Elsevier

Progress in Neurobiology

Volume 197, February 2021, 101902
Progress in Neurobiology

Sleep and its regulation: An emerging pathogenic and treatment frontier in Alzheimer’s disease

https://doi.org/10.1016/j.pneurobio.2020.101902Get rights and content

Highlights

  • Sleep disturbance is increasingly identified as contributing to Alzheimer's disease pathogenesis with potential treatment implications.

  • Patients with AD dementia exhibit lower sleep efficiency and lower amounts of SWS, compared to healthy elderly controls.

  • There is strong evidence in mouse models of AD that sleep directly affects the accumulation of Aβ and tau.

  • There is no consensus for how acute sleep loss affects AD biomarkers in cognitively healthy adults.

  • There are a number of potential sleep-associated therapeutic targets currently being explored in clinical trials for AD.

Abstract

A majority of patients with Alzheimer’s disease (AD) experience some form of sleep disruption, including nocturnal sleep fragmentation, increased daytime napping, decreased slow-wave sleep (SWS, stage N3), and decreased rapid-eye-movement sleep (REM). Clinical studies are investigating whether such sleep disturbances are a consequence of the underlying disease, and whether they also contribute to the clinical and pathological manifestations of AD. Emerging research has provided a direct link between several of these sleep disruptions and AD pathophysiology, suggesting that treating sleep disorders in this population may target basic mechanisms of the disease. Here, we provide a comprehensive review of sleep disturbances associated with the spectrum of AD, ranging from the preclinical stages through dementia. We discuss how sleep interacts with AD pathophysiology and, critically, whether sleep impairments can be targeted to modify the disease course in a subgroup of affected AD patients. Ultimately, larger studies that fully utilize new diagnostic and experimental tools will be required to better define the most relevant sleep disturbance to target in AD, the interventions that best modulate this target symptom, and whether successful early intervention can modify AD risk and prevent dementia.

Introduction

Alzheimer’s disease (AD) is clinically heterogeneous, with numerous risk factors that affect its clinical and neuropathologic course. Recent advances in neuroimaging and biomarkers, have led to AD being re-conceptualized with pathological processes that begin well before clinical symptoms and which evolve with progressive neuropathological neurodegenerative changes. The clinical symptoms manifest through a spectrum that can include subjective cognitive impairment (SCI), mild cognitive impairment (MCI), and mild-severe AD dementia (Bateman et al., 2012; Jack et al., 2018). Table 1 provides the nomenclature associated with the AD continuum as outlined by Jack and colleagues (2018) (Jack et al., 2018). The long asymptomatic period of AD pathophysiology opens up opportunities for early intervention to prevent clinical symptom evolution and dementia.

Recently, there has been growing interest in the sleep disruption associated with AD across this continuum, and as an important component of AD pathophysiology (Mander et al., 2016). It is hypothesized that sleep disturbances might present prior to other clinical symptoms, contribute to the preclinical cascade of neuropathology, and emerge when individuals have objective measurable impairment in cognition (Fig. 1).

Sleep disturbances are common in AD, highly disruptive to quality of life (Petrovsky et al., 2018), increase caregiver burden (Okuda et al., 2019), and are reported to be a leading cause of patients requiring institutional care (Bianchetti et al., 1995). A recent meta-analysis of 27 observational studies concluded that ∼15 % of AD cases could be delayed in onset or potentially prevented through effective approaches to address sleep disorders (Bubu et al., 2017). While broad disturbances in sleep have been recognized in AD for decades, more recent efforts have elucidated overlapping mechanisms underlying sleep disturbances and AD pathophysiology, highlighting the potential of treating specific aspects of sleep disruption as a disease modifying therapy (reviewed by Cedernaes et al., 2017; Mander et al., 2016).

In this review, we critically evaluate the evidence for the emerging link between sleep and AD pathophysiology (Box 1). We review subjective and objective methods for sleep assessment in patients along the spectrum of AD, the evidence for sleep disorders as a risk factor for AD, as well as the relationship between sleep and disease pathogenesis. We conclude with a review of potential treatment targets.

The historical and current evaluation of the literature is complicated by the different reference standards for AD diagnosis, from clinically-based diagnosis with lower rates of accuracy, to the new evolving biomarker-based diagnosis and the longstanding historical gold standard of neuropathologically verified disease. Only recently have well-validated and accurate CSF assays and PET biomarkers become available for diagnostic purposes (Jack et al., 2018). Within the biomarker-based criteria there are still challenges to address including studies that have relied exclusively on CSF Aβ for diagnostic reporting. While low levels of CSF Aβ42 are commonly reported for diagnostic purposes, assays and studies vary in the cut-off points used to indicate plaque accumulation (Ju et al., 2013; Molano et al., 2017; Varga et al., 2016). Furthermore, an increase of CSF Aβ42 prior to plaque formation has been proposed (Varga et al., 2016), but requires further validation.

Section snippets

Sleep across the Alzheimer’s disease spectrum

Estimates of the prevalence of sleep disorders in AD vary between studies, with one study reporting that 65% of patients with AD or MCI met the diagnostic criteria for a major sleep disorder, including insomnia, sleep disordered breathing, REM sleep behaviour disorder, restless legs syndrome, and excessive daytime sleepiness (Guarnieri et al., 2012). With the increasing availability and utilization of a wide variety of tools including wearables to evaluate sleep, the prevalence of specific

Sleep disturbance as a risk factor for cognitive decline or dementia

In addition to the evidence that sleep disturbances are associated with AD biomarkers in cognitively normal individuals (e.g., Branger et al., 2016; Carvalho et al., 2018; Spira et al., 2013), which suggests that sleep disturbances are an early symptom in the preclinical stage of the AD continuum, longitudinal studies also identify sleep as a risk factor for dementia. Most of these longitudinal studies do not measure AD biomarkers, so it is unknown whether the sleep disturbances occur prior to

Beyond a symptom: Sleep disruption contributes to Alzheimer’s disease pathogenesis

Although the precise mechanisms leading to AD are still a major focus of investigation and debate, the accumulation of misfolded Aβ and tau proteins remain the key pathological hallmarks of the disease (Ittner and Götz, 2011), with evidence that sleep disturbances directly contribute to Aβ metabolism, clearance, and deposition (Ju et al., 2014).

Mechanisms underlying the relationship between sleep disturbances and Alzheimer’s disease

There is substantial evidence that sleep disturbances occur throughout the AD spectrum and that sleep may directly affect the accumulation of Aβ and tau. Table 5 summarizes some of the proposed mechanisms driving the relationship between AD and sleep.

Treating sleep disorders

Treating sleep disorders in patients with AD is becoming a topic of greater importance given its potential role in the pathogenesis and course of the disease. There are several potential mechanisms underlying the relationship between sleep and AD from neurodegeneration of sleep promoting brain regions to changes in neurotransmitters and circadian rhythms, while there is also growing evidence that sleep is not only a symptom of the underlying pathology but that sleep disturbances contribute to

Conclusion

There is growing evidence suggesting that sleep is a potential therapeutic target and novel outcome measure for AD. Sleep will be an important component of a prevention platform for dementia: personalizing treatment and stratifying risk. Given AD is a heterogenous multifactorial disease, we predict only a subset of AD cases have sleep as a critical risk factor. The goal will be to identify those individuals who would benefit most from interventions targeting sleep as a way to change the

Acknowledgments

The authors gratefully acknowledge funding from Michael Smith Foundation for Health Research, Canada (BAK, HBN), CIHR Banting, Canada (#378933) (BAK), Pacific Alzheimer Research Foundation (HBN), The Association for Frontotemporal Dementia (HBN), and NIH-NINDS, United States (#K99NS109909) (BAK). The authors would also like to thank Meghan Chen for her input and editing support as well as Alexandre Shadyab for help with Fig. 1.

References (209)

  • P. Dykierek et al.

    The value of REM sleep parameters in differentiating Alzheimer’s disease from old-age depression and normal aging

    J. Psychiatr. Res.

    (1998)
  • R. Fronczek et al.

    Hypocretin (orexin) loss in Alzheimer’s disease

    Neurobiol. Aging

    (2012)
  • A. Gabelle et al.

    Cerebrospinal fluid levels of orexin-A and histamine, and sleep profile within the Alzheimer process

    Neurobiol. Aging

    (2017)
  • E.A. Hahn et al.

    A change in sleep pattern may predict Alzheimer disease

    Am. J. Geriatr. Psychiatry

    (2014)
  • D.G. Harper et al.

    Disturbance of endogenous circadian rhythm in aging and alzheimer disease

    Am. J. Geriatr. Psychiatry

    (2005)
  • E. Hennevin et al.

    Neural representations during sleep: from sensory processing to memory traces

    Neurobiol. Learn. Mem.

    (2007)
  • P. Hot et al.

    Changes in sleep theta rhythm are related to episodic memory impairment in early Alzheimer’s disease

    Biol. Psychol.

    (2011)
  • V. Ibanez et al.

    A survey on sleep questionnaires and diaries

    Sleep Med.

    (2018)
  • C.R. Jack et al.

    NIA-AA Research Framework: toward a biological definition of Alzheimer’s disease

    Alzheimers Dement.

    (2018)
  • C. Jorge et al.

    The STOP-Bang and Berlin questionnaires to identify obstructive sleep apnoea in Alzheimer’s disease patients

    Sleep Med.

    (2019)
  • F. Kamenetz et al.

    APP processing and synaptic function

    Neuron

    (2003)
  • H.A. Keage et al.

    What sleep characteristics predict cognitive decline in the elderly?

    Sleep Med.

    (2012)
  • B.A. Kent et al.

    Sleep and hippocampal neurogenesis: implications for Alzheimer’s disease

    Front. Neuroendocrinol.

    (2017)
  • B.A. Kent et al.

    Delayed daily activity and reduced NREM slow wave power in the APPswe/PS1dE9 mouse model of Alzheimer’s disease

    Neurobiol. Aging

    (2019)
  • J.C. Kreutzmann et al.

    Sleep deprivation and hippocampal vulnerability: changes in neuronal plasticity, neurogenesis and cognitive function

    Neuroscience

    (2015)
  • J.E. Lee et al.

    Sleep-disordered breathing and Alzheimer’s disease: a nationwide cohort study

    Psychiatry Res.

    (2019)
  • G. Adler et al.

    Short-term rivastigmine treatment reduces EEG slow-wave power in Alzheimer patients

    Neuropsychobiology

    (2001)
  • S. Ancoli-Israel et al.

    Sleep in non-institutionalized Alzheimer’s disease patients

    Aging Clin. Exp. Res.

    (1994)
  • S. Ancoli-Israel et al.

    Use of wrist activity for monitoring sleep/wake in demented nursing-home patients

    Sleep

    (1997)
  • S. Ancoli-Israel et al.

    Cognitive effects of treating obstructive sleep apnea in Alzheimer’s disease: a randomized controlled study

    J. Am. Geriatr. Soc.

    (2008)
  • J. Arendt et al.

    Efficacy of melatonin treatment in jet lag, shift work, and blindness

    J. Biol. Rhythms

    (1997)
  • S. Balkan et al.

    Effect of donepezil on EEG spectral analysis in Alzheimer’s disease

    Acta Neurol. Belg.

    (2003)
  • N.R. Barthélemy et al.

    Sleep deprivation affects tau phosphorylation in human cerebrospinal fluid

    Ann. Neurol.

    (2020)
  • M. Basta et al.

    Associations between sleep duration and cognitive impairment in mild cognitive impairment

    J. Sleep Res.

    (2019)
  • R.J. Bateman et al.

    Clinical and biomarker changes in dominantly inherited Alzheimer’s disease

    N. Engl. J. Med.

    (2012)
  • C. Benedict et al.

    Effects of acute sleep loss on diurnal plasma dynamics of CNS health biomarkers in young men

    Neurology

    (2020)
  • A. Bianchetti et al.

    Predictors of mortality and institutionalization in Alzheimer disease patients 1 year after discharge from an Alzheimer dementia unit

    Dement. Geriatr. Cogn. Disord.

    (1995)
  • S. Billioti de Gage et al.

    Benzodiazepine use and risk of Alzheimer’s disease: case-control study

    BMJ

    (2014)
  • F. Boddy et al.

    Subjectively reported sleep quality and excessive daytime somnolence in Parkinson’s disease with and without dementia, dementia with Lewy bodies and Alzheimer’s disease

    Int. J. Geriatr. Psychiatry

    (2007)
  • E. Bonanni et al.

    Daytime sleepiness in mild and moderate Alzheimer’s disease and its relationship with cognitive impairment

    J. Sleep Res.

    (2005)
  • P. Bourgin et al.

    Hypocretin-1 modulates rapid eye movement sleep through activation of locus coeruleus neurons

    J. Neurosci.

    (2000)
  • H. Braak et al.

    Stages of the pathologic process in Alzheimer disease: age categories from 1 to 100 years

    J. Neuropathol. Exp. Neurol.

    (2011)
  • S. Brassen et al.

    Short-term effects of acetylcholinesterase inhibitor treatment on EEG and memory performance in Alzheimer patients: an open, controlled trial

    Pharmacopsychiatry

    (2003)
  • O.M. Bubu et al.

    Sleep, cognitive impairment, and Alzheimer’s disease: a systematic review and meta-analysis

    Sleep

    (2017)
  • O.M. Bubu et al.

    Obstructive sleep apnea and longitudinal Alzheimer’s disease biomarker changes

    Sleep

    (2019)
  • S.L. Burke et al.

    Treatment of sleep disturbance may reduce the risk of future probable Alzheimer’s disease

    J. Aging Health

    (2019)
  • D.J. Buysse et al.

    The Pittsburgh Sleep Quality Index: a new instrument for psychiatric practice and research

    Psychiatry Res.

    (1988)
  • L. Carnicelli et al.

    A longitudinal study of polysomnographic variables in patients with mild cognitive impairment converting to Alzheimer’s disease

    J. Sleep Res.

    (2019)
  • J. Carrier et al.

    The effects of age and gender on sleep EEG power spectral density in the middle years of life (ages 20–60 years old)

    Psychophysiology

    (2001)
  • J. Carrier et al.

    Sleep slow wave changes during the middle years of life

    Eur. J. Neurosci.

    (2011)
  • Cited by (35)

    • Adjunctive and alternative treatments of circadian rhythm sleep disorders

      2023, Encyclopedia of Sleep and Circadian Rhythms: Volume 1-6, Second Edition
    • Comparison of polysomnography in people with Alzheimer's disease and insomnia versus non-demented elderly people with insomnia

      2023, Sleep Medicine
      Citation Excerpt :

      Age matching is particularly important as many of the reported alterations in PSG for AD individuals appear to be exaggerations of those found in non-demented elderly rather than qualitative differences. Confirmatory evidence regarding differences in PSG profiles for those with AD versus non-demented controls is important given that such differences have been proposed as potential biomarkers for the disease (e.g., the putative reduction in REM) or suggested therapeutic approaches (e.g., promotion of slow wave sleep) [10,11,16]. In the present analysis we attempted to address some of these limitations by using baseline PSG data obtained during the clinical development for suvorexant, an orexin receptor antagonist for the treatment of insomnia [17].

    • Alterations of sleep oscillations in Alzheimer's disease: A potential role for GABAergic neurons in the cortex, hippocampus, and thalamus

      2022, Brain Research Bulletin
      Citation Excerpt :

      In the final section, we consider evidence that alterations in GABAergic neurons in the cortex, hippocampus and thalamic reticular nucleus could account for sleep oscillatory changes in AD. For more general reviews on mechanisms of sleep, and sleep and circadian changes in AD, readers may refer to other comprehensive reviews (Brown et al., 2012; Byron et al., 2021; Hanke et al., 2022; Kent et al., 2021; Lee et al., 2020; Mander, 2020; Ning and Jorfi, 2019; Wang and Holtzman, 2020; Weng et al., 2020). Another article in this special issue focuses on pharmacological and brain stimulation approaches to influence sleep and arousal in brain disorders including dementia (Brown et al., 2022).

    • The role of orexin in Alzheimer disease: From sleep-wake disturbance to therapeutic target

      2021, Neuroscience Letters
      Citation Excerpt :

      It is known that REM sleep behavior disorder is commonly associated with neurodegenerative disorders. In AD patients, REM sleep has been documented to be severely impaired, and the significant change is already evident in the MCI stage of AD [40,82,83]. Prospective study reveals that both shorter REM sleep percentage and longer latency to REM sleep are independently associated with a higher risk of dementia [4,84].

    View all citing articles on Scopus
    View full text