to sites in the pre-Bötzinger region in the medulla
or phrenic nucleus leads to a dose-dependent, a significant increase in diaphragm
electromyographic activity (Nattie and Li 2010). Injection of orexin into the
248 F. L. Xiao et al.
who carry the HLADQB1*06:02, the same abnormal hypoxic response could also be
found as in narcoleptic patients with HLADQB1*06:02 (Han 2012b). However,
since the pathogenesis of narcolepsy is an autoimmune disorder, it has been reported
that HLADQB1*06:02 associated immune-mediated destruction of type I glomus
cells in the carotid bodies (the peripheral chemoreceptor for detection of hypoxia)
might contribute to the abnormal hypoxic response in human narcolepsy (Kornum
et al. 2011a).
Animal models, especially canine models, have been used to understand the neuro-
nal mechanisms that underlie the pharmacological control of narcolepsy (Nishino
2007a). The cholinergic system, which plays a significant role in triggering REM
sleep and REM sleep atonia, was identified as an investigatory target (Nishino
2007b). Although cholinergic blockade with muscarinic antagonists could signifi-
cantly reduce cataplexy in canine models, this class of compounds has not been used
in human narcolepsy due to their obvious peripheral side effects (Nishino 2007a).
Treatment of narcolepsy is focused on control of the two core symptoms, excessive
daytime sleepiness, and cataplexy. Recently, disturbed nocturnal sleep is increas-
ingly recognized as an important symptom of narcolepsy. Generally speaking, all
therapeutic agents used to treat cataplexy are aimed to act on the monoaminergic
system (Nishino 2007a). Wakefulness-stimulant drugs such as modafinil and
amphetamine are used to treat excessive daytime sleepiness (Sakurai 2013). Modu-
lation of γ-aminobutyric acid B (GABAB) receptors or histamine H3 receptors (H3R)
has effects on both EDS and cataplexy (Thorpy 2020). Pitolisant, an H3R antagonist,
and solriamfetol, a dopamine and norepinephrine reuptake inhibitor, are the most
recently approved treatments for narcolepsy EDS in the European Union (pitolisant)
and the United States (pitolisant and solriamfetol) (Thorpy 2020). Several new
agents are being developed and tested as potential treatments for EDS and cataplexy
in narcolepsy (Thorpy 2020), including novel oxybate formulations (once-nightly
[FT218]; low sodium [JZP-258]), a selective norepinephrine reuptake inhibitor
(AXS-12), and a product combining modafinil and an astroglial connexin inhibitor
(THN102). Continuous deep brain stimulation to lateral hypothalamus and zona
incerta dose-responsively reversed sleep and cataplexy episodes in narcolepsy
mouse model without negative sequelae (Rogers et al. 2020). Since the finding of
association between Orexin/hypocretin and narcolepsy, this system has been a target
for therapy. Previous studies showed that central administration of orexin can
ameliorate cataplexy and improve the wakefulness in narcolepsy animals (Zeitzer
et al. 2006), which demonstrated that deficiency of orexin does not induce a
permanent inability in orexin system. Thus, pharmacological modulation aims to
the orexin system and sets an example for the ideal approach to narcolepsy treat-
ment. Promising orexin-based narcolepsy treatment includes orexin administration
via different pathways (intravenous, intranasal, intra-cerebral ventricle), gene
250 F. L. Xiao et al.
In narcolepsy mice with orexin gene knockout, orexin gene transfer to neurons in
the lateral hypothalamus using replication-defective herpes simplex virus 1 (HSV-1)
reduces cataplexy and restores normal sleep–wake cycle during the 4-day lifetime of
the vector (Liu et al. 2008). However, unlike those in orexin gene knockout mice,
orexin neurons are missing in human narcolepsy patients as well as in another animal
model, orexin-ataxin-3 mice (Hara et al. 2001). Orexin neurons project to wide areas
of the brain (Peyron et al. 1998), including the dorsal pons, which involve in the
maintenance of muscle tone during waking. Indeed, transfer of the orexin gene to
surrogate neurons in the dorsal pons using an adeno-associated virus (AAV) vector
decreased cataplexy and restored normal sleep–wake cycle 3 weeks after injection in
orexin gene knockout mice (Blanco-Centurion et al. 2013). More specifically,
restoration of orexin receptor expression in the serotonergic neurons in dorsal
raphe nuclei decreased cataplexy and orexin receptor expression in noradrenergic
neurons in the locus coeruleus consolidated sleep fragmentation in orexin receptor
knockout mice (Hasegawa et al. 2014).
Cell Transplantation
It is not known in which brain regions the loss of orexin signaling contributes most
significantly to narcolepsy. Some studies suggest that orexin innervation deficiency
in pontine reticular formation plays a crucial role in the development of narcolepsy
(Blanco-Centurion et al. 2004). Orexin neurons grafted onto the pons can survive
most likely because the pons is mainly innervated by orexin neurons and secretes
factors stimulating axonal growth in orexin neurons (Peyron et al. 1998). The main
interest is now focused on the derivation of orexin neuroblasts for transplantation
from stem cells. This concept can promote higher rates of graft survival and
functional integration into host brain tissue.
Once the orexin neuron survival rate problem is resolved, the question of whether
transplanted orexin neurons indeed restore sleep–wake cycle in narcolepsy animal
models will remain. Arias-Carrion and Murillo-Rodriguez reported the first evidence
that transplantation of orexin neurons into the lateral hypothalamus diminishes
narcolepsy-like behaviors in narcolepsy rats (Arias-Carrion and Murillo-Rodriguez
2014).
Orexin Receptor Agonists
Nonpeptide orexin receptor agonists, currently under development, may be promis-
ing candidates for treating narcolepsy. Peripheral administration of YNT-185, a
nonpeptide orexin-2 receptor agonist, significantly ameliorates the narcolepsy symp-
toms in narcolepsy model mice (Irukayama-Tomobe et al. 2017). YNT-185 also
improves wakefulness in wild-type mice, suggesting that orexin receptor agonists
may be useful for treating sleepiness due to other disorders (Irukayama-Tomobe
et al. 2017). However, YNT-185 was limited in vivo efficacy and appears not
suitable for further clinical development. A second orexin 2 receptor-selective
agonist, TAK-925, when injected intravenously showed robust wake-promoting
effects in wild-type mice and nonhuman primates (marmosets and cynomolgus
monkeys), and increased wakefulness time and completely removed daytime sleep-
iness and cataplexy in orexin deficiency narcolepsy mice (Yukitake et al. 2019).
252 F. L. Xiao et al.
Preliminary data also showed that TAK-925 attenuated the body weight gain in
orexin/ataxin-3 mice (another narcolepsy animal model) without changing the daily
food intake (Yukitake et al. 2019). These results persisted after 14 days of systemic
administration, favoring that, TAK-925 may treat a broad range of narcolepsy
symptoms without causing orexin receptor desensitization. New promising
nonpeptide orexin receptor agonists are also currently under development. In the
future, the use of orexin 2 receptor agonists as efficient stimulants could also be of
interest in decreasing daytime sleepiness in patients with type 2 narcolepsy and
idiopathic hypersomnia, conditions with normal CSF orexin levels.
Immune-Based Therapy
Current treatment for narcolepsy is only symptomatic based on our understanding of
the neuro-pathophysiology of narcolepsy. However, deficiency in orexin resulting
from a possible autoimmune mechanism is the core pathophysiology of narcolepsy.
So far, in animal models, orexin peptides or orexin-producing neuronal transplanta-
tion are orexin-based therapies for a new approach to treatment of narcolepsy.
Immune-based therapy, such as intravenous immunoglobulins (IVIgs), cortico-
steroids, and plasmapheresis, have been tested in several cases given the role of
autoimmune mechanisms (Dauvilliers et al. 2004; Lecendreux et al. 2003). If applied
at disease onset, immune-based therapy might modify the course of narcolepsy but
delays in recognition of the disease limit this approach. Attempts to use immune-
based therapy in narcolepsy patients have been reported and summarized (Barateau
and Dauvilliers 2019), but they are all uncontrolled case studies, with very small
numbers of involved patients. IVIgs were usually evaluated, possibly due to the
good efficacy and tolerability in other autoimmune disorders. One adult patient
received IVIg treatment 15 days after narcolepsy onset and completely reversed
EDS and cataplexy, with normalized orexin levels in CSF (Dauvilliers et al. 2009).
But the difficulty is to administer the treatment at very early narcolepsy onset and the
destruction of orexin neurons may still be reversed at that period. So, there is no
current evidence to guide these immunomodulatory treatments in narcolepsy. Other
innovative immune-based treatments in narcolepsy were proposed: natalizumab,
fingolimod, abatacept, monoclonal antibodies targeting T or B cells, tumor necrosis
factor alpha blockers, anakinra, antigen-specific therapies, or cyclophosphamide
(Barateau et al. 2017). But these medications may also have the risk of serious
side effects. In future clinical trials, immune-based drugs should be given to highly
selected narcolepsy patients: these patients should have an ongoing autoimmune
response (Barateau and Dauvilliers 2019). More randomized controlled trials and
animal model studies are being conducted to study the benefits of immune-based
therapy in narcolepsy.
12 Narcolepsy and Orexin/Hypocretin 253
12.4 Conclusion
This is an exciting time for narcolepsy research. Over the last two decades, we have
learned much about the pathogenesis of the condition and the key role that orexin
plays. Animals models—both in dogs and mice—have played a major role in new
discoveries. While we have not yet reached the goal of orexin replacement in
humans, there have been extensive clinical trials with respect to medications to
combat excessive sleepiness and cataplexy. Thus, the current treatment of narco-
lepsy has a much more solid scientific base. We can anticipate more progress in the
future that should avoid the problem of cases of the disorder going for years
unrecognized and untreated.
References
Aldrich MS. Automobile accidents in patients with sleep disorders. Sleep. 1989;12:487–94.
American Academy of Sleep Medicine. The international classification of sleep disorders.
Westchester: American Academy of Sleep Medicine; 2014.
Arias-Carrion O, Murillo-Rodriguez E. Effects of hypocretin/orexin cell transplantation on
narcoleptic-like sleep behavior in rats. PLoS One. 2014;9:e95342.
Barateau L, Dauvilliers Y. Recent advances in treatment for narcolepsy. Ther Adv Neurol Disord.
2019;12:1756286419875622.
Barateau L, Liblau R, Peyron C, Dauvilliers Y. Narcolepsy type 1 as an autoimmune disorder:
evidence, and implications for pharmacological treatment. CNS Drugs. 2017;31:821–34.
Behar E, Lin X, Grumet FC, Mignot E. A new DRB1*1202 allele (DRB1*12022) found in
association with DQA1*0102 and DQB1*0602 in two black narcoleptic subjects. Immunoge-
netics. 1995;41:52.
Beuckmann CT, Sinton CM, Williams SC, Richardson JA, Hammer RE, Sakurai T, Yanagisawa
M. Expression of a poly-glutamine-ataxin-3 transgene in orexin neurons induces narcolepsy-
cataplexy in the rat. J Neurosci. 2004;24:4469–77.
Blanco-Centurion C, Gerashchenko D, Salin-Pascual RJ, Shiromani PJ. Effects of hypocretin2-
saporin and antidopamine-beta-hydroxylase-saporin neurotoxic lesions of the dorsolateral pons
on sleep and muscle tone. Eur J Neurosci. 2004;19:2741–52.
Blanco-Centurion C, Liu M, Konadhode R, Pelluru D, Shiromani PJ. Effects of orexin gene transfer
in the dorsolateral pons in orexin knockout mice. Sleep. 2013;36:31–40.
Blouin AM, Thannickal TC, Worley PF, Baraban JM, Reti IM, Siegel JM. Narp immunostaining of
human hypocretin (orexin) neurons: loss in narcolepsy. Neurology. 2005;65:1189–92.
Boehmer LN, Wu MF, John J, Siegel JM. Treatment with immunosuppressive and anti-
inflammatory agents delays onset of canine genetic narcolepsy and reduces symptom severity.
Exp Neurol. 2004;188:292–9.
Bourgin P, Zeitzer JM, Mignot E. CSF hypocretin-1 assessment in sleep and neurological disorders.
Lancet Neurol. 2008;7:649–62.
Broberger C, De Lecea L, Sutcliffe JG, Hokfelt T. Hypocretin/orexin- and melanin-concentrating
hormone-expressing cells form distinct populations in the rodent lateral hypothalamus: rela-
tionship to the neuropeptide Y and agouti gene-related protein systems. J Comp Neurol.
1998;402:460–74.
Broughton R, Dunham W, Newman J, Lutley K, Duschesne P, Rivers M. Ambulatory 24 hour
sleep-wake monitoring in narcolepsy-cataplexy compared to matched controls.
Electroencephalogr Clin Neurophysiol. 1988;70:473–81.
254 F. L. Xiao et al.
Brown RE, Sergeeva OA, Eriksson KS, Haas HL. Convergent excitation of dorsal raphe serotonin
neurons by multiple arousal systems (orexin/hypocretin, histamine and noradrenaline). J
Neurosci. 2002;22:8850–9.
Carskadon MA, Dement WC, Mitler MM, Roth T, Westbrook PR, Keenan S. Guidelines for the
multiple sleep latency test (MSLT): a standard measure of sleepiness. Sleep. 1986;9:519–24.
Chemelli RM, Willie JT, Sinton CM, Elmquist JK, Scammell T, Lee C, Richardson JA, Williams
SC, Xiong Y, Kisanuki Y, Fitch TE, Nakazato M, Hammer RE, Saper CB, Yanagisawa
M. Narcolepsy in orexin knockout mice: molecular genetics of sleep regulation. Cell.
1999;98:437–51.
Chervin RD, Kraemer HC, Guilleminault C. Correlates of sleep latency on the multiple sleep
latency test in a clinical population. Electroencephalogr Clin Neurophysiol. 1995;95:147–53.
Chetrit M, Besset A, Damci D, Lelarge C, Billiard M. Hypnogogic hallucinations associated with
sleep onset REM period in narcolepsy-cataplexy. J Sleep Res. 1994;3:43.
Chokroverty S. Sleep apnea in narcolepsy. Sleep. 1986;9:250–3.
Crocker A, Espana RA, Papadopoulou M, Saper CB, Faraco J, Sakurai T, Honda M, Mignot E,
Scammell TE. Concomitant loss of dynorphin, NARP, and orexin in narcolepsy. Neurology.
2005;65:1184–8.
Cvetkovic-Lopes V, Bayer L, Dorsaz S, Maret S, Pradervand S, Dauvilliers Y, Lecendreux M,
Lammers GJ, Donjacour CE, Du Pasquier RA, Pfister C, Petit B, Hor H, Muhlethaler M, Tafti
M. Elevated Tribbles homolog 2-specific antibody levels in narcolepsy patients. J Clin Invest.
2010;120:713–9.
Dahlitz M, Parkes JD. Sleep paralysis. Lancet. 1993;341:406–7.
Date Y, Ueta Y, Yamashita H, Yamaguchi H, Matsukura S, Kangawa K, Sakurai T, Yanagisawa M,
Nakazato M. Orexins, orexigenic hypothalamic peptides, interact with autonomic, neuroendo-
crine and neuroregulatory systems. Proc Natl Acad Sci U S A. 1999;96:748–53.
Dausset J. Correlation between histocompatibility antigens and susceptibility to illness. Prog Clin
Immunol. 1972;1:183–210.
Dauvilliers Y. Differential diagnosis in hypersomnia. Curr Neurol Neurosci Rep. 2006;6:156–62.
Dauvilliers Y, Montplaisir J, Molinari N, Carlander B, Ondze B, Besset A, Billiard M. Age at onset
of narcolepsy in two large populations of patients in France and Quebec. Neurology. 2001;57:
2029–33.
Dauvilliers Y, Carlander B, Rivier F, Touchon J, Tafti M. Successful management of cataplexy with
intravenous immunoglobulins at narcolepsy onset. Ann Neurol. 2004;56:905–8.
Dauvilliers Y, Arnulf I, Mignot E. Narcolepsy with cataplexy. Lancet. 2007;369:499–511.
Dauvilliers Y, Abril B, Mas E, Michel F, Tafti M. Normalization of hypocretin-1 in narcolepsy after
intravenous immunoglobulin treatment. Neurology. 2009;73:1333–4.
de Lecea L, Huerta R. Hypocretin (orexin) regulation of sleep-to-wake transitions. Front Pharmacol.
2014;5:16.
de Lecea L, Kilduff TS, Peyron C, Gao X, Foye PE, Danielson PE, Fukuhara C, Battenberg EL,
Gautvik VT, Bartlett FS 2nd, Frankel WN, van den Pol AN, Bloom FE, Gautvik KM, Sutcliffe
JG. The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity. Proc Natl
Acad Sci U S A. 1998;95:322–7.
Deadwyler SA, Porrino L, Siegel JM, Hampson RE. Systemic and nasal delivery of orexin-A
(Hypocretin-1) reduces the effects of sleep deprivation on cognitive performance in nonhuman
primates. J Neurosci. 2007;27:14239–47.
Dhuria SV, Hanson LR, Frey WH 2nd. Intranasal drug targeting of hypocretin-1 (orexin-A) to the
central nervous system. J Pharm Sci. 2009;98:2501–15.
Dube MG, Kalra SP, Kalra PS. Food intake elicited by central administration of orexins/
hypocretins: identification of hypothalamic sites of action. Brain Res. 1999;842:473–7.
Ebrahim IO, Howard RS, Kopelman MD, Sharief MK, Williams AJ. The hypocretin/orexin system.
J R Soc Med. 2002;95:227–30.
12 Narcolepsy and Orexin/Hypocretin 255
Edwards CM, Abusnana S, Sunter D, Murphy KG, Ghatei MA, Bloom SR. The effect of the orexins
on food intake: comparison with neuropeptide Y, melanin-concentrating hormone and galanin. J
Endocrinol. 1999;160:R7–12.
Eriksson KS, Sergeeva O, Brown RE, Haas HL. Orexin/hypocretin excites the histaminergic
neurons of the tuberomammillary nucleus. J Neurosci. 2001;21:9273–9.
Espana RA, McCormack SL, Mochizuki T, Scammell TE. Running promotes wakefulness and
increases cataplexy in orexin knockout mice. Sleep. 2007;30:1417–25.
Fernandez-Vina M, Moraes JR, Moraes ME, Miller S, Stastny P. HLA class II haplotypes in
Amerindians and in black North and South Americans. Tissue Antigens. 1991;38:235–7.
Fujiki N, Morris L, Mignot E, Nishino S. Analysis of onset location, laterality and propagation of
cataplexy in canine narcolepsy. Psychiatry Clin Neurosci. 2002;56:275–6.
Fujiki N, Yoshida Y, Ripley B, Mignot E, Nishino S. Effects of IV and ICV hypocretin-1 (orexin A)
in hypocretin receptor-2 gene mutated narcoleptic dogs and IV hypocretin-1 replacement
therapy in a hypocretin-ligand-deficient narcoleptic dog. Sleep. 2003;26:953–9.
Fukuda K, Miyasita A, Inugami M, Ishihara K. High prevalence of isolated sleep paralysis:
kanashibari phenomenon in Japan. Sleep. 1987;10:279–86.
Gautvik KM, de Lecea L, Gautvik VT, Danielson PE, Tranque P, Dopazo A, Bloom FE, Sutcliffe
JG. Overview of the most prevalent hypothalamus-specific mRNAs, as identified by directional
tag PCR subtraction. Proc Natl Acad Sci U S A. 1996;93:8733–8.
Goode GB. Sleep paralysis. Arch Neurol. 1962;6:228–34.
Grivel J, Cvetkovic V, Bayer L, Machard D, Tobler I, Muhlethaler M, Serafin M. The wake-
promoting hypocretin/orexin neurons change their response to noradrenaline after sleep depri-
vation. J Neurosci. 2005;25:4127–30.
Guilleminault C, Grumet C. HLA-DR2 and narcolepsy: not all narcoleptic-cataplectic patients are
DR2. Hum Immunol. 1986;17:1–2.
Guilleminault C, Mignot E, Grumet FC. Familial patterns of narcolepsy. Lancet. 1989;2:1376–9.
Gulyani S, Wu MF, Nienhuis R, John J, Siegel JM. Cataplexy-related neurons in the amygdala of
the narcoleptic dog. Neuroscience. 2002;112:355–65.
Hagan JJ, Leslie RA, Patel S, Evans ML, Wattam TA, Holmes S, Benham CD, Taylor SG,
Routledge C, Hemmati P, Munton RP, Ashmeade TE, Shah AS, Hatcher JP, Hatcher PD,
Jones DN, Smith MI, Piper DC, Hunter AJ, Porter RA, Upton N. Orexin A activates locus
coeruleus cell firing and increases arousal in the rat. Proc Natl Acad Sci U S A. 1999;96:10911–
6.
Hallmayer J, Faraco J, Lin L, Hesselson S, Winkelmann J, Kawashima M, Mayer G, Plazzi G,
Nevsimalova S, Bourgin P, Hong SC, Honda Y, Honda M, Hogl B, Longstreth WT Jr,
Montplaisir J, Kemlink D, Einen M, Chen J, Musone SL, Akana M, Miyagawa T, Duan J,
Desautels A, Erhardt C, Hesla PE, Poli F, Frauscher B, Jeong JH, Lee SP, Ton TG, Kvale M,
Kolesar L, Dobrovolna M, Nepom GT, Salomon D, Wichmann HE, Rouleau GA, Gieger C,
Levinson DF, Gejman PV, Meitinger T, Young T, Peppard P, Tokunaga K, Kwok PY, Risch N,
Mignot E. Narcolepsy is strongly associated with the T-cell receptor alpha locus. Nat Genet.
2009;41:708–11.
Han F. Respiratory regulation in narcolepsy. Sleep Breath. 2012a;16:241–5.
Han F. Sleepiness that cannot be overcome: narcolepsy and cataplexy. Respirology. 2012b;17:
1157–65.
Han F, Chen E, Wei H, He Q, Ding D, Strohl KP. Treatment effects on carbon dioxide retention in
patients with obstructive sleep apnea-hypopnea syndrome. Chest. 2001;119:1814–9.
Han F, Mignot E, Wei YC, Dong SX, Li J, Lin L, An P, Wang LH, Wang JS, He MZ, Gao HY,
Li M, Gao ZC, Strohl KP. Ventilatory chemoresponsiveness, narcolepsy-cataplexy and human
leukocyte antigen DQB1*0602 status. Eur Respir J. 2010;36:577–83.
Han F, Lin L, Li J, Aran A, Dong SX, An P, Zhao L, Li M, Li QY, Yan H, Wang JS, Gao HY, Li M,
Gao ZC, Strohl KP, Mignot E. Presentations of primary hypersomnia in Chinese children. Sleep.
2011;34:627–32.
256 F. L. Xiao et al.
Han F, Lin L, Li J, Aran A, Dong SX, An P, Zhao L, Li QY, Yan H, Wang JS, Gao HY, Li M, Gao
ZC, Strohl KP, Mignot E. TCRA, P2RY11, and CPT1B/CHKB associations in Chinese
narcolepsy. Sleep Med. 2012a;13:269–72.
Han F, Lin L, Li J, Dong SX, An P, Zhao L, Liu NY, Li QY, Yan H, Gao ZC, Faraco J, Strohl KP,
Liu X, Miyadera H, Mignot E. HLA-DQ association and allele competition in Chinese narco-
lepsy. Tissue Antigens. 2012b;80:328–35.
Hara J, Beuckmann CT, Nambu T, Willie JT, Chemelli RM, Sinton CM, Sugiyama F, Yagami K,
Goto K, Yanagisawa M, Sakurai T. Genetic ablation of orexin neurons in mice results in
narcolepsy, hypophagia, and obesity. Neuron. 2001;30:345–54.
Hasegawa E, Yanagisawa M, Sakurai T, Mieda M. Orexin neurons suppress narcolepsy via
2 distinct efferent pathways. J Clin Invest. 2014;124:604–16.
Haynes AC, Jackson B, Overend P, Buckingham RE, Wilson S, Tadayyon M, Arch JR. Effects of
single and chronic intracerebroventricular administration of the orexins on feeding in the rat.
Peptides. 1999;20:1099–105.
Hishikawa Y, Wakamatsu H, Furuya E, Sugita Y, Masaoka S. Sleep satiation in narcoleptic
patients. Electroencephalogr Clin Neurophysiol. 1976;41:1–18.
Hoang QV, Bajic D, Yanagisawa M, Nakajima S, Nakajima Y. Effects of orexin (hypocretin) on
GIRK channels. J Neurophysiol. 2003;90:693–702.
Honda Y, Doi Y, Ninomiya R, Ninomiya C. Increased frequency of non-insulin-dependent diabetes
mellitus among narcoleptic patients. Sleep. 1986;9:254–9.
Horvath TL, Peyron C, Diano S, Ivanov A, Aston-Jones G, Kilduff TS, van Den Pol
AN. Hypocretin (orexin) activation and synaptic innervation of the locus coeruleus noradren-
ergic system. J Comp Neurol. 1999;415:145–59.
Hublin C, Kaprio J, Partinen M, Koskenvuo M, Heikkila K, Koskimies S, Guilleminault C. The
prevalence of narcolepsy: an epidemiological study of the Finnish Twin Cohort. Ann Neurol.
1994;35:709–16.
Ida T, Nakahara K, Katayama T, Murakami N, Nakazato M. Effect of lateral cerebroventricular
injection of the appetite-stimulating neuropeptide, orexin and neuropeptide Y, on the various
behavioral activities of rats. Brain Res. 1999;821:526–9.
Ida T, Nakahara K, Kuroiwa T, Fukui K, Nakazato M, Murakami T, Murakami N. Both cortico-
tropin releasing factor and neuropeptide Y are involved in the effect of orexin (hypocretin) on
the food intake in rats. Neurosci Lett. 2000;293:119–22.
Irukayama-Tomobe Y, Ogawa Y, Tominaga H, Ishikawa Y, Hosokawa N, Ambai S, Kawabe Y,
Uchida S, Nakajima R, Saitoh T, Kanda T, Vogt K, Sakurai T, Nagase H, Yanagisawa
M. Nonpeptide orexin type-2 receptor agonist ameliorates narcolepsy-cataplexy symptoms in
mouse models. Proc Natl Acad Sci U S A. 2017;114:5731–6.
John J, Wu MF, Siegel JM. Systemic administration of hypocretin-1 reduces cataplexy and
normalizes sleep and waking durations in narcoleptic dogs. Sleep Res Online. 2000;3:23–8.
John J, Thannickal TC, McGregor R, Ramanathan L, Ohtsu H, Nishino S, Sakai N, Yamanaka A,
Stone C, Cornford M, Siegel JM. Greatly increased numbers of histamine cells in human
narcolepsy with cataplexy. Ann Neurol. 2013;74:786–93.
Johnson PL, Truitt W, Fitz SD, Minick PE, Dietrich A, Sanghani S, Traskman-Bendz L, Goddard
AW, Brundin L, Shekhar A. A key role for orexin in panic anxiety. Nat Med. 2010;16:111–5.
Juji T, Satake M, Honda Y, Doi Y. HLA antigens in Japanese patients with narcolepsy. All the
patients were DR2 positive. Tissue Antigens. 1984;24:316–9.
Kadotani H, Faraco J, Mignot E. Genetic studies in the sleep disorder narcolepsy. Genome Res.
1998;8:427–34.
Kaitin KI, Kilduff TS, Dement WC. Evidence for excessive sleepiness in canine narcoleptics.
Electroencephalogr Clin Neurophysiol. 1986a;64:447–54.
Kaitin KI, Kilduff TS, Dement WC. Sleep fragmentation in canine narcolepsy. Sleep. 1986b;9:116–
9.
12 Narcolepsy and Orexin/Hypocretin 257
Kales A, Soldatos CR, Bixler EO, Caldwell A, Cadieux RJ, Verrechio JM, Kales JD. Narcolepsy-
cataplexy. II. Psychosocial consequences and associated psychopathology. Arch Neurol.
1982;39:169–71.
Kalogiannis M, Hsu E, Willie JT, Chemelli RM, Kisanuki YY, Yanagisawa M, Leonard
CS. Cholinergic modulation of narcoleptic attacks in double orexin receptor knockout mice.
PLoS One. 2011;6:e18697.
Kantor S, Mochizuki T, Lops SN, Ko B, Clain E, Clark E, Yamamoto M, Scammell TE. Orexin
gene therapy restores the timing and maintenance of wakefulness in narcoleptic mice. Sleep.
2013;36:1129–38.
Kilduff TS, Peyron C. The hypocretin/orexin ligand-receptor system: implications for sleep and
sleep disorders. Trends Neurosci. 2000;23:359–65.
Kirchgessner AL, Liu M. Orexin synthesis and response in the gut. Neuron. 1999;24:941–51.
Kiyashchenko LI, Mileykovskiy BY, Lai YY, Siegel JM. Increased and decreased muscle tone with
orexin (hypocretin) microinjections in the locus coeruleus and pontine inhibitory area. J
Neurophysiol. 2001;85:2008–16.
Kiyashchenko LI, Mileykovskiy BY, Maidment N, Lam HA, Wu MF, John J, Peever J, Siegel
JM. Release of hypocretin (orexin) during waking and sleep states. J Neurosci. 2002;22:5282–6.
Kornum BR, Faraco J, Mignot E. Narcolepsy with hypocretin/orexin deficiency, infections and
autoimmunity of the brain. Curr Opin Neurobiol. 2011a;21:897–903.
Kornum BR, Kawashima M, Faraco J, Lin L, Rico TJ, Hesselson S, Axtell RC, Kuipers H,
Weiner K, Hamacher A, Kassack MU, Han F, Knudsen S, Li J, Dong X, Winkelmann J,
Plazzi G, Nevsimalova S, Hong SC, Honda Y, Honda M, Hogl B, Ton TG, Montplaisir J,
Bourgin P, Kemlink D, Huang YS, Warby S, Einen M, Eshragh JL, Miyagawa T, Desautels A,
Ruppert E, Hesla PE, Poli F, Pizza F, Frauscher B, Jeong JH, Lee SP, Strohl KP, Longstreth WT
Jr, Kvale M, Dobrovolna M, Ohayon MM, Nepom GT, Wichmann HE, Rouleau GA, Gieger C,
Levinson DF, Gejman PV, Meitinger T, Peppard P, Young T, Jennum P, Steinman L,
Tokunaga K, Kwok PY, Risch N, Hallmayer J, Mignot E. Common variants in P2RY11 are
associated with narcolepsy. Nat Genet. 2011b;43:66–71.
Korotkova TM, Eriksson KS, Haas HL, Brown RE. Selective excitation of GABAergic neurons in
the substantia nigra of the rat by orexin/hypocretin in vitro. Regul Pept. 2002;104:83–9.
Korotkova TM, Sergeeva OA, Eriksson KS, Haas HL, Brown RE. Excitation of ventral tegmental
area dopaminergic and nondopaminergic neurons by orexins/hypocretins. J Neurosci. 2003;23:
7–11.
Krahn LE, Pankratz VS, Oliver L, Boeve BF, Silber MH. Hypocretin (orexin) levels in cerebro-
spinal fluid of patients with narcolepsy: relationship to cataplexy and HLA DQB1*0602 status.
Sleep. 2002;25:733–6.
Kukkonen JP, Holmqvist T, Ammoun S, Akerman KE. Functions of the orexinergic/
hypocretinergic system. Am J Physiol Cell Physiol. 2002;283:C1567–91.
Kuru M, Ueta Y, Serino R, Nakazato M, Yamamoto Y, Shibuya I, Yamashita H. Centrally
administered orexin/hypocretin activates HPA axis in rats. Neuroreport. 2000;11:1977–80.
Kuwaki T. Orexinergic modulation of breathing across vigilance states. Respir Physiol Neurobiol.
2008;164:204–12.
Kuwaki T. Hypothalamic modulation of breathing. Adv Exp Med Biol. 2010;669:243–7.
Kuwaki T, Li A, Nattie E. State-dependent central chemoreception: a role of orexin. Respir Physiol
Neurobiol. 2010;173:223–9.
Lai YY, Siegel JM. Medullary regions mediating atonia. J Neurosci. 1988;8:4790–6.
Lammers GJ, Pijl H, Iestra J, Langius JA, Buunk G, Meinders AE. Spontaneous food choice in
narcolepsy. Sleep. 1996;19:75–6.
Latorre D, Kallweit U, Armentani E, Foglierini M, Mele F, Cassotta A, Jovic S, Jarrossay D,
Mathis J, Zellini F, Becher B, Lanzavecchia A, Khatami R, Manconi M, Tafti M, Bassetti CL,
Sallusto F. T cells in patients with narcolepsy target self-antigens of hypocretin neurons. Nature.
2018;562:63–8.
258 F. L. Xiao et al.
Lecendreux M, Maret S, Bassetti C, Mouren MC, Tafti M. Clinical efficacy of high-dose intrave-
nous immunoglobulins near the onset of narcolepsy in a 10-year-old boy. J Sleep Res. 2003;12:
347–8.
Li Y, Gao XB, Sakurai T, van den Pol AN. Hypocretin/Orexin excites hypocretin neurons via a
local glutamate neuron-A potential mechanism for orchestrating the hypothalamic arousal
system. Neuron. 2002;36:1169–81.
Li J, Hu Z, de Lecea L. The hypocretins/orexins: integrators of multiple physiological functions. Br
J Pharmacol. 2014;171:332–50.
Liblau RS, Vassalli A, Seifinejad A, Tafti M. Hypocretin (orexin) biology and the pathophysiology
of narcolepsy with cataplexy. Lancet Neurol. 2015;14:318–28.
Lin L, Faraco J, Li R, Kadotani H, Rogers W, Lin X, Qiu X, de Jong PJ, Nishino S, Mignot E. The
sleep disorder canine narcolepsy is caused by a mutation in the hypocretin (orexin) receptor
2 gene. Cell. 1999;98:365–76.
Liu ZW, Gao XB. Adenosine inhibits activity of hypocretin/orexin neurons by the A1 receptor in
the lateral hypothalamus: a possible sleep-promoting effect. J Neurophysiol. 2007;97:837–48.
Liu M, Thankachan S, Kaur S, Begum S, Blanco-Centurion C, Sakurai T, Yanagisawa M, Neve R,
Shiromani PJ. Orexin (hypocretin) gene transfer diminishes narcoleptic sleep behavior in mice.
Eur J Neurosci. 2008;28:1382–93.
Longstreth WT Jr, Koepsell TD, Ton TG, Hendrickson AF, van Belle G. The epidemiology of
narcolepsy. Sleep. 2007;30:13–26.
Lu XY, Bagnol D, Burke S, Akil H, Watson SJ. Differential distribution and regulation of OX1 and
OX2 orexin/hypocretin receptor messenger RNA in the brain upon fasting. Horm Behav.
2000;37:335–44.
Lund PE, Shariatmadari R, Uustare A, Detheux M, Parmentier M, Kukkonen JP, Akerman KE. The
orexin OX1 receptor activates a novel Ca2+ influx pathway necessary for coupling to phospho-
lipase C. J Biol Chem. 2000;275:30806–12.
Luo G, Ambati A, Lin L, Bonvalet M, Partinen M, Ji X, Maecker HT, Mignot EJ. Autoimmunity to
hypocretin and molecular mimicry to flu in type 1 narcolepsy. Proc Natl Acad Sci U S
A. 2018;115:E12323–32.
Lutter M, Krishnan V, Russo SJ, Jung S, McClung CA, Nestler EJ. Orexin signaling mediates the
antidepressant-like effect of calorie restriction. J Neurosci. 2008;28:3071–5.
Matsuki K, Honda Y, Juji T. Diagnostic criteria for narcolepsy and HLA-DR2 frequencies. Tissue
Antigens. 1987;30:155–60.
Matsuki K, Grumet FC, Lin X, Gelb M, Guilleminault C, Dement WC, Mignot E. DQ (rather than
DR) gene marks susceptibility to narcolepsy. Lancet. 1992;339:1052.
Mayer G, Hellmann F, Leonhard E, Meier-Ewert K. Circadian temperature and activity rhythms in
unmedicated narcoleptic patients. Pharmacol Biochem Behav. 1997;58:395–402.
McDevitt HO, Tyan ML. Genetic control of the antibody response in inbred mice. Transfer of
response by spleen cells and linkage to the major histocompatibility (H-2) locus. J Exp Med.
1968;128:1–11.
Mignot E. Genetic and familial aspects of narcolepsy. Neurology. 1998;50:S16–22.
Mignot E. Perspectives in narcolepsy and hypocretin (orexin) research. Sleep Med. 2000;1:87–90.
Mignot EJ. History of narcolepsy at Stanford University. Immunol Res. 2014;58:315–39.
Mignot E, Lin X, Hesla PE, Dement WC, Guilleminault C, Grumet FC. A novel HLA DR17, DQ1
(DQA1-0102/DQB1-0602 positive) haplotype predisposing to narcolepsy in Caucasians. Sleep.
1993;16:764–5.
Mignot E, Lin X, Arrigoni J, Macaubas C, Olive F, Hallmayer J, Underhill P, Guilleminault C,
Dement WC, Grumet FC. DQB1*0602 and DQA1*0102 (DQ1) are better markers than DR2 for
narcolepsy in Caucasian and black Americans. Sleep. 1994;17:S60–7.
Mignot E, Kimura A, Lattermann A, Lin X, Yasunaga S, Mueller-Eckhardt G, Rattazzi C, Lin L,
Guilleminault C, Grumet FC, Mayer G, Dement WC, Underhill P. Extensive HLA class II
studies in 58 non-DRB1*15 (DR2) narcoleptic patients with cataplexy. Tissue Antigens.
1997;49:329–41.
12 Narcolepsy and Orexin/Hypocretin 259
Mignot E, Lin L, Rogers W, Honda Y, Qiu X, Lin X, Okun M, Hohjoh H, Miki T, Hsu S, Leffell M,
Grumet F, Fernandez-Vina M, Honda M, Risch N. Complex HLA-DR and -DQ interactions
confer risk of narcolepsy-cataplexy in three ethnic groups. Am J Hum Genet. 2001;68:686–99.
Mignot E, Lammers GJ, Ripley B, Okun M, Nevsimalova S, Overeem S, Vankova J, Black J,
Harsh J, Bassetti C, Schrader H, Nishino S. The role of cerebrospinal fluid hypocretin measure-
ment in the diagnosis of narcolepsy and other hypersomnias. Arch Neurol. 2002;59:1553–62.
Mileykovskiy BY, Kiyashchenko LI, Siegel JM. Behavioral correlates of activity in identified
hypocretin/orexin neurons. Neuron. 2005;46:787–98.
Mitler MM, Walsleben J, Sangal RB, Hirshkowitz M. Sleep latency on the maintenance of
wakefulness test (MWT) for 530 patients with narcolepsy while free of psychoactive drugs.
Electroencephalogr Clin Neurophysiol. 1998;107:33–8.
Mochizuki T, Crocker A, McCormack S, Yanagisawa M, Sakurai T, Scammell TE. Behavioral state
instability in orexin knock-out mice. J Neurosci. 2004;24:6291–300.
Montplaisir J, Billiard M, Takahashi S, Bell IR, Guilleminault C, Dement WC. Twenty-four-hour
recording in REM-narcoleptics with special reference to nocturnal sleep disruption. Biol
Psychiatry. 1978;13:73–89.
Moriguchi T, Sakurai T, Nambu T, Yanagisawa M, Goto K. Neurons containing orexin in the lateral
hypothalamic area of the adult rat brain are activated by insulin-induced acute hypoglycemia.
Neurosci Lett. 1999;264:101–4.
Mosko SS, Shampain DS, Sassin JF. Nocturnal REM latency and sleep disturbance in narcolepsy.
Sleep. 1984;7:115–25.
Mueller-Eckhardt G, Meier-Ewert K, Schendel DJ, Reinecker FB, Multhoff G, Mueller-Eckhardt
C. HLA and narcolepsy in a German population. Tissue Antigens. 1986;28:163–9.
Nakamura A, Zhang W, Yanagisawa M, Fukuda Y, Kuwaki T. Vigilance state-dependent attenu-
ation of hypercapnic chemoreflex and exaggerated sleep apnea in orexin knockout mice. J Appl
Physiol (1985). 2007;102:241–8.
Nattie E, Li A. Central chemoreception in wakefulness and sleep: evidence for a distributed network
and a role for orexin. J Appl Physiol (1985). 2010;108:1417–24.
Neely SE, Rosenberg RS, Spire JP, Antel J, Arnason BGW. Familial Narcolepsy and Hla Antigens.
Ann Neurol. 1986;20:168.
Nishino S. Clinical and neurobiological aspects of narcolepsy. Sleep Med. 2007a;8:373–99.
Nishino S. Narcolepsy: pathophysiology and pharmacology. J Clin Psychiatry. 2007b;68(Suppl
13):9–15.
Nishino S, Kanbayashi T. Symptomatic narcolepsy, cataplexy and hypersomnia, and their impli-
cations in the hypothalamic hypocretin/orexin system. Sleep Med Rev. 2005;9:269–310.
Nishino S, Mignot E. Pharmacological aspects of human and canine narcolepsy. Prog Neurobiol.
1997;52:27–78.
Nishino S, Mignot E, Fruhstorfer B, Dement WC, Hayaishi O. Prostaglandin E2 and its methyl ester
reduce cataplexy in canine narcolepsy. Proc Natl Acad Sci U S A. 1989;86:2483–7.
Nishino S, Tafti M, Reid MS, Shelton J, Siegel JM, Dement WC, Mignot E. Muscle atonia is
triggered by cholinergic stimulation of the basal forebrain: implication for the pathophysiology
of canine narcolepsy. J Neurosci. 1995;15:4806–14.
Nishino S, Riehl J, Hong J, Kwan M, Reid M, Mignot E. Is narcolepsy a REM sleep disorder?
Analysis of sleep abnormalities in narcoleptic Dobermans. Neurosci Res. 2000a;38:437–46.
Nishino S, Ripley B, Overeem S, Lammers GJ, Mignot E. Hypocretin (orexin) deficiency in human
narcolepsy. Lancet. 2000b;355:39–40.
Nishino S, Ripley B, Overeem S, Nevsimalova S, Lammers GJ, Vankova J, Okun M, Rogers W,
Brooks S, Mignot E. Low cerebrospinal fluid hypocretin (Orexin) and altered energy homeo-
stasis in human narcolepsy. Ann Neurol. 2001;50:381–8.
Nishino S, Kanbayashi T, Fujiki N, Uchino M, Ripley B, Watanabe M, Lammers GJ, Ishiguro H,
Shoji S, Nishida Y, Overeem S, Toyoshima I, Yoshida Y, Shimizu T, Taheri S, Mignot E. CSF
hypocretin levels in Guillain-Barre syndrome and other inflammatory neuropathies. Neurology.
2003;61:823–5.
260 F. L. Xiao et al.
Ohayon MM, Priest RG, Zulley J, Smirne S, Paiva T. Prevalence of narcolepsy symptomatology
and diagnosis in the European general population. Neurology. 2002;58:1826–33.
Overeem S, Dalmau J, Bataller L, Nishino S, Mignot E, Verschuuren J, Lammers GJ. Hypocretin-1
CSF levels in anti-Ma2 associated encephalitis. Neurology. 2004;62:138–40.
Pataka AD, Frangulyan RR, Mackay TW, Douglas NJ, Riha RL. Narcolepsy and sleep-disordered
breathing. Eur J Neurol. 2012;19:696–702.
Peyron C, Tighe DK, van den Pol AN, de Lecea L, Heller HC, Sutcliffe JG, Kilduff TS. Neurons
containing hypocretin (orexin) project to multiple neuronal systems. J Neurosci. 1998;18:9996–
10015.
Peyron C, Faraco J, Rogers W, Ripley B, Overeem S, Charnay Y, Nevsimalova S, Aldrich M,
Reynolds D, Albin R, Li R, Hungs M, Pedrazzoli M, Padigaru M, Kucherlapati M, Fan J,
Maki R, Lammers GJ, Bouras C, Kucherlapati R, Nishino S, Mignot E. A mutation in a case of
early onset narcolepsy and a generalized absence of hypocretin peptides in human narcoleptic
brains. Nat Med. 2000;6:991–7.
Poryazova R, Siccoli M, Werth E, Bassetti CL. Unusually prolonged rebound cataplexy after
withdrawal of fluoxetine. Neurology. 2005;65:967–8.
Richardson GS, Carskadon MA, Flagg W, Van den Hoed J, Dement WC, Mitler MM. Excessive
daytime sleepiness in man: multiple sleep latency measurement in narcoleptic and control
subjects. Electroencephalogr Clin Neurophysiol. 1978;45:621–7.
Ripley B, Overeem S, Fujiki N, Nevsimalova S, Uchino M, Yesavage J, Di Monte D, Dohi K,
Melberg A, Lammers GJ, Nishida Y, Roelandse FW, Hungs M, Mignot E, Nishino S. CSF
hypocretin/orexin levels in narcolepsy and other neurological conditions. Neurology. 2001;57:
2253–8.
Rogers AE, Rosenberg RS. Tests of memory in narcoleptics. Sleep. 1990;13:42–52.
Rogers AA, Aiani LM, Blanpain LT, Yuxian S, Moore R, Willie JT. Deep brain stimulation of
hypothalamus for narcolepsy-cataplexy in mice. Brain Stimul. 2020;13:1305–16.
Sakurai T. Orexin deficiency and narcolepsy. Curr Opin Neurobiol. 2013;23:760–6.
Sakurai T, Amemiya A, Ishii M, Matsuzaki I, Chemelli RM, Tanaka H, Williams SC, Richarson JA,
Kozlowski GP, Wilson S, Arch JR, Buckingham RE, Haynes AC, Carr SA, Annan RS,
McNulty DE, Liu WS, Terrett JA, Elshourbagy NA, Bergsma DJ, Yanagisawa M. Orexins
and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors
that regulate feeding behavior. Cell. 1998;92:1. page following 696
Samson WK, Resch ZT. The hypocretin/orexin story. Trends Endocrinol Metab. 2000;11:257–62.
Scammell TE, Willie JT, Guilleminault C, Siegel JM, International Working Group on Rodent
Models of Narcolepsy. A consensus definition of cataplexy in mouse models of narcolepsy.
Sleep. 2009;32:111–6.
Schenck CH, Mahowald MW. Motor dyscontrol in narcolepsy: rapid-eye-movement (REM) sleep
without atonia and REM sleep behavior disorder. Ann Neurol. 1992;32:3–10.
Schuld A, Hebebrand J, Geller F, Pollmacher T. Increased body-mass index in patients with
narcolepsy. Lancet. 2000;355:1274–5.
Scott MM, Marcus JN, Pettersen A, Birnbaum SG, Mochizuki T, Scammell TE, Nestler EJ,
Elmquist JK, Lutter M. Hcrtr1 and 2 signaling differentially regulates depression-like behaviors.
Behav Brain Res. 2011;222:289–94.
Sergeeva OA, Korotkova TM, Scherer A, Brown RE, Haas HL. Co-expression of non-selective
cation channels of the transient receptor potential canonical family in central aminergic
neurones. J Neurochem. 2003;85:1547–52.
Siegel JM, Nienhuis R, Fahringer HM, Paul R, Shiromani P, Dement WC, Mignot E, Chiu
C. Neuronal activity in narcolepsy: identification of cataplexy-related cells in the medial
medulla. Science. 1991;252:1315–8.
Silber MH, Krahn LE, Olson EJ, Pankratz VS. The epidemiology of narcolepsy in Olmsted County,
Minnesota: a population-based study. Sleep. 2002;25:197–202.
Strohl KP, Cherniack NS, Gothe B. Physiologic basis of therapy for sleep apnea. Am Rev Respir
Dis. 1986;134:791–802.
12 Narcolepsy and Orexin/Hypocretin 261
Sunanaga J, Deng BS, Zhang W, Kanmura Y, Kuwaki T. CO2 activates orexin-containing neurons
in mice. Respir Physiol Neurobiol. 2009;166:184–6.
Sutcliffe JG, de Lecea L. The hypocretins: excitatory neuromodulatory peptides for multiple
homeostatic systems, including sleep and feeding. J Neurosci Res. 2000;62:161–8.
Tabuchi S, Tsunematsu T, Black SW, Tominaga M, Maruyama M, Takagi K, Minokoshi Y,
Sakurai T, Kilduff TS, Yamanaka A. Conditional ablation of orexin/hypocretin neurons: a
new mouse model for the study of narcolepsy and orexin system function. J Neurosci.
2014;34:6495–509.
Tafti M, Lammers GJ, Dauvilliers Y, Overeem S, Mayer G, Nowak J, Pfister C, Dubois V, Eliaou
JF, Eberhard HP, Liblau R, Wierzbicka A, Geisler P, Bassetti CL, Mathis J, Lecendreux M,
Khatami R, Heinzer R, Haba-Rubio J, Feketeova E, Baumann CR, Kutalik Z, Tiercy
JM. Narcolepsy-associated HLA class I alleles implicate cell-mediated cytotoxicity. Sleep.
2016;39:581–7.
Thannickal TC, Moore RY, Nienhuis R, Ramanathan L, Gulyani S, Aldrich M, Cornford M, Siegel
JM. Reduced number of hypocretin neurons in human narcolepsy. Neuron. 2000;27:469–74.
Thorpy MJ. Recently approved and upcoming treatments for narcolepsy. CNS Drugs. 2020;34:9–
27.
Trivedi P, Yu H, MacNeil DJ, Van der Ploeg LH, Guan XM. Distribution of orexin receptor mRNA
in the rat brain. FEBS Lett. 1998;438:71–5.
Tsujino N, Sakurai T. Orexin/hypocretin: a neuropeptide at the interface of sleep, energy homeo-
stasis, and reward system. Pharmacol Rev. 2009;61:162–76.
Tsujino N, Yamanaka A, Ichiki K, Muraki Y, Kilduff TS, Yagami K, Takahashi S, Goto K, Sakurai
T. Cholecystokinin activates orexin/hypocretin neurons through the cholecystokinin A receptor.
J Neurosci. 2005;25:7459–69.
Valko PO, Gavrilov YV, Yamamoto M, Reddy H, Haybaeck J, Mignot E, Baumann CR, Scammell
TE. Increase of histaminergic tuberomammillary neurons in narcolepsy. Ann Neurol. 2013;74:
794–804.
van den Pol AN. Hypothalamic hypocretin (orexin): robust innervation of the spinal cord. J
Neurosci. 1999;19:3171–82.
van den Pol AN, Gao XB, Obrietan K, Kilduff TS, Belousov AB. Presynaptic and postsynaptic
actions and modulation of neuroendocrine neurons by a new hypothalamic peptide, hypocretin/
orexin. J Neurosci. 1998;18:7962–71.
White DP, Douglas NJ, Pickett CK, Zwillich CW, Weil JV. Sleep deprivation and the control of
ventilation. Am Rev Respir Dis. 1983;128:984–6.
Williams RH, Burdakov D. Hypothalamic orexins/hypocretins as regulators of breathing. Expert
Rev Mol Med. 2008;10:e28.
Williams RH, Jensen LT, Verkhratsky A, Fugger L, Burdakov D. Control of hypothalamic orexin
neurons by acid and CO2. Proc Natl Acad Sci U S A. 2007;104:10685–90.
Willie JT, Chemelli RM, Sinton CM, Tokita S, Williams SC, Kisanuki YY, Marcus JN, Lee C,
Elmquist JK, Kohlmeier KA, Leonard CS, Richardson JA, Hammer RE, Yanagisawa
M. Distinct narcolepsy syndromes in Orexin receptor-2 and Orexin null mice: molecular genetic
dissection of Non-REM and REM sleep regulatory processes. Neuron. 2003;38:715–30.
Wing YK, Li RH, Lam CW, Ho CK, Fong SY, Leung T. The prevalence of narcolepsy among
Chinese in Hong Kong. Ann Neurol. 2002;51:578–84.
Wu MF, Gulyani SA, Yau E, Mignot E, Phan B, Siegel JM. Locus coeruleus neurons: cessation of
activity during cataplexy. Neuroscience. 1999;91:1389–99.
Wu M, Zhang Z, Leranth C, Xu C, van den Pol AN, Alreja M. Hypocretin increases impulse flow in
the septohippocampal GABAergic pathway: implications for arousal via a mechanism of
hippocampal disinhibition. J Neurosci. 2002;22:7754–65.
Xie X, Crowder TL, Yamanaka A, Morairty SR, Lewinter RD, Sakurai T, Kilduff TS. GABA
(B) receptor-mediated modulation of hypocretin/orexin neurones in mouse hypothalamus. J
Physiol. 2006;574:399–414.
262 F. L. Xiao et al.
Yamanaka A, Tsujino N, Funahashi H, Honda K, Guan JL, Wang QP, Tominaga M, Goto K,
Shioda S, Sakurai T. Orexins activate histaminergic neurons via the orexin 2 receptor. Biochem
Biophys Res Commun. 2002;290:1237–45.
Yamanaka A, Beuckmann CT, Willie JT, Hara J, Tsujino N, Mieda M, Tominaga M, Yagami K,
Sugiyama F, Goto K, Yanagisawa M, Sakurai T. Hypothalamic orexin neurons regulate arousal
according to energy balance in mice. Neuron. 2003a;38:701–13.
Yamanaka A, Muraki Y, Tsujino N, Goto K, Sakurai T. Regulation of orexin neurons by the
monoaminergic and cholinergic systems. Biochem Biophys Res Commun. 2003b;303:120–9.
Yamanaka A, Muraki Y, Ichiki K, Tsujino N, Kilduff TS, Goto K, Sakurai T. Orexin neurons are
directly and indirectly regulated by catecholamines in a complex manner. J Neurophysiol.
2006;96:284–98.
Yang B, Ferguson AV. Orexin-A depolarizes nucleus tractus solitarius neurons through effects on
nonselective cationic and K+ conductances. J Neurophysiol. 2003;89:2167–75.
Yukitake H, Fujimoto T, Ishikawa T, Suzuki A, Shimizu Y, Rikimaru K, Ito M, Suzuki M, Kimura
H. TAK-925, an orexin 2 receptor-selective agonist, shows robust wake-promoting effects in
mice. Pharmacol Biochem Behav. 2019;187:172794.
Zeitzer JM, Nishino S, Mignot E. The neurobiology of hypocretins (orexins), narcolepsy and related
therapeutic interventions. Trends Pharmacol Sci. 2006;27:368–74.
Zhu Y, Miwa Y, Yamanaka A, Yada T, Shibahara M, Abe Y, Sakurai T, Goto K. Orexin receptor
type-1 couples exclusively to pertussis toxin-insensitive G-proteins, while orexin receptor type-
2 couples to both pertussis toxin-sensitive and -insensitive G-proteins. J Pharmacol Sci.
2003;92:259–66.
Part V
Circadian Rhythm Disorders
Chapter 13
Circadian Rhythm Sleep–Wake Disorders:
Mechanisms and Treatment
Abstract Nearly all biological processes exhibit circadian rhythms that are gener-
ated by circadian clocks in central and peripheral tissues. In mammals a central
circadian clock, the suprachiasmatic nucleus helps to align behaviors and physio-
logical processes, including the sleep–wake cycle with the 24-h environment.
Disruption of the proper alignment of circadian clocks with the required sleep–
wake time leads to development of circadian rhythm sleep–wake disorders. These
disorders can develop as a result of pathology at the level of the internal clock,
disruption of the ability to receive or process environmental synchronizing signals,
or changes to the external environmental time. Treatment of circadian rhythm sleep–
wake disorders depends on behavioral adjustments, often in conjunction with spe-
cific timing of light and/or melatonin. This chapter will highlight the six primary
circadian rhythm sleep–wake disorders, focusing on what is known about their
underlying pathogenic mechanisms and the currently recommended treatment
strategies.
13.1 Introduction
Circadian rhythms are the near 24-oscillations observed in nearly all physiological
processes and behaviors. They serve to align, or entrain, these behaviors with the
external environment. While the most apparent of these rhythms is the daily pattern
of the sleep–wake cycle, these rhythms can also be observed in everything from
feeding behaviors to daily patterns of body temperature and hormone release. In
mammals, these rhythms are regulated by the suprachiasmatic nucleus (SCN), a set
of paired nuclei, located in the hypothalamus, directly above the optic chiasm
(Stephan and Zucker 1972). The SCN sends direct projections to the paraventricular
© Springer Nature B.V. and Shanghai Jiao Tong University Press 2022 265
A. I. Pack, Q. Y. Li (eds.), Sleep and its Disorders, Translational Medicine Research,
https://doi.org/10.1007/978-94-024-2168-2_13
266 S. M. Abbott and P. C. Zee
Light Sleep-wake
CLOCK
PER PER
Activity BMAL1
CRY P CRY P Thermoregulation
PER
CRY
Pineal
Fig. 13.1 Illustration of the circadian system. Cells within the suprachiasmatic nucleus (SCN)
contain a set of core clock genes. The transcription–translation feedback loop completed by these
genes maintains the 24-h clock. Peripheral inputs to the SCN can reset or adjust circadian timing,
while the SCN in turn provides outputs that regulate the timing of many of these behaviors
Symptoms:
Difficulty Falling Asleep at Night
and/or
Daytime Sleepiness
Evaluation:
Clinical History
Actigraphy and/or Sleep Logs
Circadian phase markers (optional)
Fig. 13.2 Algorithm for the evaluation of a patient with a circadian rhythm sleep–wake disorder.
Treatment strategies are based on targeting the primary underlying problem, either using phase
shifting strategies to adjust misalignment, or using strong time cues to improve amplitude
photoreceptors also appear to play a role in irradiance detection (Gooley et al. 2001;
van Diepen et al. 2013). Light in the evening, prior to the core body temperature
nadir causes delays in the circadian rhythm, while light in the morning, after the core
body temperature nadir causes circadian advances (St Hilaire et al. 2012).
Melatonin is one of the other key resetting signals for the mammalian circadian
clock. Melatonin is secreted by the pineal gland, and levels normally increase shortly
before sleep onset, peaking in the middle of the night, and dropping off the following
morning (Lewy and Sack 1989). Opposite to the effects seen with light, melatonin in
the evening can cause circadian advances, while melatonin in the morning causes
circadian delays (Burgess et al. 2010). The circadian resetting properties of light and
melatonin play a key role in the treatment of circadian rhythms sleep–wake
disorders.
The circadian rhythm sleep–wake disorders result when there is a mismatch
between the internal circadian time and the external environment. Symptoms should
be present for at least 3 months. Figure 13.2 outlines a schematic for evaluating a
patient suspected to have a circadian rhythm sleep–wake disorder. Delayed sleep–
wake phase disorder, advanced sleep–wake phase disorder irregular sleep–wake
rhythm disorder and non-24-h sleep–wake rhythm disorder are thought to be pri-
marily due to abnormalities at the level of biological clock, though behavioral factors
can often contribute to the development of these disorders. Conversely, shift work
disorder and jet lag disorder develop when an individual is behaviorally active
during the time period they would normally be sleeping, either because of work
requirements or as a result of crossing multiple time zones. However, there are likely
biological factors contributing to these disorders as well, as some individuals
exposed to these environmental conditions appear to be able to adapt to these
symptoms, and do not develop a chronic circadian disorder (ICSD-3 2014). In the
268 S. M. Abbott and P. C. Zee
Table 13.1 Summary of the strength of evidence for current treatment strategies for circadian
rhythm sleep–wake disorders (Auger et al. 2015)
Diagnosis Treatment Evidence
DSWPD Timed evening melatonin Weak for
Post-awakening light Weak for
ASWPD Evening light Weak for
ISWRD Light therapy Weak for
Melatonin in elderly with dementia Weak against
Hypnotics for elderly patients Strong against
N24SWD Timed melatonin (in blind adults) Weak for
following sections, we will detail what is currently known about the underlying
pathophysiology and treatment options for each of these disorders (Table 13.1).
Patients with delayed sleep–wake phase disorder (DSWPD) have chronic or recur-
rent habitual sleep times that are significantly later than average, often not going to
bed until at least 2 am, or sometimes much later, and are unable to wake until late
morning/early afternoon. These individuals are often diagnosed with insomnia due
to their difficulty falling asleep, however, if allowed to sleep during their preferred
times, sleep duration and quality are normal for their age (ICSD-3 2014). The
prevalence of DSWPD varies depending on the population being studied, but can
be as high as 7–16% in adolescents and decreases with age (ICSD-3 2014; Paine
et al. 2014). In addition, it has been estimated that ~10% of patients presenting to a
sleep clinic with a chief complaint of insomnia actually have DSWPD (Flynn-Evans
et al. 2017).
The diagnosis of DSWPD is confirmed through the use of sleep logs, preferably
with the addition of actigraphy, collected for at least 7 days, but ideally 14 days to
confirm a delayed sleep–wake pattern (Fig. 13.3). Standard chronotype question-
naires such as the Munich which measures the actual timing of daily sleep–wake
patterns, or the Horne-Ostberg which measures sleep–wake preferences (Zavada
et al. 2005) can be used to confirm a later sleep midpoint and evening chronotype,
though these are not required for diagnosis. In addition, the pattern of secretion of
melatonin can also be measured in the saliva to confirm a delay in the daily onset of
melatonin secretion (ICSD-3 2014). Overnight sleep studies are generally not
indicated unless there is a concern for another underlying sleep disorder such as
obstructive sleep apnea.
The underlying pathophysiology of DSWPD is still not fully understood; how-
ever, there are several current theories regarding the underlying mechanism, with
varying degrees of supportive evidence: (1) Individuals with DSWPD are either
more sensitive to the delaying effects of evening light or less sensitive to the
13 Circadian Rhythm Sleep–Wake Disorders: Mechanisms and Treatment 269
Fig. 13.3 Example actigraphy from a subject with delayed sleep–wake phase disorder. Black bars
indicate activity, while the yellow line represents light levels. Each line is 24 h. Sleep onset is
typically around 2–3 am, while sleep offset is generally around 10 am
advancing effects of morning light; (2) Due to behavioral changes, individuals with
DSWPD are exposed to more light in the evening during times of maximal phase
delay, and are exposed to less light in the morning during times of maximal phase
advance; or (3) Individuals with DSWPD have a longer intrinsic period, making it
more difficult to fully entrain to a 24-h schedule. A very small study of individuals
with DSWPD did demonstrate that these individuals exhibit a greater degree of
melatonin suppression in response to light, suggesting greater sensitivity to evening
light (Aoki et al. 2001). However, similar studies have not been performed evaluat-
ing the sensitivity of these individuals to light during the phase advance time. The
support for behavioral factors contributing to DSWPD is mixed. Studies in adoles-
cents have demonstrated a greater exposure to evening light, and decreased exposure
to morning light in evening type individuals with respect to clock time. However,
when corrected for circadian time, there were no significant differences observed
suggesting that they are not receiving excess light at circadian times that would
promote a delay (Auger et al. 2011). Finally, to evaluate the potential for increased
period length as a contributing factor, polymorphisms of core clock genes have been
analyzed in individuals with DSWPD. It has been demonstrated that polymorphisms
in the gene hPer3 near the phosphorylation site for CK1ε are associated with
DSWPD (Ebisawa et al. 2001). Alterations in phosphorylation presumably affect
the degradation rate of hPER3, in turn impacting the overall period.
Treatment of DSWPD focuses on very specific timing of both light exposure and
avoidance, along with timed melatonin. In addition, close attention to general
270 S. M. Abbott and P. C. Zee
In advanced sleep–wake phase disorder (ASWPD) patients fall asleep and wake up
much earlier than desired, with typical bedtimes around 6–9 pm and wake times
around 2–5 am. Sleep complaints include difficulty staying awake for evening
activities, along with early morning awakenings. If they are able to make themselves
stay up later, they develop daytime sleepiness because they are still unable to sleep
later. Similar to DSWPD, patients with ASWPD have a normal duration and quality
of sleep when allowed to sleep during their preferred times (ICSD-3 2014). The
13 Circadian Rhythm Sleep–Wake Disorders: Mechanisms and Treatment 271
Fig. 13.4 Example actigraphy from a subject with non-24-h sleep–wake rhythm disorder. Black
bars indicate activity, while the yellow line represents light levels. Each line is 24 h. The duration of
the sleep window remains relatively similar from day to day, however, the onset shifts later by 1–2 h
each day. Note the overall amplitude of activity and light
13 Circadian Rhythm Sleep–Wake Disorders: Mechanisms and Treatment 273
light perception who are unaffected have an internal clock that is closer to 24 h, so it
is easier for them to maintain entrainment through non-photic cues, such as social
interactions and activity. Sighted individuals with N24SWD are thought to result
from a combination of factors. These individuals often initially present as cases of
severe DSWPD, and they eventually are unable to maintain entrainment (Hayakawa
et al. 2005). There is also evidence that sighted individuals with N24SWD have an
internal period that is significantly longer than 24 h, making it more difficult to
maintain entrainment through traditional signals (Kitamura et al. 2013). In addition,
other potential possibilities include limited exposure to strong entraining signals, or
an inability to respond normally to the entraining signals of light.
The treatment of N24SWD is dependent on the underlying etiology. In blind
individuals treatment with daily melatonin at a fixed time has been effective.
Previous studies have evaluated doses ranging from 10 mg (Sack et al. 2000) to
0.5 mg (Lewy et al. 2001) given 1 h before the desired bedtime. Currently lower
doses are preferred. More recently tasimelteon, a melatonin agonist was developed
as the first FDA approved medication for the treatment of N24SWD (The Medical
Letter 2014). In sighted individuals treatment is more complicated, and there is less
data available on effective treatment regimens. Currently, suggestions include a
combination of enforcing available entraining signals, including regular timed
light exposure, strong social cues and regular sleep–wake cycles. Melatonin may
be effective; however, it is not as well studied in sighted N24SWD when compared
to blind individuals (Auger et al. 2015).
Shift work sleep disorder results when individuals develop sleep complaints as a
result of being required to follow a recurrent work schedule that occurs during the
time they would normally be sleeping. Symptoms can include both excessive
sleepiness while at work, and insomnia when allowed time to sleep. The diagnosis
is confirmed with sleep logs and/or actigraphy for at least 14 days, demonstrating a
disrupted sleep–wake pattern as a result of this work schedule (ICSD-3 2014).
Of note, not all individuals who are shift workers develop shift work disorder,
with some estimates suggesting that only 5–10% of shift workers have shift work
sleep disorder (Drake et al. 2004). Factors that may contribute to the development of
shift work disorder include age, gender, shift schedule, and circadian preference or
chronotype (Harma et al. 1994). Those who are more likely to develop shift work
disorder include those with a morning chronotype, older individuals, and individuals
with more daytime responsibilities, which may limit their ability to sleep during their
time off (Colligan and Rosa 1990). In addition, more recent studies have demon-
strated that polymorphisms in the hPer3 gene have been associated with a greater
tendency to develop symptoms of shift work disorder (Gumenyuk et al. 2015; Drake
et al. 2015).
276 S. M. Abbott and P. C. Zee
Jet lag disorder consists of sleep complaints that result from crossing at least two
time zones. In addition to complaints of insomnia and/or excessive sleepiness,
patients often also develop somatic symptoms such as gastrointestinal distress.
Diagnosis is made based on clinical history of experiencing the above symptoms
in temporal correlation with jet travel. The potential for and severity of symptoms
depends on both the direction of travel and the number of time zones crossed, with
eastward travel generally producing more symptoms (ICSD-3 2014).
Treatment of jet lag disorder depends on multiple factors, including the direction
of travel, number of time zones crossed, and duration of time spent at the destination.
In general, if trips are short (<48 h) it is often easier to try to maintain the home
schedule, rather than trying to adapt to the new schedule. For longer trips, targeted
use of light and melatonin to reset the clock can be beneficial. In all cases attention to
good sleep hygiene is important, making sure that the sleeping environment is cool,
dark, and quiet. In addition, adapting behaviors to the new environmental time as
soon as possible can be beneficial, including sleeping, and eating meals at the local
time. Avoiding excessive alcohol and caffeine can also help to limit the symptoms of
jet lag.
When traveling east the goal is to advance, or move the clock earlier. As the
human circadian clock naturally is slightly longer than 24 h, most individuals find
this transition to be much more challenging than the delays required for traveling
west. The required circadian phase shifts are generally accomplished through a
combination of timed melatonin, light exposure, and light avoidance. Depending
on the commitments prior to travel, attempts can be made to begin to adjust the clock
prior to leaving, or can start once arriving at the new destination. If trying to adjust
prior to departure, starting 3 days before leaving, patients should take melatonin
(1–3 mg) prior to their habitual sleep time, get at least an hour of bright light (5000
lux) in the morning, and move everything 1 h earlier each day (Burgess et al. 2003).
At the new time zone, to maximize the ability to re-entrain to the new time zone,
general recommendations are to avoid bright light in the morning and maximize
bright light exposure in the afternoon (Boulos et al. 1995). In addition, melatonin
(2–5 mg) can be taken before bed, both to help advance the clock, and as a mild
hypnotic (Suhner et al. 2001). Hypnotics such as zolpidem (10 mg) have also been
beneficial for improving sleep in the new location, though were associated with more
side effects when compared to melatonin (Suhner et al. 2001).
The goal when traveling west is to delay or move the clock later. As was
mentioned previously, it is generally easier for humans to delay than it is to advance.
Prior to traveling to the new destination, just one night of bright light exposure prior
to bedtime (2 h, ~4000 lux) can effectively delay the clock by ~1.5 h (Canton et al.
2009). After arriving at the new destination, patients should avoid bright light in the
morning, and seek bright light in the afternoon/evening to further delay the clock
(Boulos et al. 1995). Melatonin (5 mg) right before bedtime may provide additional
benefit (Petrie et al. 1993).
278 S. M. Abbott and P. C. Zee
13.8 Conclusions
References
Afonso P, Brissos S, Figueira ML, Paiva T. Schizophrenia patients with predominantly positive
symptoms have more disturbed sleep-wake cycles measured by actigraphy. Psychiatry Res.
2011;189(1):62–6.
Alessi CA, Martin JL, Webber AP, Cynthia Kim E, Harker JO, Josephson KR. Randomized,
controlled trial of a nonpharmacological intervention to improve abnormal sleep/wake patterns
in nursing home residents. J Am Geriatr Soc. 2005;53(5):803–10.
Ancoli-Israel S, Gehrman P, Martin JL, Shochat T, Marler M, Corey-Bloom J, et al. Increased light
exposure consolidates sleep and strengthens circadian rhythms in severe Alzheimer’s disease
patients. Behav Sleep Med. 2003;1(1):22–36.
Antunes LC, Levandovski R, Dantas G, Caumo W, Hidalgo MP. Obesity and shift work: chrono-
biological aspects. Nutr Res Rev. 2010;23(1):155–68.
Aoki H, Ozeki Y, Yamada N. Hypersensitivity of melatonin suppression in response to light in
patients with delayed sleep phase syndrome. Chronobiol Int. 2001;18(2):263–71.
Auger RR, Burgess HJ, Dierkhising RA, Sharma RG, Slocumb NL. Light exposure among
adolescents with delayed sleep phase disorder: a prospective cohort study. Chronobiol Int.
2011;28(10):911–20.
Auger RR, Burgess HJ, Emens JS, Deriy LV, Thomas SM, Sharkey KM. Clinical Practice
Guideline for the Treatment of Intrinsic Circadian Rhythm Sleep-Wake Disorders: Advanced
Sleep-Wake Phase Disorder (ASWPD), Delayed Sleep-Wake Phase Disorder (DSWPD),
Non-24-Hour Sleep-Wake Rhythm Disorder (N24SWD), and Irregular Sleep-Wake Rhythm
Disorder (ISWRD). An update for 2015: an American Academy of Sleep Medicine Clinical
Practice Guideline. J Clin Sleep Med. 2015;11(10):1199–236.
Ayalon L, Borodkin K, Dishon L, Kanety H, Dagan Y. Circadian rhythm sleep disorders following
mild traumatic brain injury. Neurology. 2007;68(14):1136–40.
Boivin DB, James FO. Circadian adaptation to night-shift work by judicious light and darkness
exposure. J Biol Rhythm. 2002;17(6):556–67.
Borodkin K, Ayalon L, Kanety H, Dagan Y. Dysregulation of circadian rhythms following
prolactin-secreting pituitary microadenoma. Chronobiol Int. 2005;22(1):145–56.
13 Circadian Rhythm Sleep–Wake Disorders: Mechanisms and Treatment 279
Boulos Z, Campbell SS, Lewy AJ, Terman M, Dijk DJ, Eastman CI. Light treatment for sleep
disorders: consensus report. VII. Jet lag. J Biol Rhythm. 1995;10(2):167–76.
Bromundt V, Koster M, Georgiev-Kill A, Opwis K, Wirz-Justice A, Stoppe G, et al. Sleep-wake
cycles and cognitive functioning in schizophrenia. Br J Psychiatry. 2011;198(4):269–76.
Brown DL, Feskanich D, Sanchez BN, Rexrode KM, Schernhammer ES, Lisabeth LD. Rotating
night shift work and the risk of ischemic stroke. Am J Epidemiol. 2009;169(11):1370–7.
Burgess HJ, Crowley SJ, Gazda CJ, Fogg LF, Eastman CI. Preflight adjustment to eastward travel:
3 days of advancing sleep with and without morning bright light. J Biol Rhythm. 2003;18(4):
318–28.
Burgess HJ, Revell VL, Molina TA, Eastman CI. Human phase response curves to three days of
daily melatonin: 0.5 mg versus 3.0 mg. J Clin Endocrinol Metab. 2010;95(7):3325–31.
Burkhart K, Phelps JR. Amber lenses to block blue light and improve sleep: a randomized trial.
Chronobiol Int. 2009;26(8):1602–12.
Campbell SS, Dawson D, Anderson MW. Alleviation of sleep maintenance insomnia with timed
exposure to bright light. J Am Geriatr Soc. 1993;41(8):829–36.
Canton JL, Smith MR, Choi HS, Eastman CI. Phase delaying the human circadian clock with a
single light pulse and moderate delay of the sleep/dark episode: no influence of iris color. J
Circadian Rhythms. 2009;7:8.
Cermakian N, Lamont EW, Boudreau P, Boivin DB. Circadian clock gene expression in brain
regions of Alzheimer’s disease patients and control subjects. J Biol Rhythm. 2011;26(2):
160–70.
Colligan MJ, Rosa RR. Shiftwork effects on social and family life. Occup Med. 1990;5(2):315–22.
Cortesi F, Giannotti F, Ivanenko A, Johnson K. Sleep in children with autistic spectrum disorder.
Sleep Med. 2010;11(7):659–64.
Crowley SJ, Eastman CI. Phase advancing human circadian rhythms with morning bright light,
afternoon melatonin, and gradually shifted sleep: can we reduce morning bright-light duration?
Sleep Med. 2015;16(2):288–97.
Czeisler CA, Walsh JK, Roth T, Hughes RJ, Wright KP, Kingsbury L, et al. Modafinil for excessive
sleepiness associated with shift-work sleep disorder. N Engl J Med. 2005;353(5):476–86.
Czeisler CA, Walsh JK, Wesnes KA, Arora S, Roth T. Armodafinil for treatment of excessive
sleepiness associated with shift work disorder: a randomized controlled study. Mayo Clin Proc.
2009;84(11):958–72.
Dai J, Swaab DF, Van der Vliet J, Buijs RM. Postmortem tracing reveals the organization of
hypothalamic projections of the suprachiasmatic nucleus in the human brain. J Comp Neurol.
1998;400(1):87–102.
DelRosso LM, Hoque R, James S, Gonzalez-Toledo E, Chesson AL Jr. Sleep-wake pattern
following gunshot suprachiasmatic damage. J Clin Sleep Med. 2014;10(4):443–5.
Drake CL, Roehrs T, Richardson G, Walsh JK, Roth T. Shift work sleep disorder: prevalence and
consequences beyond that of symptomatic day workers. Sleep. 2004;27(8):1453–62.
Drake C, Gumenyuk V, Roth T, Howard R. Effects of armodafinil on simulated driving and
alertness in shift work disorder. Sleep. 2014;37(12):1987–94.
Drake CL, Belcher R, Howard R, Roth T, Levin AM, Gumenyuk V. Length polymorphism in the
period 3 gene is associated with sleepiness and maladaptive circadian phase in night-shift
workers. J Sleep Res. 2015;24(3):254–61.
Ebisawa T, Uchiyama M, Kajimura N, Mishima K, Kamei Y, Katoh M, et al. Association of
structural polymorphisms in the human period3 gene with delayed sleep phase syndrome.
EMBO Rep. 2001;2(4):342–6.
Flynn-Evans EE, Tabandeh H, Skene DJ, Lockley SW. Circadian rhythm disorders and melatonin
production in 127 blind women with and without light perception. J Biol Rhythm. 2014;29(3):
215–24.
Flynn-Evans EE, Shekleton JA, Miller B, Epstein LJ, Kirsch D, Brogna LA, et al. Circadian phase
and phase angle disorders in primary insomnia. Sleep. 2017;40(12) https://doi.org/10.1093/
sleep/zsx163.
280 S. M. Abbott and P. C. Zee
Gooley JJ, Lu J, Chou TC, Scammell TE, Saper CB. Melanopsin in cells of origin of the
retinohypothalamic tract. Nat Neurosci. 2001;4(12):1165.
Gradisar M, Dohnt H, Gardner G, Paine S, Starkey K, Menne A, et al. A randomized controlled trial
of cognitive-behavior therapy plus bright light therapy for adolescent delayed sleep phase
disorder. Sleep. 2011;34(12):1671–80.
Guilleminault C, McCann CC, Quera-Salva M, Cetel M. Light therapy as treatment of dyschronosis
in brain impaired children. Eur J Pediatr. 1993;152(9):754–9.
Gumenyuk V, Belcher R, Drake CL, Roth T. Differential sleep, sleepiness, and neurophysiology in
the insomnia phenotypes of shift work disorder. Sleep. 2015;38(1):119–26.
Harma MI, Hakola T, Akerstedt T, Laitinen JT. Age and adjustment to night work. Occup Environ
Med. 1994;51(8):568–73.
Hart CL, Ward AS, Haney M, Foltin RW. Zolpidem-related effects on performance and mood
during simulated night-shift work. Exp Clin Psychopharmacol. 2003;11(4):259–68.
Hayakawa T, Uchiyama M, Kamei Y, Shibui K, Tagaya H, Asada T, et al. Clinical analyses of
sighted patients with non-24-hour sleep-wake syndrome: a study of 57 consecutively diagnosed
cases. Sleep. 2005;28(8):945–52.
Heikkila E, Hatonen TH, Telakivi T, Laakso ML, Heiskala H, Salmi T, et al. Circadian rhythm
studies in neuronal ceroid-lipofuscinosis (NCL). Am J Med Genet. 1995;57(2):229–34.
ICSD-3. The international classification of sleep disorders: diagnostic and coding manual. 2nd
ed. Darien, IL: American Academy of Sleep Medicine; 2014.
Jan JE, Espezel H, Appleton RE. The treatment of sleep disorders with melatonin. Dev Med Child
Neurol. 1994;36(2):97–107.
Jones CR, Campbell SS, Zone SE, Cooper F, DeSano A, Murphy PJ, et al. Familial advanced sleep-
phase syndrome: a short-period circadian rhythm variant in humans. Nat Med. 1999;5(9):
1062–5.
Kitamura S, Hida A, Enomoto M, Watanabe M, Katayose Y, Nozaki K, et al. Intrinsic circadian
period of sighted patients with circadian rhythm sleep disorder, free-running type. Biol Psychi-
atry. 2013;73(1):63–9.
Lack L, Wright H. The effect of evening bright light in delaying the circadian rhythms and
lengthening the sleep of early morning awakening insomniacs. Sleep. 1993;16(5):436–43.
Lack L, Wright H, Kemp K, Gibbon S. The treatment of early-morning awakening insomnia with
2 evenings of bright light. Sleep. 2005;28(5):616–23.
Lamont EW, James FO, Boivin DB, Cermakian N. From circadian clock gene expression to
pathologies. Sleep Med. 2007;8(6):547–56.
Lewy AJ, Sack RL. The dim light melatonin onset as a marker for circadian phase position.
Chronobiol Int. 1989;6(1):93–102.
Lewy AJ, Bauer VK, Hasler BP, Kendall AR, Pires ML, Sack RL. Capturing the circadian rhythms
of free-running blind people with 0.5 mg melatonin. Brain Res. 2001;918(1–2):96–100.
McCurry SM, Gibbons LE, Logsdon RG, Vitiello MV, Teri L. Nighttime insomnia treatment and
education for Alzheimer’s disease: a randomized, controlled trial. J Am Geriatr Soc. 2005;53(5):
793–802.
Mundey K, Benloucif S, Harsanyi K, Dubocovich ML, Zee PC. Phase-dependent treatment of
delayed sleep phase syndrome with melatonin. Sleep. 2005;28(10):1271–8.
Nagoshi E, Saini C, Bauer C, Laroche T, Naef F, Schibler U. Circadian gene expression in
individual fibroblasts: cell-autonomous and self-sustained oscillators pass time to daughter
cells. Cell. 2004;119(5):693–705.
Nagtegaal JE, Kerkhof GA, Smits MG, Swart AC, Van Der Meer YG. Delayed sleep phase
syndrome: a placebo-controlled cross-over study on the effects of melatonin administered five
hours before the individual dim light melatonin onset. J Sleep Res. 1998;7(2):135–43.
Okawa M, Nanami T, Wada S, Shimizu T, Hishikawa Y, Sasaki H, et al. Four congenitally blind
children with circadian sleep-wake rhythm disorder. Sleep. 1987;10(2):101–10.
13 Circadian Rhythm Sleep–Wake Disorders: Mechanisms and Treatment 281
Takaesu Y, Komada Y, Inoue Y. Melatonin profile and its relation to circadian rhythm sleep
disorders in Angelman syndrome patients. Sleep Med. 2012;13(9):1164–70.
The Medical Letter. Tasimelteon (Hetlioz) for non-24-hour sleep-wake disorder. Med Lett Drugs
Ther. 2014;56(1441):34–5.
Toh KL, Jones CR, He Y, Eide EJ, Hinz WA, Virshup DM, et al. An hPer2 phosphorylation site
mutation in familial advanced sleep phase syndrome. Science. 2001;291(5506):1040–3.
Tranah GJ, Blackwell T, Stone KL, Ancoli-Israel S, Paudel ML, Ensrud KE, et al. Circadian activity
rhythms and risk of incident dementia and mild cognitive impairment in older women. Ann
Neurol. 2011;70(5):722–32.
van Diepen HC, Ramkisoensing A, Peirson SN, Foster RG, Meijer JH. Irradiance encoding in the
suprachiasmatic nuclei by rod and cone photoreceptors. FASEB J. 2013;27(10):4204–12.
Wang JL, Lim AS, Chiang WY, Hsieh WH, Lo MT, Schneider JA, et al. Suprachiasmatic neuron
numbers and rest-activity circadian rhythms in older humans. Ann Neurol. 2015;78(2):317–22.
Wilhelmsen-Langeland A, Saxvig IW, Pallesen S, Nordhus IH, Vedaa O, Lundervold AJ, et al. A
randomized controlled trial with bright light and melatonin for the treatment of delayed sleep
phase disorder: effects on subjective and objective sleepiness and cognitive function. J Biol
Rhythm. 2013;28(5):306–21.
Witting W, Kwa IH, Eikelenboom P, Mirmiran M, Swaab DF. Alterations in the circadian rest-
activity rhythm in aging and Alzheimer’s disease. Biol Psychiatry. 1990;27(6):563–72.
Xu Y, Padiath QS, Shapiro RE, Jones CR, Wu SC, Saigoh N, et al. Functional consequences of a
CKIdelta mutation causing familial advanced sleep phase syndrome. Nature. 2005;434(7033):
640–4.
Zavada A, Gordijn MC, Beersma DG, Daan S, Roenneberg T. Comparison of the Munich
chronotype questionnaire with the Horne-Ostberg’s morningness-eveningness score.
Chronobiol Int. 2005;22(2):267–78.
Zee PC. Melantonin for the treatment of advanced sleep phase disorder. Sleep. 2008;31(7):923.
author reply 5
Zee PC, Vitiello MV. Circadian rhythm sleep disorder: irregular sleep wake rhythm type. Sleep
Med Clin. 2009;4(2):213–8.